Functions of 55 Newfound Proteins and Their Medicinal Application in the Treatment and Prevention of Disease

Abstract
The invention relates to 55 newly discovered proteins, which are present in isolated purified protein complexes, derived medicinal products, recombinant DNA, engineered DNA, cDNA, monoclonal and natural products or synthesized products as part of nutrition, food, and/or supplemental products and their applications.
Description
FIELD OF THE INVENTION

55 novel proteins have been identified in various biological constructs and have been sequenced, and tested for their medicinal application in the treatment and prevention of disease.


BACKGROUND OF THE INVENTION

Cells are the smallest component in an organism and are responsible for the production of proteins that can either cause harm to the organism or prevent and treat disease and infection in the organism. Since the discovery of cells as a basic building block of human life, scientists have been studying them in live (in vivo) and cultured mediums (in vitro) to try and discover the key to disease prevention.


As determined by the World Health Organization in 2014, cancer accounts for 8.2 million deaths per year and 14.6% of all deaths worldwide. There are various methods and drugs used in the treatment of cancer, including surgery excision, chemotherapy, radiotherapy, hormonal medications, and for sexual tract cancers the removal of sex organs that produce tumor cell-stimulating hormones, e.g. ER/PR positive cancers. While effective, many of these therapies contribute to side effects that can be almost as debilitating as the original disease. For example, it is extremely common to treat breast or ovarian cancers with surgical removal of the tumor(s) in addition to chemotherapy, which may produce early onset menopause and require long-term drug therapy to offset the premature aging of the body. This premature aging process can lead to osteopenia, osteoporosis, hot flashes, and vaginal dryness—and subsequent long-term treatment with expensive bisphosphonates and hormonal drugs to offset its effects. It is therefore desirable to develop cancer treatments that can be used alone or in combination with other cancer therapies, which have little or no side effects, yet are effective in treating or preventing the progression of the disease.


While cancer is perhaps the most nefarious affliction affecting human health, other diseases and infections that have been identified and studied in detail by scientists remain prevalent and without cure in society. The methods and drugs that have been developed to treat those diseases and infections often come with their own repercussions and adverse side effects. Examples of some of the most widely studied diseases and viruses that require better treatment options include, HIV, hepatitis, diabetes, atherosclerosis and related cardiovascular diseases, influenza, and Parkinson's disease.


Therefore, KH cells and proteins have been discovered, isolated, and purified in combination for treating a wide variety of diseases, infections, and other physical conditions and disorders, without many of the repercussions and adverse side effects of previously discovered drugs and methods of treatment.


SUMMARY OF THE INVENTION

55 proteins have been discovered, isolated, and purified for use in various constructs helpful in treating and preventing a wide variety of diseases, infections, disorders, and afflictions which adversely affect health.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows graphical representation of prism files containing dose-dependent curves.



FIG. 2 is a graph depicting the inhibition rate of HIV-1 treated with various plasma products.



FIG. 3 is a graph depicting the inhibition rate of HIV-1 compared with the dilution of various plasma products.



FIG. 4 is a graph of OD 450 and dilution of various plasma products used in treating HIV-1.



FIGS. 5 and 6 are graphs showing the dose dependent curves of various plasma products used in the treatment of HVC.



FIGS. 7-15 are graphs comparing FSC/SSC on FACS.



FIGS. 16-22 are graphs comparing human T/B cells on FACS.



FIGS. 23-30A are graphs comparing human granulocytes on FACS.



FIG. 30B is a graph comparing human NK cells on FACS.



FIG. 31A is a graph depicting AFOD 1 vs. human insulin results.



FIG. 31B is a graph depicting AFOD RAAS 101 vs. human insulin results.



FIG. 31C is a graph depicting AFOD RAAS 103 vs. human insulin results.



FIG. 31D is a graph depicting AFOD RAAS 107 vs. human insulin results.



FIG. 31E is a graph depicting AFOD RAAS 108 vs. human insulin results.



FIG. 31F is a graph depicting AFOD RAAS 109 vs. human insulin results.



FIG. 31G is a graph depicting AFOD RAAS 110 vs. human insulin results.



FIG. 31H is a graph depicting AFOD RAAS 120 vs. human insulin results.



FIG. 31I is a graph depicting AFOD RAAS 121 vs. human insulin results.



FIG. 31J is a graph depicting AFOD RAAS KH vs. human insulin results.



FIG. 31K is a graph depicting AFCC KH1 vs. human insulin results.



FIG. 31L is a graph depicting KH105(1) vs. human insulin results.



FIG. 31M is a graph depicting KH105(2) vs. human insulin results.



FIG. 31N shows dose response graphs of glucose uptake for AFOD RAAS 107 and AFOD KH_N1.



FIG. 31O shows dose response graphs of glucose uptake for AFOD RAAS 107 and AFOD KH_N2.



FIG. 31P shows dose response graphs of glucose uptake for AFOD RAAS 107 and AFOD KH_N3.



FIG. 31Q shows dose response graphs of glucose uptake for AFOD 1 and AFOD RAAS 109_N1.



FIG. 31R shows dose response graphs of glucose uptake for AFOD 1 and AFOD RAAS 109_N2.



FIGS. 31S-31Z show additional response graphs of glucose uptake.



FIGS. 32-36 are pictures of the aorta of mice in the study of APOAI protein in preventing atherosclerosis and related cardiovascular diseases. FIG. 32 is the vehicle control group. FIG. 33 is the low dose group. FIG. 34 is the medium dose group. FIG. 35 is the high dose group. FIG. 36E is the positive control (Lipitor) group.



FIGS. 36A-36D are pictures of organs of mice in the study of APOAI protein in preventing atherosclerosis and related cardiovascular diseases. FIG. 36A shows liver fatty change of control and treated animals. FIG. 36B shows fat deposits on the heart of control and treated animals. FIG. 36C shows atherosclerosis on control and treated animals, first view. FIG. 36D shows atherosclerosis change on control and treated animals, second view.



FIGS. 37-38 are pictures of dissected mice in the study: pre-clinical animal test of Apo-AI—for the antiatherogenic and cholesterol-lowering properties. FIG. 37 shows the fatty streak lesions and liver change of a animals fed with a high fat diet and sacrificed on week 10. FIG. 38 shows the plaque area change with normal diet after 10 weeks with a high fat diet in the control group—animals which were given a normal diet after establishing fatty streak lesions in aortas.



FIG. 39 is a graph showing change of weight and plasma lipid concentrations compared between APOAI treated and control animals.



FIG. 40 is a picture of a normal rabbit aorta without fatty streak lesion.



FIG. 41 is a picture of the area of fatty streak lesion in the aorta from the APOAI group 1.



FIG. 42 is a picture of the area of fatty streak lesion in the aorta from the APOAI group 2.



FIG. 43 is a graph comparing number of days treated with AFCC vs. bodyweight.



FIG. 44 is a graph comparing days post infection vs. survival rate.



FIG. 45 is a graph comparing days post infection vs. bodyweight.



FIG. 46 is a graph comparing days post infection vs. bodyweight change.



FIG. 47 is a graph comparing days post infection vs. bodyweight change.



FIG. 48 is a graph comparing days post infection vs. survival distribution function expressed as a percentage.



FIG. 49 is a graph comparing days before and after infection vs. bodyweight change.



FIG. 50 is a graph comparing HBV copies/ul plasma vs. days after infection for various test groups.



FIG. 51 is a graph of the effect of prophylactic treatment or therapeutic treatment of RAAS 8 on ETV on the HBsAg in mouse blood.



FIG. 52 is a graph of the effect of prophylactic treatment or therapeutic treatment of RAAS 8 on ETV on the intermediate HBV replication in the mouse livers by qPCR.



FIG. 53 is a southern blot determination of intermediate HBV DNA in mouse livers.



FIG. 54 is a graph depicting the bodyweight of mice treated with vehicle or indicated compounds during the course of experiment.



FIG. 55 is a graph depicting the efficacy of therapeutic treatment of RAAS 105 on in vivo HBV replication in plasma in mouse HDI model.



FIG. 56 is a graph depicting the efficacy of prophylactic treatment of RAAS 105 on in vivo HBV replication in plasma in mouse HDI model.



FIG. 57 is a graph depicting the effect of therapeutic treatment of RAAS 105 on the HBsAg in mouse plasma.



FIG. 58 is a graph depicting the effect of prophylactic treatment of RAAS 105 on the HBsAg in mouse plasma.



FIG. 59 is a graph depicting the effect of therapeutic treatment of RAAS 105 on the intermediate HBV replication in the mouse livers by qPCR.



FIG. 60 is a graph depicting the effect of prophylactic treatment of RAAS 105 on the intermediate HBV replication in the mouse livers by qPCR.



FIG. 61 is a graph depicting the body weights of mice in all groups treated with vehicle or indicated compounds during the course of the experiment “Efficacy of A Human Plasma Derived Protein AFOD RAAS 105 in Inhibition of the HBV Replication in the Mouse Hydrodynamic Injection Model” (n=10).



FIG. 62 is a graph depicting percentages of T and B lymphocytes in peripheral blood, with and without therapeutic RAAS 105 treatment.



FIG. 63 is a graph depicting percentages of T and B lymphocytes in peripheral blood, with further analysis done on CD4 and CD8 T cell lineages, with and without therapeutic RAAS 105 treatment.



FIG. 64 is a graph depicting percentages of CD4 and CD8 T cells in peripheral blood, with and without therapeutic RAAS 105 treatment.



FIG. 65 is a graph depicting percentages of CD4 and CD8 T cells in peripheral blood, with further analysis done on the percentages of CD11c+ dendritic cells (DC) and Gr-1+ granulocytes.



FIG. 66 is graphs depicting percentages of dendritic cells and granulocytes in peripheral blood, with and without therapeutic RAAS 105 treatment.



FIG. 67 is graph showing another representation of Gr-1 vs. CD 11c cells, with and without therapeutic RAAS 105 treatment.



FIG. 68 is a graph depicting the percentage of monocytes in peripheral blood, with and without therapeutic RAAS 105 treatment.



FIG. 69 is a graph showing another representation of monocytes in peripheral blood, with and without therapeutic RAAS 105 treatment.



FIG. 70 is graphs depicting percentages of T and B lymphocytes in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 71 is a graph showing another representation of T and B lymphocytes in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 72 is graphs depicting percentages of CD4 and CD8 T cells in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 73 is a graph showing another representation of CD4 and CD8 T cells in the spleen, with CD3 T cells being gated, with and without therapeutic RAAS 105 treatment.



FIG. 74 is graphs depicting T cell subset percentages in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 75 is a graph of CD4 T cell subset percentages in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 76 is graphs depicting T cell subset percentages in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 77 is a graph of CD8 T cell subset percentages in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 78 is a graph depicting percentages of regulatory T cells in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 79 is another graphical representation of percentages of regulatory T cells in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 80 is graphs depicting percentages of mDc and pDcs in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 81 is another graphical representation of mDC and pDcs in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 82 is graphs depicting percentages of macrophages and granulocytes in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 83 is another graphical representation of percentages of macrophages and granulocytes in the spleen, with and without therapeutic RAAS 105 treatment.



FIG. 84 is a graph depicting percentages of T cells in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 85 is graphs showing percentages of CD3 T cells in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 86 is graphs depicting percentages of CD4 and CD8 T cells in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 87 is another graphical representation of CD4 and CD8 T cells in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 88 is graphs depicting CD4 T cell subset percentages in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 89 is another graphical representation of CD4 T cell subset percentages in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 90 is graphs depicting CD8 T cell subset percentages in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 91 is another graphical representation of CD8 T cell subset percentages in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 92 is a graph depicting percentages of Foxp3 regulatory T cells in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 93 is another graphical representation of Foxp3 regulatory T cells in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 94 is a graph depicting percentages of DCs in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 95 is another graphical representation of percentages of DCs in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 96 is graphs depicting percentages of macrophages and granulocytes in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 97 is another graphical representation of percentages of macrophages and granulocytes in the lymph nodes, with and without therapeutic RAAS 105 treatment.



FIG. 98 is graphs depicting T and B lymphocytes in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 99 is another graphical representation of T and B cells in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 100 is graphs depicting percentages of CD4 and CD 8 T cells in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 101 is another graphical representation of CD4 and CD 8 T cells in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 102 is graphs depicting percentages of dendritic cells and granulocytes in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 103 is another graphical representation of dendritic cells and granulocytes in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 104 is a graph depicting percentages of monocytes in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 105 is another graphical representation of percentages of monocytes in peripheral blood, with and without prophylactic RAAS 105 treatment.



FIG. 106 is graphs depicting percentages of T and B lymphocytes in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 107 is another graphical representation of percentages of T and B lymphocytes in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 108 is graphs depicting percentages of CD4 and CD8 T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 109 is another graphical representation of percentages of CD4 and CD8 T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 110 is graphs depicting subset percentages of T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 111 is another graphical representation of subset percentages of T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 112 is graphs depicting subset percentages of T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 113 is another graphical representation of subset percentages of T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 114 is a graph depicting Foxp3 regulator T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 115 is another graphical representation of Foxp3 regulator T cells in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 116 is graphs depicting percentages of pDCs and mDCs in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 117 is another graphical representation of percentages of pDCs and mDCs in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 118 is graphs depicting percentages of macrophages and granulocytes in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 119 is another graphical representation of percentages of macrophages and granulocytes in the spleen, with and without prophylactic RAAS 105 treatment.



FIG. 120 is a graph depicting percentages of T cells in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 121 is another graphical representation of percentages of CD3 T cells in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 122 is graphs depicting percentages of CD4 and CD8 T cells in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 123 is another graphical representation of percentages of CD4 and CD8 T cells in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 124 is graphs depicting T cell subset percentages in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 125 is another graphical representation of T cell subset percentages in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 126 is graphs depicting T cell subset percentages in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 127 is another graphical representation of T cell subset percentages in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 128 is a graph depicting percentages of Foxp3 regulatory T cells in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 129 is another graphical representation of Foxp3 regulatory T cells in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 130 is a graph depicting percentages of DCs in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 131 is another graphical representation of percentages of DCs in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 132 is graphs depicting percentages of macrophages and granulocytes in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 133 is another graphical representation of percentages of macrophages and granulocytes in the lymph nodes, with and without prophylactic RAAS 105 treatment.



FIG. 134 is graphs depicting the effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108, and AFOD 1 on bodyweight (A) and bodyweight change (B).



FIG. 135 is graphs depicting the effects of AFCC KH, AFOD 101 and AFOD 102 on bodyweight (A) and bodyweight change (B).



FIG. 136 is graphs depicting the effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108, and AFOD 1 on delta paw volume (A) and AUC of paw swelling (B).



FIG. 137 is graphs depicting the effects of AFCC KH, AFOD 101, and AFOD 102 on delta paw volume (A) and AUC of paw swelling (B).



FIG. 138 is a graph depicting the effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108, and AFOD 1 on arthritic score.



FIG. 139 is a graph depicting the effects of AFCC KH, AFOD 101 and AFOD 102 on arthritic score.



FIG. 140 is a graph depicting the effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108, and AFOD 1 on the incidence rate of arthritis.



FIG. 141 is a graph depicting the effects of AFCC KH, AFOD 101, and AFOD 102 on the incidence rate of arthritis.



FIG. 142 is a graph depicting the effect of various doses of APOA 1 on bodyweight.



FIG. 143 is a graph depicting the effect of HFD on the lipid profile in ApoE mice.



FIG. 144 is a graph depicting the effect of various doses of APOA 1 on plasma TC.



FIG. 145 is a graph depicting the net change of plasma TC with various doses of APOA 1.



FIG. 146 is a graph depicting the effect of various doses of APOA 1 on plasma triglycerides.



FIG. 147 is a graph depicting the effect of various doses of APOA 1 on plasma HDL levels.



FIG. 148 is a graph depicting the effect of various doses of APOA 1 on the net change of plasma LDL levels.



FIG. 149 is a graph depicting the effect of various doses of APOA 1 on plasma LDL levels.



FIG. 150 is a graph depicting the effect of various doses of APOA 1 on the net change of plasma HDL levels.



FIG. 151 is pictures of the effect of APOA 1 on the atherosclerosis plaque lesion of a mouse.



FIG. 152 is pictures of the effect of APOA 1 on the atherosclerosis plaque lesion of a mouse.



FIG. 153 is a graph depicting the percentage of plaque area in the total inner vascular area of mice treated with various doses of APOA 1.



FIG. 154 is a picture of the arterial arch area of a mouse.



FIG. 155 is a graph depicting the percent of root plaque area in the arterial arch area of mice treated with various doses of APOA 1.



FIG. 156 is a picture of the area analyzed of the root to the right renal artery.



FIG. 157 is a graph depicting the percentage of plaque area from the root to the right renal artery of mice treated with various doses of APOA 1.



FIG. 158 is a graph of the effect of various doses of APOA 1 on liver weight.



FIG. 159 is a graph of the effect of various doses of APOA 1 on liver/body weight.



FIG. 160 is a graph comparing percentages of plaque area of mice fed high fat diets for 4, 19, and 27 weeks, treated with various doses of APOA 1.



FIG. 161 is a graph comparing TC levels of mice fed high fat dies for 4, 19, and 27 weeks, treated with various vehicles.



FIG. 162 is a graph comparing LDL levels of mice fed high fat diets for 4, 19, and 27 weeks, treated with various vehicles.



FIG. 163 is another graphical representation comparing LDL levels of mice fed high fat dies for 4, 19, and 27 weeks, treated with various vehicles.



FIG. 164 is pictures of aorta plaque lesions after 16 weeks of treatment with various doses of APOA 1.



FIG. 165 is a graph depicting the anti-colo-rectal tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and AFOD at various doses in PDX model CO-04-0002.



FIG. 166 is a graph depicting the anti-colo-rectal tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and AFOD at various doses in PDX model CO-04-0002 and CO-04-0001.



FIG. 167 is a picture of colo-rectal derived tumors dissected from the abdominal cavity of mice.



FIG. 168 is a graph depicting the relative change of bodyweight of mice treated with high concentrated fibrinogen enriched a1at thrombin and AFOD at various doses.



FIG. 169 is a graph depicting the anti-lung-derived tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and AFOD at various doses in PDX model LU-01-0032.



FIG. 170 is a picture of lung-derived tumors dissected from the abdominal cavity of mice.



FIG. 171 is a graph depicting ratios of mice with palpable lung cancer derived tumors observed in mice treated with high concentrated fibrinogen enriched a1at thrombin and AFOD at various doses.



FIG. 172 is a graph depicting the relative change of bodyweight of mice treated with high concentrated fibrinogen enriched a1at thrombin and AFOD at various doses.



FIG. 173-182 are ovarian cancer survival curve graphs of mice treated with AFOD RAAS 1, AFOD RAAS 104, AFOD RAAS 108, AFOD RAAS 109/121, AFOD RAAS 110, AFOD 113, AFOD RAAS 114, AFOD RAAS 120 and AFFC RAAS 1, AFCC RAAS 2, and positive and vehicle, respectively.



FIG. 183 is pictures of dissected mice in the ovarian cancer vehicle group vs. AFOD KH1 treatment group.



FIG. 184 is pictures of dissected mice in the ovarian cancer AFOD KH1 group.



FIG. 185 is pictures of dissected mice in the ovarian cancer positive control group.



FIG. 186 is pictures of dissected mice in the ovarian cancer AFOD RAAS 1 group.



FIG. 187 is pictures of dissected mice in the ovarian cancer AFOD RAAS 104 group.



FIG. 188 is pictures of dissected mice in the ovarian cancer AFOD RAAS 108 group.



FIG. 189 is pictures of dissected mice in the ovarian cancer AFOD RAAS 109/121 group.



FIG. 190 is pictures of dissected mice in the ovarian cancer AFOD RAAS 110 group.



FIG. 191 is pictures of dissected mice in the ovarian cancer AFOD RAAS 113 group.



FIG. 192 is pictures of dissected mice in the ovarian cancer AFOD RAAS 114 group.



FIG. 193 is pictures of dissected mice in the ovarian cancer AFCC RAAS 1 group.



FIG. 194 is pictures of dissected mice in the ovarian cancer AFCC RAAS 2 group.



FIG. 195 is pictures of dissected mice in the ovarian cancer AFOD RAAS 120 group.



FIG. 196 is a picture of a mouse implanted with MDA-MB-231-Luc tumor cells.



FIG. 197 is a picture of one of the 10 nude mice with MDA-MB-231-Luc tumor cells used in the re-implantation study, with dissection over tumor area revealed.



FIG. 198 is a picture of a mouse implanted with MDA-MB-231-Luc tumor cells.



FIG. 199 is a graph depicting tumor volume vs. days post implantation for mice implanted with MDA-MB-231-Luc tumor cells.



FIG. 200 is pictures of a mouse implanted with MDA-MB-231-Luc tumor cells with tumor site dissected.



FIG. 201 is pictures of the re-implantation of tumor tissues in nude mice.



FIG. 202 is a raw data of tumor growth in mice treated with AFCC prior to re-implantation



FIG. 203 is a graph showing 5 groups of nude mice after tumor volume change after the second re-implantation with breast tumor cancer.



FIG. 204 is pictures of re-implantation of tumor-tissues in nude mice.



FIG. 205 is a picture of a mouse implanted with a re-implanted tumor.



FIG. 206 is a graph of tumor weight for a mice re-implanted with tumors.



FIG. 207 is a picture of a mouse showing no tumor at the dissected at the implant site.



FIG. 208 is a picture of a mouse with no tumor after implantation.



FIG. 209 is pictures of nude mice re-implanted with tumor tissues.



FIG. 210 is a picture of a nude naïve mouse at 8 weeks old used as a negative normal control.



FIG. 211 is a picture of a nude naïve mouse at 8 weeks old used as a negative normal control.



FIG. 212 is a picture of a mouse used in tumor studies.



FIG. 213 is a graph depicting the percentages of B cells in peripheral blood.



FIG. 214 is a graph depicting the percentages of activated B lymphocytes in peripheral blood.



FIG. 215 is a graph depicting the percentages of monocytes and macrophages in peripheral blood.



FIG. 216 is a graph depicting the percentages of mDC and pDC in peripheral blood.



FIG. 217 is a graph depicting the percentages of CD3+ T cells in the spleen.



FIG. 218 is a graph depicting the percentages of B cells in the spleen.



FIG. 219 is a graph depicting the percentages of mDc and pDc in the spleen.



FIG. 220 is a graph of the percentages of activated B lymphocytes in the spleen.



FIG. 221 is a graph of the percentages of monocytes and macrophages in the spleen.



FIG. 222 is a graph of the percentages of granulocytes in the spleen.



FIG. 223 is a graph of percentages of CD3+ T cells in the draining lymph nodes.



FIG. 224 is a graph of the percentages of B cells in the draining lymph nodes.



FIG. 225 is a graph of the percentages of mDC and pDC in the draining lymph nodes.



FIG. 226 is a graph of the percentages of granulocytes in the draining lymph nodes.



FIG. 227 is a graph of the percentages of monocytes and macrophages in the draining lymph nodes.



FIG. 228 is a graph of the percentages of activated B lymphocytes in the draining lymph nodes.



FIG. 229 is a graph of APOE KO mice by area of atherosclerosis of mice treated with various doses of ApoA1.



FIG. 230 is a table showing the inhibition of inflammation factors RNA transcription.



FIG. 231 is a picture of a western blot construct.



FIG. 232 is a graph showing optical density in the APOA1 mouse group.



FIG. 233 is a graph showing optical density in APOA1 vehicle mouse groups.



FIG. 234 is a graph of lipid change in LDLR knock out mice.



FIG. 235 is a graph of lipid change in LDLR knock out mice.



FIG. 236 is a graph depicting the percentage of lesion area for LDLR knock out mice.



FIG. 237-242 are PET/CT scans.



FIG. 243 is a flow chart depicting the process of purifying blood plasma Fraction IV and extracting APOA1 from Fraction IV.



FIG. 244 is a table showing changes in body weight in tumor treated groups with various purified blood plasma products.





DETAIL DESCRIPTION OF THE INVENTION

55 proteins (defined in this application as KH1-55) have been isolated and identified in a variety of human plasmas. 538 functions, processes, and components for these proteins have been determined. In certain embodiments of the current invention the discovery of these proteins and their unique characteristics has led to the development purified plasma products containing KH proteins and methods of use for treating and preventing a wide range of diseases and infections.


KH1—newly discovered protein synthesized by good healthy KH cells is found in Cryoprecipitate which is used to make HemoRAAS® (Factor VIII) and FibroRAAS® (Fibrinogen) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH2—newly discovered protein synthesized by good healthy KH cells is found in Cryoprecipitate which is used to make HemoRAAS® (Factor VIII) and FibroRAAS® (Fibrinogen) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH3—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH4—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH5—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH6—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH7—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH8—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH9—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH10—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 1® through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH11—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH12—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH13—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH14—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH15—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH16—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH17—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH18—newly discovered protein synthesized by good healthy KH cells is found in ProthroRAAS® (Prothrombin complex concentrate) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH19—newly discovered protein synthesized by good healthy KH cells is found in AFCC KH® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH20—newly discovered protein synthesized by good healthy KH cells is found in AFCC KH® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH21—newly discovered protein synthesized by good healthy KH cells is found in fraction IV which is used to manufacture human Albumin, APOA1, Transferrin, Alpha1 Antitripsin, Anti Thrombin III, CP98 and 16 AFOD RAAS 1® through AFOD RAAS 16® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH22—newly discovered protein synthesized by good healthy KH cells is found in fraction IV which is used to manufacture human Albumin, APOA1, Transferrin, Alpha1 Antitripsin, Anti Thrombin III, CP98 and 16 AFOD RAAS 1® through AFOD RAAS 16® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH23—newly discovered protein synthesized by good healthy KH cells is found in fraction IV which is used to manufacture human Albumin, APOA1, Transferrin, Alpha1 Antitripsin, Anti Thrombin III, CP98 and 16 AFOD RAAS 1® through AFOD RAAS 16® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH24—newly discovered protein synthesized by good healthy KH cells is found in AFOD KH® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH25—newly discovered protein synthesized by good healthy KH cells is found in AFOD KH® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH26—newly discovered protein synthesized by good healthy KH cells is found in AFOD KH® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH27—newly discovered protein synthesized by good healthy KH cells is found in AFOD KH® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH28—newly discovered protein synthesized by good healthy KH cells is found in HemoRAAS® (Human Factor VIII) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH29—newly discovered protein synthesized by good healthy KH cells is found in HemoRAAS® (Human Factor VIII) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH30—newly discovered protein synthesized by good healthy KH cells is found in FibroRAAS® (Human Fibrinogen) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH31—newly discovered protein synthesized by good healthy KH cells is found in FibroRAAS® (Human Fibrinogen) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH32—newly discovered protein synthesized by good healthy KH cells is found in FibroRAAS® (Human Fibrinogen) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH33—newly discovered protein synthesized by good healthy KH cells is found in GammaRAAS® (Human Immunoglobulin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH34—newly discovered protein synthesized by good healthy KH cells is found in GammaRAAS® (Human Immunoglobulin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH35—newly discovered protein synthesized by good healthy KH cells is found in GammaRAAS® (Human Immunoglobulin) preventing and/or for treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH36—newly discovered protein synthesized by good healthy KH cells is found in GammaRAAS® (Human Immunoglobulin) for preventing and/or, treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH37—newly discovered protein synthesized by good healthy KH cells is found in GammaRAAS® (Human Immunoglobulin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH38—newly discovered protein synthesized by good healthy KH cells is found in AFCC RAAS 3® through 16 for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH39—newly discovered protein synthesized by good healthy KH cells is found in AFCC RAAS 3® through 16 for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH40—newly discovered protein synthesized by good healthy KH cells is found in AFCC RAAS 3® through 16 for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH41—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 10 through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH42—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 10 through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH43—newly discovered protein synthesized by good healthy KH cells is found in Fraction III which is used to make ProthoRAAS® (Prothrombin complex concentrate), all 16 AFCC RAAS 10 through AFCC RAAS 16® developed products and Immunoglobulin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH44—newly discovered protein synthesized by good healthy KH cells is found in ThrombiRAAS® (Human Thrombin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH45—newly discovered protein synthesized by good healthy KH cells is found in ThrombiRAAS® (Human Thrombin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH46—newly discovered protein synthesized by good healthy KH cells is found in ThrombiRAAS® (Human Thrombin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH47—newly discovered protein synthesized by good healthy KH cells is found in ThrombiRAAS® (Human Thrombin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH48—newly discovered protein synthesized by good healthy KH cells is found in AFOD RAAS 1® through AFOD RAAS 16® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH49—newly discovered protein synthesized by good healthy KH cells is found in AFOD RAAS 10 through AFOD RAAS 16® for preventing and/or treating a wide variety of diseases, cancers infections and other physical conditions and disorders and for maintaining health.


KH50—newly discovered protein synthesized by good healthy KH cells is found in AFOD RAAS 1® through AFOD RAAS 16® for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH51—newly discovered protein synthesized by good healthy KH cells is found in AlbuRAAS® (Human Albumin) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH52—newly discovered protein synthesized by good healthy KH cells is found in FibringluRAAS® (Human high concentrate Fibrinogen) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH53—newly discovered protein synthesized by good healthy KH cells is found in AFCC RAAS 2® (Fraction IV) for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH54—newly discovered protein synthesized by good healthy KH cells is found in Transferrin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


KH55—newly discovered protein synthesized by good healthy KH cells is found in Transferrin for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


Embodiments of the invention include KH proteins found in purified blood plasma products including:


AFOD comprising CP 98 kDa protein, CP reuloplasmin, KRT2 Keratin, type II cytoskeletal epidermal, KH22, KH23, KH24, KH25, APOA1 Apolipoprotein A1, human albumin, transferrin, vimentin, and haptoglobin;


AFCC comprising C3 complement C3, ENO1 Isoform ENO1, TUFM elongation factor, ASS1 argininosuccinate, ANXA2 isoform 2 of annexin A2, glyceraldehyde-3-phosphate dehydrogenase, KHT 86 keratin, type II cuticular HB6, KH20, LDHA isoform 1 of L-lactate dehydrogenase A chain, fibrin beta, KH21, growth inhibiting protein 25, fibrinogen gamma, chain L crystal structure of human fibrinogen, chain A of IgM, chain A crystal structure of the Fab fragment of a human monoclonal Igm cold agglutinin, immunoglobulin light chain, and chain C molecular basis for complement recognition;


AFOD KH comprising CP 98 kDa, CP ceruloplasmin, KRT2 keratin type II cytoskeletal 2 epidermal, KH proteins, APOA1, human albumin, transferrin, vimentin, and haptoglobin;


AFOD RAAS 8 (also known as AFOD RAAS 104) comprising TF serotransferrin derived from fraction III WIG;


AFOD RAAS 101 comprising ALB uncharacterized protein, HPR 31 kDa protein, albumin uncharacterized protein, AIBG isoform 1 of alpha-1B-glycoprotein, HPR haptoglobin, and KH51;


AFOD RAAS 102 (main component of immunoglobulin) comprising 120/E19 IGHV4-31, IGHG1 44 kDa, 191/H18 IGHV4 31, IGHG1 32 kDa, IGHG1 putative uncharacterized protein, DKFZp686G11190, and KH proteins 33-37;


AFOD RAAS 107 comprising protein 1CP 98 kDa including NUP98 and Nup 96, which play a role in bidirectional transport;


AFOD RAAS 109 comprising transferrin and KH proteins 21-27 and KH proteins 48-50; AFOD RAAS 110 comprising anti-thrombin III and KH proteins 22-27 and KH proteins 48-50; and


AFCC RAAS 1 (also known as AFCC RAAS 105) comprising factor II, factor VII, factor IX, factor X, and KH proteins 111-118.


In certain embodiments any two or many of these new found proteins KH1 through KH55 synthesized by good healthy KH cells are combined for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


Embodiments of the invention include any recombinant DNA or many of these new found proteins KH1 through KH55 synthesized by good healthy KH cells are combined for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


In other embodiments any monoclonal or many of these new found proteins KH1 through KH55 synthesized by good healthy KH cells are combined for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


Another embodiment of the invention include processes for the isolating, purifying and concentrating of any KH1 to KH55 protein, and/or the combination of more than of KH1 to KH 55 from all natural products, recombinant DNA, cDNA, or synthesized products for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


Certain embodiments also include the use of any of these KH proteins in combination, either singly or more than two with any natural products, products from recombinant DNA, engineered DNA, cDNA and for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


Other embodiments encompass the use of any of these KH proteins in combination, either singly or more than two KH proteins with any chemical products, medication, small molecules, any future medication for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


Another embodiment of the invention includes the process to isolating and producing and use a mixture of any of these KH proteins in combination, either singly or more than two KH proteins with any natural products or synthesized products as part of nutrition, food, and/or supplemental products in any capacity for preventing and/or treating a wide variety of diseases, cancers, infections and other physical conditions and disorders and for maintaining health.


538 functions have been identified for the 55 KH proteins, which provide them with unique characteristics for treating a wide range of disease, infection, and other cellular disturbances as expressed in some embodiments of the invention as described.


















Fraction - P






(process), C




(component), F
Sequence


Number
GI code
(function)
name
Sequence desc.







KH 1
21749960
cryopaste
gi|21749960
dock4_humandedicator of






cytokinesis protein 4






os = homo sapiens gn = dock4






pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
F
GO: 0005102
receptor binding



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
P
GO: 0043547
positive regulation of



cytokinesis protein 4


GTPase activity



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
P
GO: 0016477
cell migration



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
P
GO: 0007165
signal transduction



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
P
GO: 0006935
chemotaxis



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
C
GO: 0005737
cytoplasm



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
F
GO: 0005083
small GTPase regulator



cytokinesis protein 4


activity



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
F
GO: 0019904
protein domain specific



cytokinesis protein 4


binding



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
P
GO: 0048583
regulation of response to



cytokinesis protein 4


stimulus



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
F
GO: 0005096
GTPase activator activity



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
F
GO: 0051020
GTPase binding



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


gi|21749960
dock4_humandedicator of
C
GO: 0016020
membrane



cytokinesis protein 4



os = homo sapiens gn = dock4



pe = 1 sv = 3


KH 2
215415640
cryopaste
gi|215415640
apoa1_humanapolipoprotein






a-i os = homo sapiens






gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0070508
cholesterol import



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
C
GO: 0030139
endocytic vesicle



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0050728
negative regulation of



a-i os = homo sapiens


inflammatory response



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0033344
cholesterol efflux



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0034115
negative regulation of



a-i os = homo sapiens


heterotypic cell-cell



gn = apoa1 pe = 1 sv = 1


adhesion


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0018206
peptidyl-methionine



a-i os = homo sapiens


modification



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0042157
lipoprotein metabolic



a-i os = homo sapiens


process



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0043691
reverse cholesterol transport



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0005543
phospholipid binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0002740
negative regulation of



a-i os = homo sapiens


cytokine secretion involved



gn = apoa1 pe = 1 sv = 1


in immune response


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0008203
cholesterol metabolic



a-i os = homo sapiens


process



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0050713
negative regulation of



a-i os = homo sapiens


interleukin-1 beta secretion



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0018158
protein oxidation



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0006656
phosphatidylcholine



a-i os = homo sapiens


biosynthetic process



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0001540
beta-amyloid binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0060228
phosphatidylcholine-sterol



a-i os = homo sapiens


O-acyltransferase activator



gn = apoa1 pe = 1 sv = 1


activity


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0042632
cholesterol homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0015485
cholesterol binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0042060
wound healing



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0034191
apolipoprotein A-I receptor



a-i os = homo sapiens


binding



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0042802
identical protein binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0010903
negative regulation of very-



a-i os = homo sapiens


low-density lipoprotein



gn = apoa1 pe = 1 sv = 1


particle remodeling


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0010804
negative regulation of tumor



a-i os = homo sapiens


necrosis factor-mediated



gn = apoa1 pe = 1 sv = 1


signaling pathway


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0034380
high-density lipoprotein



a-i os = homo sapiens


particle assembly



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0007186
G-protein coupled receptor



a-i os = homo sapiens


signaling pathway



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0050821
protein stabilization



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
C
GO: 0034361
very-low-density lipoprotein



a-i os = homo sapiens


particle



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0032488
Cdc42 protein signal



a-i os = homo sapiens


transduction



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0060354
negative regulation of cell



a-i os = homo sapiens


adhesion molecule



gn = apoa1 pe = 1 sv = 1


production


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0055091
phospholipid homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0010873
positive regulation of



a-i os = homo sapiens


cholesterol esterification



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0017127
cholesterol transporter



a-i os = homo sapiens


activity



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0019899
enzyme binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
F
GO: 0070653
high-density lipoprotein



a-i os = homo sapiens


particle receptor binding



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0070328
triglyceride homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
C
GO: 0034366
spherical high-density



a-i os = homo sapiens


lipoprotein particle



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0033700
phospholipid efflux



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415640
apoa1_humanapolipoprotein
P
GO: 0051345
positive regulation of



a-i os = homo sapiens


hydrolase activity



gn = apoa1 pe = 1 sv = 1


KH 3
215415638
Fr III
gi|215415638
apoa1_humanapolipoprotein






a-i os = homo sapiens






gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0070508
cholesterol import



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
C
GO: 0030139
endocytic vesicle



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0050728
negative regulation of



a-i os = homo sapiens


inflammatory response



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0033344
cholesterol efflux



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0034115
negative regulation of



a-i os = homo sapiens


heterotypic cell-cell



gn = apoa1 pe = 1 sv = 1


adhesion


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0018206
peptidyl-methionine



a-i os = homo sapiens


modification



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0042157
lipoprotein metabolic



a-i os = homo sapiens


process



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0043691
reverse cholesterol transport



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0005543
phospholipid binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0002740
negative regulation of



a-i os = homo sapiens


cytokine secretion involved



gn = apoa1 pe = 1 sv = 1


in immune response


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0008203
cholesterol metabolic



a-i os = homo sapiens


process



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0050713
negative regulation of



a-i os = homo sapiens


interleukin-1 beta secretion



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0018158
protein oxidation



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0006656
phosphatidylcholine



a-i os = homo sapiens


biosynthetic process



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0001540
beta-amyloid binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0060228
phosphatidylcholine-sterol



a-i os = homo sapiens


O-acyltransferase activator



gn = apoa1 pe = 1 sv = 1


activity


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0042632
cholesterol homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0015485
cholesterol binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0042060
wound healing



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0034191
apolipoprotein A-I receptor



a-i os = homo sapiens


binding



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0042802
identical protein binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0010903
negative regulation of very-



a-i os = homo sapiens


low-density lipoprotein



gn = apoa1 pe = 1 sv = 1


particle remodeling


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0010804
negative regulation of tumor



a-i os = homo sapiens


necrosis factor-mediated



gn = apoa1 pe = 1 sv = 1


signaling pathway


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0034380
high-density lipoprotein



a-i os = homo sapiens


particle assembly



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0007186
G-protein coupled receptor



a-i os = homo sapiens


signaling pathway



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0050821
protein stabilization



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
C
GO: 0034361
very-low-density lipoprotein



a-i os = homo sapiens


particle



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0032488
Cdc42 protein signal



a-i os = homo sapiens


transduction



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0060354
negative regulation of cell



a-i os = homo sapiens


adhesion molecule



gn = apoa1 pe = 1 sv = 1


production


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0055091
phospholipid homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0010873
positive regulation of



a-i os = homo sapiens


cholesterol esterification



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0017127
cholesterol transporter



a-i os = homo sapiens


activity



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0019899
enzyme binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
F
GO: 0070653
high-density lipoprotein



a-i os = homo sapiens


particle receptor binding



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0070328
triglyceride homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
C
GO: 0034366
spherical high-density



a-i os = homo sapiens


lipoprotein particle



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0033700
phospholipid efflux



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


gi|215415638
apoa1_humanapolipoprotein
P
GO: 0051345
positive regulation of



a-i os = homo sapiens


hydrolase activity



gn = apoa1 pe = 1 sv = 1


KH 4
40044478
Fr III


KH 5
194383496
Fr III
gi|194383496
thrb_humanprothrombin






os = homo sapiens gn = f2






pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0032879
regulation of localization



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0048468
cell development



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 2000026
regulation of multicellular



os = homo sapiens gn = f2


organismal development



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0010557
positive regulation of



os = homo sapiens gn = f2


macromolecule biosynthetic



pe = 1 sv = 2


process


gi|194383496
thrb_humanprothrombin
P
GO: 0030194
positive regulation of blood



os = homo sapiens gn = f2


coagulation



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
F
GO: 0005102
receptor binding



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0009967
positive regulation of signal



os = homo sapiens gn = f2


transduction



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
C
GO: 0005615
extracellular space



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0030168
platelet activation



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
F
GO: 0008236
serine-type peptidase activity



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0016477
cell migration



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0006508
proteolysis



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0001934
positive regulation of protein



os = homo sapiens gn = f2


phosphorylation



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0007166
cell surface receptor



os = homo sapiens gn = f2


signaling pathway



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0048523
negative regulation of



os = homo sapiens gn = f2


cellular process



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0006810
transport



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0042730
fibrinolysis



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
C
GO: 0005622
intracellular



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0048731
system development



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
C
GO: 0016020
membrane



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194383496
thrb_humanprothrombin
P
GO: 0051480
cytosolic calcium ion



os = homo sapiens gn = f2


homeostasis



pe = 1 sv = 2


KH 6
28071026
Fr III
gi|28071026
ighm_humanig mu chain c






region os = homo sapiens






gn = ighm pe = 1 sv = 3


gi|28071026
ighm_humanig mu chain c
F
GO: 0005488
binding



region os = homo sapiens



gn = ighm pe = 1 sv = 3


gi|28071026
ighm_humanig mu chain c
C
GO: 0044464
cell part



region os = homo sapiens



gn = ighm pe = 1 sv = 3


gi|28071026
ighm_humanig mu chain c
C
GO: 0016020
membrane



region os = homo sapiens



gn = ighm pe = 1 sv = 3


gi|28071026
ighm_humanig mu chain c
P
GO: 0006955
immune response



region os = homo sapiens



gn = ighm pe = 1 sv = 3


KH 7
300621695
Fr III
gi|300621695
ighm_humanig mu chain c






region os = homo sapiens






gn = ighm pe = 1 sv = 3


gi|300621695
ighm_humanig mu chain c
P
GO: 0006955
immune response



region os = homo sapiens



gn = ighm pe = 1 sv = 3


KH 8
1335098
Fr III
gi|1335098
hemo_humanhemopexin






os = homo sapiens gn = hpx






pe = 1 sv = 2


gi|1335098
hemo_humanhemopexin
P
GO: 0008152
metabolic process



os = homo sapiens gn = hpx



pe = 1 sv = 2


gi|1335098
hemo_humanhemopexin
P
GO: 0051179
localization



os = homo sapiens gn = hpx



pe = 1 sv = 2


gi|1335098
hemo_humanhemopexin
C
GO: 0005615
extracellular space



os = homo sapiens gn = hpx



pe = 1 sv = 2


gi|1335098
hemo_humanhemopexin
F
GO: 0005515
protein binding



os = homo sapiens gn = hpx



pe = 1 sv = 2


gi|1335098
hemo_humanhemopexin
P
GO: 0048522
positive regulation of



os = homo sapiens gn = hpx


cellular process



pe = 1 sv = 2


gi|1335098
hemo_humanhemopexin
P
GO: 0050896
response to stimulus



os = homo sapiens gn = hpx



pe = 1 sv = 2


KH 9
10434804
Fr III
gi|10434804
mthsd_humanmethenyltetrahydrofolate






synthase domain-






containing protein os = homo







sapiens gn = mthfsd pe = 1







sv = 2


gi|10434804
mthsd_humanmethenyltetrahydrofolate
F
GO: 0005524
ATP binding



synthase domain-



containing protein os = homo




sapiens gn = mthfsd pe = 1




sv = 2


gi|10434804
mthsd_humanmethenyltetrahydrofolate
P
GO: 0009396
folic acid-containing



synthase domain-


compound biosynthetic



containing protein os = homo


process




sapiens gn = mthfsd pe = 1




sv = 2


gi|10434804
mthsd_humanmethenyltetrahydrofolate
F
GO: 0030272
5-formyltetrahydrofolate



synthase domain-


cyclo-ligase activity



containing protein os = homo




sapiens gn = mthfsd pe = 1




sv = 2


KH 10
221044726
Fr III
gi|221044726
hemo_humanhemopexin






os = homo sapiens gn = hpx






pe = 1 sv = 2


gi|221044726
hemo_humanhemopexin
F
GO: 0005515
protein binding



os = homo sapiens gn = hpx



pe = 1 sv = 2


gi|221044726
hemo_humanhemopexin
C
GO: 0005615
extracellular space



os = homo sapiens gn = hpx



pe = 1 sv = 2


gi|221044726
hemo_humanhemopexin
P
GO: 0009987
cellular process



os = homo sapiens gn = hpx



pe = 1 sv = 2


gi|221044726
hemo_humanhemopexin
P
GO: 0065007
biological regulation



os = homo sapiens gn = hpx



pe = 1 sv = 2


KH 11
215415638
PCC
same as





KH 3


KH 12
189066554
PCC
gi|189066554
thrb_humanprothrombin






os = homo sapiens gn = f2






pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
C
GO: 0044446
intracellular organelle part



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 0048712
negative regulation of



os = homo sapiens gn = f2


astrocyte differentiation



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
C
GO: 0043233
organelle lumen



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 0030194
positive regulation of blood



os = homo sapiens gn = f2


coagulation



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
F
GO: 0005102
receptor binding



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 2000379
positive regulation of



os = homo sapiens gn = f2


reactive oxygen species



pe = 1 sv = 2


metabolic process


gi|189066554
thrb_humanprothrombin
P
GO: 0045861
negative regulation of



os = homo sapiens gn = f2


proteolysis



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
C
GO: 0005615
extracellular space



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 0030168
platelet activation



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 1900738
positive regulation of



os = homo sapiens gn = f2


phospholipase C-activating



pe = 1 sv = 2


G-protein coupled receptor






signaling pathway


gi|189066554
thrb_humanprothrombin
P
GO: 0016477
cell migration



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
C
GO: 0043231
intracellular membrane-



os = homo sapiens gn = f2


bounded organelle



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 0001934
positive regulation of protein



os = homo sapiens gn = f2


phosphorylation



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
C
GO: 0005886
plasma membrane



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
F
GO: 0070053
thrombospondin receptor



os = homo sapiens gn = f2


activity



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 0051281
positive regulation of release



os = homo sapiens gn = f2


of sequestered calcium ion



pe = 1 sv = 2


into cytosol


gi|189066554
thrb_humanprothrombin
F
GO: 0004252
serine-type endopeptidase



os = homo sapiens gn = f2


activity



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 0042730
fibrinolysis



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
C
GO: 0044444
cytoplasmic part



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|189066554
thrb_humanprothrombin
P
GO: 0032967
positive regulation of



os = homo sapiens gn = f2


collagen biosynthetic



pe = 1 sv = 2


process


KH 13
194391084
PCC
gi|194391084
kng1_humankininogen-1






os = homo sapiens gn = kng1






pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
F
GO: 0005515
protein binding



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0055065
metal ion homeostasis



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0051241
negative regulation of



os = homo sapiens gn = kng1


multicellular organismal



pe = 1 sv = 2


process


gi|194391084
kng1_humankininogen-1
P
GO: 0007596
blood coagulation



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
C
GO: 0043229
intracellular organelle



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0048523
negative regulation of



os = homo sapiens gn = kng1


cellular process



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0008152
metabolic process



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0003008
system process



os = homo sapiens gn = kng1



pe = 1 sv = 2


KH 14
158255114
PCC
gi|158255114
kng1_humankininogen-1






os = homo sapiens gn = kng1






pe = 1 sv = 2


gi|158255114
kng1_humankininogen-1
F
GO: 0005515
protein binding



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|158255114
kng1_humankininogen-1
P
GO: 0055065
metal ion homeostasis



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|158255114
kng1_humankininogen-1
P
GO: 0051241
negative regulation of



os = homo sapiens gn = kng1


multicellular organismal



pe = 1 sv = 2


process


gi|158255114
kng1_humankininogen-1
P
GO: 0007596
blood coagulation



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|158255114
kng1_humankininogen-1
C
GO: 0043229
intracellular organelle



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|158255114
kng1_humankininogen-1
P
GO: 0048523
negative regulation of



os = homo sapiens gn = kng1


cellular process



pe = 1 sv = 2


gi|158255114
kng1_humankininogen-1
P
GO: 0008152
metabolic process



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|158255114
kng1_humankininogen-1
P
GO: 0003008
system process



os = homo sapiens gn = kng1



pe = 1 sv = 2


KH 15
213506121
PCC
gi|213506121
kng1_humankininogen-1






os = homo sapiens gn = kng1






pe = 1 sv = 2


gi|213506121
kng1_humankininogen-1
F
GO: 0005515
protein binding



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506121
kng1_humankininogen-1
P
GO: 0055065
metal ion homeostasis



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506121
kng1_humankininogen-1
P
GO: 0051241
negative regulation of



os = homo sapiens gn = kng1


multicellular organismal



pe = 1 sv = 2


process


gi|213506121
kng1_humankininogen-1
P
GO: 0007596
blood coagulation



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506121
kng1_humankininogen-1
C
GO: 0043229
intracellular organelle



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506121
kng1_humankininogen-1
P
GO: 0048523
negative regulation of



os = homo sapiens gn = kng1


cellular process



pe = 1 sv = 2


gi|213506121
kng1_humankininogen-1
P
GO: 0008152
metabolic process



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506121
kng1_humankininogen-1
P
GO: 0003008
system process



os = homo sapiens gn = kng1



pe = 1 sv = 2


KH 16
213506103
PCC
gi|213506103
kng1_humankininogen-1






os = homo sapiens gn = kng1






pe = 1 sv = 2


gi|213506103
kng1_humankininogen-1
F
GO: 0005515
protein binding



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506103
kng1_humankininogen-1
P
GO: 0055065
metal ion homeostasis



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506103
kng1_humankininogen-1
P
GO: 0051241
negative regulation of



os = homo sapiens gn = kng1


multicellular organismal



pe = 1 sv = 2


process


gi|213506103
kng1_humankininogen-1
P
GO: 0007596
blood coagulation



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506103
kng1_humankininogen-1
C
GO: 0043229
intracellular organelle



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506103
kng1_humankininogen-1
P
GO: 0048523
negative regulation of



os = homo sapiens gn = kng1


cellular process



pe = 1 sv = 2


gi|213506103
kng1_humankininogen-1
P
GO: 0008152
metabolic process



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|213506103
kng1_humankininogen-1
P
GO: 0003008
system process



os = homo sapiens gn = kng1



pe = 1 sv = 2


KH 17
194376310
PCC
gi|194376310
cytoplasmic 1 os = homo







sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
P
GO: 0009888
tissue development




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
P
GO: 0030048
actin filament-based




sapiens gn = actb pe = 1 sv = 1



movement


gi|194376310
cytoplasmic 1 os = homo
P
GO: 0003012
muscle system process




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
C
GO: 0030017
sarcomere




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
P
GO: 0030239
myofibril assembly




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
P
GO: 0044238
primary metabolic process




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
C
GO: 0005884
actin filament




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
P
GO: 0072358
cardiovascular system




sapiens gn = actb pe = 1 sv = 1



development


gi|194376310
cytoplasmic 1 os = homo
P
GO: 0044237
cellular metabolic process




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
P
GO: 0048513
organ development




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
F
GO: 0005515
protein binding




sapiens gn = actb pe = 1 sv = 1



gi|194376310
cytoplasmic 1 os = homo
P
GO: 0042221
response to chemical




sapiens gn = actb pe = 1 sv = 1



stimulus


gi|194376310
cytoplasmic 1 os = homo
P
GO: 0008015
blood circulation




sapiens gn = actb pe = 1 sv = 1



KH 18
194388064
PCC
gi|194388064
cytoplasmic 2 os = homo







sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0009888
tissue development




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0030048
actin filament-based




sapiens gn = actg1 pe = 1 sv = 1



movement


gi|194388064
cytoplasmic 2 os = homo
P
GO: 0003012
muscle system process




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
C
GO: 0030017
sarcomere




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0030239
myofibril assembly




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0044238
primary metabolic process




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
C
GO: 0005884
actin filament




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0072358
cardiovascular system




sapiens gn = actg1 pe = 1 sv = 1



development


gi|194388064
cytoplasmic 2 os = homo
P
GO: 0044237
cellular metabolic process




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0048513
organ development




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0042221
response to chemical




sapiens gn = actg1 pe = 1 sv = 1



stimulus


gi|194388064
cytoplasmic 2 os = homo
F
GO: 0008092
cytoskeletal protein binding




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0065008
regulation of biological




sapiens gn = actg1 pe = 1 sv = 1



quality


gi|194388064
cytoplasmic 2 os = homo
C
GO: 0044451
nucleoplasm part




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
P
GO: 0008015
blood circulation




sapiens gn = actg1 pe = 1 sv = 1



gi|194388064
cytoplasmic 2 os = homo
F
GO: 0019899
enzyme binding




sapiens gn = actg1 pe = 1 sv = 1



gi|194391084
kng1_humankininogen-1
F
GO: 0005515
protein binding



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0055065
metal ion homeostasis



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0051241
negative regulation of



os = homo sapiens gn = kng1


multicellular organismal



pe = 1 sv = 2


process


gi|194391084
kng1_humankininogen-1
P
GO: 0007596
blood coagulation



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
C
GO: 0043229
intracellular organelle



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0048523
negative regulation of



os = homo sapiens gn = kng1


cellular process



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0008152
metabolic process



os = homo sapiens gn = kng1



pe = 1 sv = 2


gi|194391084
kng1_humankininogen-1
P
GO: 0003008
system process



os = homo sapiens gn = kng1



pe = 1 sv = 2


KH 19
IPI00964149
AFCC
IPI00964149
pacrl_humanpacrg-like






protein os = homo sapiens






gn = pacrgl pe = 1 sv = 2


KH 20
IPI00966721
AFCC
IPI00966721
ce028_humantransmembrane






protein c5orf28 os = homo







sapiens gn = c5orf28 pe = 2







sv = 1


IPI00966721
ce028_humantransmembrane
C
GO: 0016021
integral to membrane



protein c5orf28 os = homo




sapiens gn = c5orf28 pe = 2




sv = 1


KH 21
IPI00966826
FrIV
IPI00966826
pds5a_humansister






chromatid cohesion protein






pds5 homolog a os = homo







sapiens gn = pds5a pe = 1 sv = 1



IPI00966826
pds5a_humansister
P
GO: 0008156
negative regulation of DNA



chromatid cohesion protein


replication



pds5 homolog a os = homo




sapiens gn = pds5a pe = 1 sv = 1



IPI00966826
pds5a_humansister
C
GO: 0005730
nucleolus



chromatid cohesion protein



pds5 homolog a os = homo




sapiens gn = pds5a pe = 1 sv = 1



IPI00966826
pds5a_humansister
C
GO: 0000785
chromatin



chromatid cohesion protein



pds5 homolog a os = homo




sapiens gn = pds5a pe = 1 sv = 1



IPI00966826
pds5a_humansister
F
GO: 0005515
protein binding



chromatid cohesion protein



pds5 homolog a os = homo




sapiens gn = pds5a pe = 1 sv = 1



IPI00966826
pds5a_humansister
P
GO: 0008283
cell proliferation



chromatid cohesion protein



pds5 homolog a os = homo




sapiens gn = pds5a pe = 1 sv = 1



IPI00966826
pds5a_humansister
C
GO: 0005886
plasma membrane



chromatid cohesion protein



pds5 homolog a os = homo




sapiens gn = pds5a pe = 1 sv = 1



KH 22
IPI00760788
FrIV
IPI00760788
klh22_humankelch-like






protein 22 os = homo sapiens






gn = klhl22 pe = 1 sv = 2


IPI00760788
klh22_humankelch-like
P
GO: 0051301
cell division



protein 22 os = homo sapiens



gn = klhl22 pe = 1 sv = 2


IPI00760788
klh22_humankelch-like
C
GO: 0031463
Cul3-RING ubiquitin ligase



protein 22 os = homo sapiens


complex



gn = klhl22 pe = 1 sv = 2


KH 23
IPI00917278
FrIV


KH 24
IPI00966721
AFOD
same as





KH 20


KH 25
IPI01012037
AFOD
IPI01012037
mcm8_humandna helicase






mcm8 os = homo sapiens






gn = mcm8 pe = 1 sv = 2


IPI01012037
mcm8_humandna helicase
P
GO: 0051329
interphase of mitotic cell



mcm8 os = homo sapiens


cycle



gn = mcm8 pe = 1 sv = 2


IPI01012037
mcm8_humandna helicase
P
GO: 0034645
cellular macromolecule



mcm8 os = homo sapiens


biosynthetic process



gn = mcm8 pe = 1 sv = 2


IPI01012037
mcm8_humandna helicase
P
GO: 0090304
nucleic acid metabolic



mcm8 os = homo sapiens


process



gn = mcm8 pe = 1 sv = 2


KH 26
IPI00940730
AFOD
IPI00940730
enoph_humanenolase-






phosphatase e1 os = homo







sapiens gn = enoph1 pe = 1







sv = 1


IPI00940730
enoph_humanenolase-
P
GO: 0019509
L-methionine salvage from



phosphatase e1 os = homo


methylthioadenosine




sapiens gn = enoph1 pe = 1




sv = 1


IPI00940730
enoph_humanenolase-
F
GO: 0043874
acireductone synthase



phosphatase e1 os = homo


activity




sapiens gn = enoph1 pe = 1




sv = 1


KH 27
IPI00977191
AFOD


KH 28
IPI00022434
HemoRAAS
IPI00022434
albu_humanserum albumin






os = homo sapiens gn = alb






pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0008202
steroid metabolic process



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0051704
multi-organism process



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
C
GO: 0044446
intracellular organelle part



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0051641
cellular localization



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0051716
cellular response to stimulus



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
F
GO: 0008289
lipid binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0043069
negative regulation of



os = homo sapiens gn = alb


programmed cell death



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0044260
cellular macromolecule



os = homo sapiens gn = alb


metabolic process



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0031667
response to nutrient levels



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
C
GO: 0043231
intracellular membrane-



os = homo sapiens gn = alb


bounded organelle



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0044281
small molecule metabolic



os = homo sapiens gn = alb


process



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
F
GO: 0005515
protein binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0006810
transport



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0065008
regulation of biological



os = homo sapiens gn = alb


quality



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0007154
cell communication



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
F
GO: 0019842
vitamin binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0006950
response to stress



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
C
GO: 0044444
cytoplasmic part



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
P
GO: 0032501
multicellular organismal



os = homo sapiens gn = alb


process



pe = 1 sv = 2


IPI00022434
albu_humanserum albumin
C
GO: 0044421
extracellular region part



os = homo sapiens gn = alb



pe = 1 sv = 2


KH 29
IPI00022434
HemoRAAS
same as





KH 28


KH 30
IPI00219713
FibroRAAS
IPI00219713
fibg_humanfibrinogen






gamma chain os = homo







sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
P
GO: 0009987
cellular process



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
C
GO: 0009897
external side of plasma



gamma chain os = homo


membrane




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
F
GO: 0043499
eukaryotic cell surface



gamma chain os = homo


binding




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
C
GO: 0005615
extracellular space



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
C
GO: 0031091
platelet alpha granule



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
P
GO: 0032501
multicellular organismal



gamma chain os = homo


process




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
P
GO: 0065007
biological regulation



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
P
GO: 0051592
response to calcium ion



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



KH 31
IPI00219713
FibroRAAS
same as





KH 30


KH 32
IPI00220327
FibroRAAS
IPI00220327
type ii cytoskeletal 1






os = homo sapiens gn = krt1






pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
P
GO: 0009987
cellular process



os = homo sapiens gn = krt1



pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
P
GO: 0048731
system development



os = homo sapiens gn = krt1



pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
P
GO: 0009888
tissue development



os = homo sapiens gn = krt1



pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
C
GO: 0005856
cytoskeleton



os = homo sapiens gn = krt1



pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
F
GO: 0005515
protein binding



os = homo sapiens gn = krt1



pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
P
GO: 0001867
complement activation,



os = homo sapiens gn = krt1


lectin pathway



pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
F
GO: 0030246
carbohydrate binding



os = homo sapiens gn = krt1



pe = 1 sv = 6


IPI00220327
type ii cytoskeletal 1
C
GO: 0016020
membrane



os = homo sapiens gn = krt1



pe = 1 sv = 6


KH 33
IPI00029739
GammaRAAS
IPI00029739
cfah_humancomplement






factor h os = homo sapiens






gn = cfh pe = 1 sv = 4


IPI00029739
cfah_humancomplement
P
GO: 0030449
regulation of complement



factor h os = homo sapiens


activation



gn = cfh pe = 1 sv = 4


IPI00029739
cfah_humancomplement
P
GO: 0045087
innate immune response



factor h os = homo sapiens



gn = cfh pe = 1 sv = 4


KH 34
IPI00384853
GammaRAAS


KH 35
IPI00479708
GammaRAAS
IPI00479708
ighm_humanig mu chain c






region os = homo sapiens






gn = ighm pe = 1 sv = 3


IPI00479708
ighm_humanig mu chain c
F
GO: 0005488
binding



region os = homo sapiens



gn = ighm pe = 1 sv = 3


IPI00479708
ighm_humanig mu chain c
C
GO: 0044464
cell part



region os = homo sapiens



gn = ighm pe = 1 sv = 3


IPI00479708
ighm_humanig mu chain c
C
GO: 0016020
membrane



region os = homo sapiens



gn = ighm pe = 1 sv = 3


IPI00479708
ighm_humanig mu chain c
P
GO: 0006955
immune response



region os = homo sapiens



gn = ighm pe = 1 sv = 3


KH 36
IPI00298497
GammaRAAS
IPI00298497
fibb_humanfibrinogen beta






chain os = homo sapiens






gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
F
GO: 0051087
chaperone binding



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
P
GO: 0051592
response to calcium ion



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
C
GO: 0005615
extracellular space



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
P
GO: 0051179
localization



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
C
GO: 0031091
platelet alpha granule



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
C
GO: 0009897
external side of plasma



chain os = homo sapiens


membrane



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
P
GO: 0050794
regulation of cellular process



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
P
GO: 0006950
response to stress



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
F
GO: 0043499
eukaryotic cell surface



chain os = homo sapiens


binding



gn = fgb pe = 1 sv = 2


IPI00298497
fibb_humanfibrinogen beta
P
GO: 0032501
multicellular organismal



chain os = homo sapiens


process



gn = fgb pe = 1 sv = 2


KH 37
IPI00021841
GammaRAAS
IPI00021841
apoa1_humanapolipoprotein






a-i os = homo sapiens






gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0070508
cholesterol import



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
C
GO: 0030139
endocytic vesicle



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0050728
negative regulation of



a-i os = homo sapiens


inflammatory response



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0033344
cholesterol efflux



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0034115
negative regulation of



a-i os = homo sapiens


heterotypic cell-cell



gn = apoa1 pe = 1 sv = 1


adhesion


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0018206
peptidyl-methionine



a-i os = homo sapiens


modification



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0042157
lipoprotein metabolic



a-i os = homo sapiens


process



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0043691
reverse cholesterol transport



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0005543
phospholipid binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0002740
negative regulation of



a-i os = homo sapiens


cytokine secretion involved



gn = apoa1 pe = 1 sv = 1


in immune response


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0008203
cholesterol metabolic



a-i os = homo sapiens


process



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0050713
negative regulation of



a-i os = homo sapiens


interleukin-1 beta secretion



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0018158
protein oxidation



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0006656
phosphatidylcholine



a-i os = homo sapiens


biosynthetic process



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0001540
beta-amyloid binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0060228
phosphatidylcholine-sterol



a-i os = homo sapiens


O-acyltransferase activator



gn = apoa1 pe = 1 sv = 1


activity


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0042632
cholesterol homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0015485
cholesterol binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0042060
wound healing



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0034191
apolipoprotein A-I receptor



a-i os = homo sapiens


binding



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0042802
identical protein binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0010903
negative regulation of very-



a-i os = homo sapiens


low-density lipoprotein



gn = apoa1 pe = 1 sv = 1


particle remodeling


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0010804
negative regulation of tumor



a-i os = homo sapiens


necrosis factor-mediated



gn = apoa1 pe = 1 sv = 1


signaling pathway


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0034380
high-density lipoprotein



a-i os = homo sapiens


particle assembly



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0007186
G-protein coupled receptor



a-i os = homo sapiens


signaling pathway



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0050821
protein stabilization



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
C
GO: 0034361
very-low-density lipoprotein



a-i os = homo sapiens


particle



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0032488
Cdc42 protein signal



a-i os = homo sapiens


transduction



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0060354
negative regulation of cell



a-i os = homo sapiens


adhesion molecule



gn = apoa1 pe = 1 sv = 1


production


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0055091
phospholipid homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0010873
positive regulation of



a-i os = homo sapiens


cholesterol esterification



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0017127
cholesterol transporter



a-i os = homo sapiens


activity



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0019899
enzyme binding



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
F
GO: 0070653
high-density lipoprotein



a-i os = homo sapiens


particle receptor binding



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0070328
triglyceride homeostasis



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
C
GO: 0034366
spherical high-density



a-i os = homo sapiens


lipoprotein particle



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0033700
phospholipid efflux



a-i os = homo sapiens



gn = apoa1 pe = 1 sv = 1


IPI00021841
apoa1_humanapolipoprotein
P
GO: 0051345
positive regulation of



a-i os = homo sapiens


hydrolase activity



gn = apoa1 pe = 1 sv = 1


KH 38
IPI00783987
AFCC
IPI00783987
co3_humancomplement c3






os = homo sapiens gn = c3






pe = 1 sv = 2


IPI00783987
co3_humancomplement c3
C
GO: 0044464
cell part



os = homo sapiens gn = c3



pe = 1 sv = 2


IPI00783987
co3_humancomplement c3
P
GO: 0010575
positive regulation vascular



os = homo sapiens gn = c3


endothelial growth factor



pe = 1 sv = 2


production


IPI00783987
co3_humancomplement c3
P
GO: 0030449
regulation of complement



os = homo sapiens gn = c3


activation



pe = 1 sv = 2


IPI00783987
co3_humancomplement c3
P
GO: 0007165
signal transduction



os = homo sapiens gn = c3



pe = 1 sv = 2


IPI00783987
co3_humancomplement c3
P
GO: 0045087
innate immune response



os = homo sapiens gn = c3



pe = 1 sv = 2


IPI00783987
co3_humancomplement c3
F
GO: 0005515
protein binding



os = homo sapiens gn = c3



pe = 1 sv = 2


IPI00783987
co3_humancomplement c3
C
GO: 0016020
membrane



os = homo sapiens gn = c3



pe = 1 sv = 2


KH 39
IPI00878282
AFCC
IPI00878282
albu_humanserum albumin






os = homo sapiens gn = alb






pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0008202
steroid metabolic process



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
F
GO: 0051087
chaperone binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
C
GO: 0044446
intracellular organelle part



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
F
GO: 0015643
toxin binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0044260
cellular macromolecule



os = homo sapiens gn = alb


metabolic process



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
C
GO: 0005615
extracellular space



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0051659
maintenance of



os = homo sapiens gn = alb


mitochondrion location



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
F
GO: 0008144
drug binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
C
GO: 0043231
intracellular membrane-



os = homo sapiens gn = alb


bounded organelle



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0044281
small molecule metabolic



os = homo sapiens gn = alb


process



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
F
GO: 0005504
fatty acid binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0042221
response to chemical



os = homo sapiens gn = alb


stimulus



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
F
GO: 0003677
DNA binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0009267
cellular response to



os = homo sapiens gn = alb


starvation



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
F
GO: 0030170
pyridoxal phosphate binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0006810
transport



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
F
GO: 0019825
oxygen binding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0050878
regulation of body fluid



os = homo sapiens gn = alb


levels



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0043066
negative regulation of



os = homo sapiens gn = alb


apoptotic process



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
C
GO: 0044444
cytoplasmic part



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0009611
response to wounding



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0019836
hemolysis by symbiont of



os = homo sapiens gn = alb


host erythrocytes



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
P
GO: 0006955
immune response



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00878282
albu_humanserum albumin
C
GO: 0019814
immunoglobulin complex



os = homo sapiens gn = alb



pe = 1 sv = 2


IPI00784842
ighg1_humanig gamma-1
P
GO: 0050776
regulation of immune



chain c region os = homo


response




sapiens gn = ighg1 pe = 1 sv = 1



IPI00784842
ighg1_humanig gamma-1
F
GO: 0005515
protein binding



chain c region os = homo




sapiens gn = ighg1 pe = 1 sv = 1



KH 40
IPI00784842
AFCC
IPI00784842
ighg1_humanig gamma-1






chain c region os = homo







sapiens gn = ighg1 pe = 1 sv = 1



IPI00784842
ighg1_humanig gamma-1
P
GO: 0050776
regulation of immune



chain c region os = homo


response




sapiens gn = ighg1 pe = 1 sv = 1



IPI00784842
ighg1_humanig gamma-1
F
GO: 0005515
protein binding



chain c region os = homo




sapiens gn = ighg1 pe = 1 sv = 1



KH 41
IPI00022434
Fraction III-
same as




II
KH 28


KH 42
IPI00298497
Fraction III
same as





KH 36


KH 43
IPI00965713
Fraction III
IPI00965713
fibb_humanfibrinogen beta






chain os = homo sapiens






gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
P
GO: 0042221
response to chemical



chain os = homo sapiens


stimulus



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
F
GO: 0005515
protein binding



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
C
GO: 0005615
extracellular space



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
P
GO: 0051179
localization



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
C
GO: 0031091
platelet alpha granule



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
C
GO: 0044425
membrane part



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
P
GO: 0050794
regulation of cellular process



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
P
GO: 0006950
response to stress



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
C
GO: 0005886
plasma membrane



chain os = homo sapiens



gn = fgb pe = 1 sv = 2


IPI00965713
fibb_humanfibrinogen beta
P
GO: 0032501
multicellular organismal



chain os = homo sapiens


process



gn = fgb pe = 1 sv = 2


KH 44
IPI00645363
FibringluRAAS ®
IPI00645363
ighg1_humanig gamma-1




Human

chain c region os = homo




Thrombin


sapiens gn = ighg1 pe = 1 sv = 1



IPI00645363
ighg1_humanig gamma-1
P
GO: 0050776
regulation of immune



chain c region os = homo


response




sapiens gn = ighg1 pe = 1 sv = 1



IPI00645363
ighg1_humanig gamma-1
F
GO: 0005515
protein binding



chain c region os = homo




sapiens gn = ighg1 pe = 1 sv = 1



KH 45
IPI00219713
FibringluRAAS ®
same as




Human
KH 30




Thrombin


IPI00219713
fibg_humanfibrinogen
P
GO: 0009987
cellular process



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
C
GO: 0009897
external side of plasma



gamma chain os = homo


membrane




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
F
GO: 0043499
eukaryotic cell surface



gamma chain os = homo


binding




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
C
GO: 0005615
extracellular space



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
C
GO: 0031091
platelet alpha granule



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
P
GO: 0032501
multicellular organismal



gamma chain os = homo


process




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
P
GO: 0065007
biological regulation



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



IPI00219713
fibg_humanfibrinogen
P
GO: 0051592
response to calcium ion



gamma chain os = homo




sapiens gn = fgg pe = 1 sv = 3



KH 46
IPI00022371
FibringluRAAS ®
IPI00022371
hrg_humanhistidine-rich




Human

glycoprotein os = homo




Thrombin


sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0043065
positive regulation of



glycoprotein os = homo


apoptotic process




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0010468
regulation of gene



glycoprotein os = homo


expression




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0032956
regulation of actin



glycoprotein os = homo


cytoskeleton organization




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0016525
negative regulation of



glycoprotein os = homo


angiogenesis




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 2000504
positive regulation of blood



glycoprotein os = homo


vessel remodeling




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0043254
regulation of protein



glycoprotein os = homo


complex assembly




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0002839
positive regulation of



glycoprotein os = homo


immune response to tumor




sapiens gn = hrg pe = 1 sv = 1



cell


IPI00022371
hrg_humanhistidine-rich
F
GO: 0008201
heparin binding



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0010593
negative regulation of



glycoprotein os = homo


lamellipodium assembly




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0050832
defense response to fungus



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
F
GO: 0020037
heme binding



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
F
GO: 0019865
immunoglobulin binding



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0030168
platelet activation



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
F
GO: 0043395
heparan sulfate proteoglycan



glycoprotein os = homo


binding




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 1900747
negative regulation of



glycoprotein os = homo


vascular endothelial growth




sapiens gn = hrg pe = 1 sv = 1



factor signaling pathway


IPI00022371
hrg_humanhistidine-rich
F
GO: 0008270
zinc ion binding



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
F
GO: 0043498
cell surface binding



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 2001027
negative regulation of



glycoprotein os = homo


endothelial cell chemotaxis




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0008285
negative regulation of cell



glycoprotein os = homo


proliferation




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0051894
positive regulation of focal



glycoprotein os = homo


adhesion assembly




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0030193
regulation of blood



glycoprotein os = homo


coagulation




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0050730
regulation of peptidyl-



glycoprotein os = homo


tyrosine phosphorylation




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
P
GO: 0033629
negative regulation of cell



glycoprotein os = homo


adhesion mediated by




sapiens gn = hrg pe = 1 sv = 1



integrin


IPI00022371
hrg_humanhistidine-rich
P
GO: 0030308
negative regulation of cell



glycoprotein os = homo


growth




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
C
GO: 0005576
extracellular region



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



IPI00022371
hrg_humanhistidine-rich
C
GO: 0019814
immunoglobulin complex



glycoprotein os = homo




sapiens gn = hrg pe = 1 sv = 1



KH 47
IPI00022371
FibringluRAAS ®
same as




Human
KH 46




Thrombin


KH 48
IPI00022463
AFOD
IPI00022463
trfe_humanserotransferrin






os = homo sapiens gn = tf pe = 1






sv = 3


IPI00022463
trfe_humanserotransferrin
P
GO: 0009987
cellular process



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
P
GO: 0065008
regulation of biological



os = homo sapiens gn = tf pe = 1


quality



sv = 3


IPI00022463
trfe_humanserotransferrin
P
GO: 0006810
transport



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0009925
basal plasma membrane



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0005739
mitochondrion



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0030139
endocytic vesicle



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0005905
coated pit



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0005770
late endosome



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0005769
early endosome



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0055037
recycling endosome



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
F
GO: 0005515
protein binding



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0048471
perinuclear region of



os = homo sapiens gn = tf pe = 1


cytoplasm



sv = 3


IPI00022463
trfe_humanserotransferrin
C
GO: 0016324
apical plasma membrane



os = homo sapiens gn = tf pe = 1



sv = 3


IPI00022463
trfe_humanserotransferrin
P
GO: 0006950
response to stress



os = homo sapiens gn = tf pe = 1



sv = 3


KH 49
IPI00023006
AFOD
IPI00023006
alpha cardiac muscle 1






os = homo sapiens gn = actc1






pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
C
GO: 0005865
striated muscle thin filament



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
F
GO: 0017022
myosin binding



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0030240
skeletal muscle thin filament



os = homo sapiens gn = actc1


assembly



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0006200
ATP catabolic process



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0072144
glomerular mesangial cell



os = homo sapiens gn = actc1


development



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0006936
muscle contraction



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0033275
actin-myosin filament



os = homo sapiens gn = actc1


sliding



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
C
GO: 0042643
actomyosin, actin part



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0042221
response to chemical



os = homo sapiens gn = actc1


stimulus



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
F
GO: 0005524
ATP binding



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
C
GO: 0001725
stress fiber



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
F
GO: 0016887
ATPase activity



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0065008
regulation of biological



os = homo sapiens gn = actc1


quality



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
C
GO: 0044451
nucleoplasm part



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0009615
response to virus



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
P
GO: 0060047
heart contraction



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
F
GO: 0019899
enzyme binding



os = homo sapiens gn = actc1



pe = 1 sv = 1


IPI00023006
alpha cardiac muscle 1
C
GO: 0016459
myosin complex



os = homo sapiens gn = actc1



pe = 1 sv = 1


KH 50
IPI00021841
AFOD
same as





KH 37


KH 51
IPI00023006
AlbuRAAS
same as





KH 49


KH 52
IPI00930226
FibringluRAAS ®
IPI00930226
cytoplasmic 2 os = homo




High


sapiens gn = actg1 pe = 1 sv = 1





Concentrate




Human




Fibrinogen


IPI00930226
cytoplasmic 2 os = homo
P
GO: 0009888
tissue development




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0030048
actin filament-based




sapiens gn = actg1 pe = 1 sv = 1



movement


IPI00930226
cytoplasmic 2 os = homo
P
GO: 0003012
muscle system process




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
C
GO: 0030017
sarcomere




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0030239
myofibril assembly




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0044238
primary metabolic process




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
C
GO: 0005884
actin filament




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0072358
cardiovascular system




sapiens gn = actg1 pe = 1 sv = 1



development


IPI00930226
cytoplasmic 2 os = homo
P
GO: 0044237
cellular metabolic process




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0048513
organ development




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0042221
response to chemical




sapiens gn = actg1 pe = 1 sv = 1



stimulus


IPI00930226
cytoplasmic 2 os = homo
F
GO: 0008092
cytoskeletal protein binding




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0065008
regulation of biological




sapiens gn = actg1 pe = 1 sv = 1



quality


IPI00930226
cytoplasmic 2 os = homo
C
GO: 0044451
nucleoplasm part




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
P
GO: 0008015
blood circulation




sapiens gn = actg1 pe = 1 sv = 1



IPI00930226
cytoplasmic 2 os = homo
F
GO: 0019899
enzyme binding




sapiens gn = actg1 pe = 1 sv = 1



KH 53
194373497
AFCC
gi|194373497
thrb_humanprothrombin




(Fraction IV)

os = homo sapiens gn = f2






pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
C
GO: 0044446
intracellular organelle part



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 0048712
negative regulation of



os = homo sapiens gn = f2


astrocyte differentiation



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
C
GO: 0043233
organelle lumen



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 0030194
positive regulation of blood



os = homo sapiens gn = f2


coagulation



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
F
GO: 0005102
receptor binding



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 2000379
positive regulation of



os = homo sapiens gn = f2


reactive oxygen species



pe = 1 sv = 2


metabolic process


gi|194373497
thrb_humanprothrombin
P
GO: 0045861
negative regulation of



os = homo sapiens gn = f2


proteolysis



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
C
GO: 0005615
extracellular space



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 0030168
platelet activation



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 1900738
positive regulation of



os = homo sapiens gn = f2


phospholipase C-activating



pe = 1 sv = 2


G-protein coupled receptor






signaling pathway


gi|194373497
thrb_humanprothrombin
P
GO: 0016477
cell migration



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
C
GO: 0043231
intracellular membrane-



os = homo sapiens gn = f2


bounded organelle



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 0001934
positive regulation of protein



os = homo sapiens gn = f2


phosphorylation



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
C
GO: 0005886
plasma membrane



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
F
GO: 0070053
thrombospondin receptor



os = homo sapiens gn = f2


activity



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 0051281
positive regulation of release



os = homo sapiens gn = f2


of sequestered calcium ion



pe = 1 sv = 2


into cytosol


gi|194373497
thrb_humanprothrombin
F
GO: 0004252
serine-type endopeptidase



os = homo sapiens gn = f2


activity



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 0042730
fibrinolysis



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
C
GO: 0044444
cytoplasmic part



os = homo sapiens gn = f2



pe = 1 sv = 2


gi|194373497
thrb_humanprothrombin
P
GO: 0032967
positive regulation of



os = homo sapiens gn = f2


collagen biosynthetic



pe = 1 sv = 2


process


KH 54
194380034
Transferrin
gi|194380034
trfe_humanserotransferrin






os = homo sapiens gn = tf pe = 1






sv = 3


gi|194380034
trfe_humanserotransferrin
P
GO: 0009987
cellular process



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
P
GO: 0065008
regulation of biological



os = homo sapiens gn = tf pe = 1


quality



sv = 3


gi|194380034
trfe_humanserotransferrin
P
GO: 0006810
transport



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0009925
basal plasma membrane



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0005739
mitochondrion



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0030139
endocytic vesicle



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0005905
coated pit



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0005770
late endosome



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0005769
early endosome



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0055037
recycling endosome



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
F
GO: 0005515
protein binding



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0048471
perinuclear region of



os = homo sapiens gn = tf pe = 1


cytoplasm



sv = 3


gi|194380034
trfe_humanserotransferrin
C
GO: 0016324
apical plasma membrane



os = homo sapiens gn = tf pe = 1



sv = 3


gi|194380034
trfe_humanserotransferrin
P
GO: 0006950
response to stress



os = homo sapiens gn = tf pe = 1



sv = 3


KH 55
194380034
Transferrin
same as





KH 54









Protein sequence data as well as sequence identifiers and accession numbers for KH proteins 1-55 are found in the table below.














KH




Protein/SEQ
Sequence



ID NO
Identifier(s)
Protein Sequence


















1
gi: 21749960
MDTYIESHFA GALAYRDLIK VLKWYVDRIT EAERQEHIQE VLKAQEYIFK YIVQSRRLFS
60



BAC03696.1
LATGGQNEEE FRCCIQELLM SVRFFLSQES KGSGALSQSQ AVFLSSFPAV YSELLKLFDV
120




REVANLVQDT LGSLPTILHV DDSLQAIKLQ CIGKTVESQL YTNPDSRYIL LPVVLHHLHI
180




HLQEQKDLIM CARILSNVFC LIKKNSSEKS VLEEIDVIVA SLLDILLRTI LEITSRPQPS
240




SSAMRFQFQD VTGEFVACLL SLLRQMTDRH YQQLLDSFNT KEELRDFLLQ IFTVFRILIR
300




PEMFPKDWTV MRLVANNVII TTVLYLSDAL RKNFLNENFD YKIWDSYFYL AVIFINQLCL
360




QLEMFTPSKK KKVLEKYGDM RVTMGCEIFS MWQNLGEHKL HFIPALIGPF LEVTLIPQPD
420




LRNVMIPIFH DMMDWEQRRS GNFKQVEAKL IDKLDSLMSE GEGDETYREL FNSIIPLFGP
480




YPSLLKKIER ETWRESGVSL IATVTRLMER LLDYRDCMKM GEVDGKKIGC TVSLLNFYKT
540




ELNKEEMYIR YIHKLYDLHL KAQNFTEAAY TLLLYDELLE WSDRPLREFL TYPMQTEWQR
600




KEHLHLTIIQ NFDRGKCWEN GIILCRKIAE QYESYYDYRN LSKMRMMEAS LYDKIMDQQR
660




LEPEFFRVGF YGKKFPFFLR NKEFVCRGHD YERLEAFQQR MLNEFPHAIA MQHANQPDET
720




IFQAEAQYLQ IYAVTPIPES QEVLQREGVP DNIKSFYKVN HIWKFRYDRP FHKGT
775





2
gi: 215415640
DEPPQSPWDR VKDLATVYVD VLKDSGRDYV SQFEGSALGK QLNLKLLDNW DSVTSTFSKL
60



CAT02162.1
REQLGPVTQE FWDNLEKETE GLRQEMSKDL EEVKAKVQPY LDDFQKKWQE EMELYRQKVE
120




PLRAELQEGA RQKLHELQEK LSPLGEEMRD CARAHVDALR THLAPYSDEL RQRLAARLEA
180




LKENGGARLA EYHAKATEHL STLSEKAKPA LEDLRQGLLP VLESFKVSFL SALEEYTKKL
240




N
241





3
gi: 215415638
DEPPQSPWDR VKDLATVYVD VLKDSGRDYV SQFEGSALGK QLNLKLLDNW DSVTSTFSKL
60



CAT02161.1
REQLGPVTQE FWDNLEKETE GLCQEMSKDL EEVKAKVQPY LDDFQKKWQE EMELYRQKVE
120




PLRAELQEGA RQKLHELQEK LSPLGEEMRD RARAHVDALR THLAPYSDEL RQRLAARLEA
180




LKENGGARLA EYHAKATEHL STLSEKAKPA LEDLRQGLLP VLESFKVSFL SALEEYTKKL
240




NTQ
243





4
gi: 40044478
MGCKRASEVC GXAVEGLRDP LKPSEPSQGA AGKRKGTEYL MKQKLEFGGR GEELLLGVHL
60



CAF01015.1
RGAQKTGGGW RR
72





5
gi: 194383496
TATDVFWAKY TACETARTPR DKLAACLEGN CAEGLGTNYR GHVNITRSGI ECQLWRSRYP
120



BAG64719.1
HKPEINSTTH PGADLQENFC RNPDSSTTGP WCYTTDPTVR RQECSIPVCG QDQVTVAMTP
180




GQQYQGRLAV TTHGLPCLAW ASAQAKALSK HQDFNSAVQL
240




VENFCRNPDG DEEGVWCYVA GKPGDFGYCD LNYCEEAVEE ETGDGLDEDS DRAIEGRTAT
300




SEYQTFFNPR TFGSGEADCG LRPLFEKKSL EDKTERELLE SYIDGRIVEG SDAEIGMSPW
360




QVMLFRKSPQ ELLCGASLIS DRWVLTAAHC LLYPPWDKNF TENDLLVRIG KHSRTRYERN
420




IEKISMLEKI YIHPRYNWRE NLDRDIALMK LKKPVAFSDY IHPVCLPDRE TAASLLQAGY
480




KGRVTGWGNL KETWTANVGK GQPSVLQVVN LPIVERPVCK DSTRIRITDN MFCAGYKPDE
540




GKRGDACEGD SGGPFVMKSP FNNRWYQMGI VSWGEGCDRD GKYGFYTHVF RLKKWIQKVI
600




DQFGE
605





6
gi: 28071026
MQGTDEHVVC KVQHPNGNKE KNVPLPVIAE LPPKVSVFVP PRDGFFGNPR KSKLICQATG
60



CAD61894.1
FSPRQIQVSW LREGKQVGSG VTTDQVQAEA KESGPTTYKV TSTLTIKESD WLSQSMFTCR
120




VDHRGLTFQQ NASSMCGPDQ DTAIRVFAIP PSFASIFLTK STKLTCLVTD LTTYDSVTIS
180




WTRQNGEAVK THTNISESHP NATFSAVGEA SICEDDWNSG ERFTCTVTHT DLPSPLKQTI
240




SRPKGVALHR PDVYLLPPAR EQLNLRESAT ITCLVTGFSP ADVFVQWMQR GQPLSPEKYV
300




TSAPMPEPQA PGRYFAHSIL TVSEEEWNTG ETYTCVVAHE ALPNRVTERT VDKSTGKPTL
360




YNVSLVMSDT AGTCY
375





7
gi: 300621695
MEFGLSWLFL VAILKGVQCE VQLLESGGGL VQPGGSLRLS CAASGFTFSS YAMSWVRQAP
60



CBU30464.1
GKGLEWVSAI SGSGYTTYYA DSVKGRFTIS RDNSKNTLYL QMNSLRAEDT AVYYCAKKPG
120




DYGSGSYYLD YWGQGTLVTV SSGSASAPTL FPLVSCENSP SDTSSVAVGC LAQDFLPDSI
180




TFSWKYKNNS DISSTRGFPS VLRGGKYAAT SQVLLPSKDV MQGTDEHVVC KVQHPNGNKE
240




KNVPLPVIAE LPPKVSVFVP PRDGFFGNPR KSKLICQATG FSPRQIQVSW LREGKQVGSG
300




VTTDQVQAEA KESGPTTYKV TSTLTIKESD WLSQSMFTCR VDHRGLTFQQ NASSMCVPDQ
360




DTAIRVFAIP PSFASIFLTK STKLTCLVTD LTTYDSVTIS WTRQNGEAVK THTNISESHP
420




NATFSAVGEA SICEDDWNSG ERFTCTVTHT DLPSPLKQTI SRPKGVALHR PDVYLLPPAR
480




EQLNLRESAT ITCLVTGFSP ADVFVQWMQR GQPLSPEKYV TSAPMPEPQA PGRYFAHSIL
540




TVSEEEWNTG ETYTCVVAHE ALPNRVTERT VDKSTGKPTL YNVSLVMSDT AGTCY
595





8
gi: 1335098
TPLPPTSAHG NVAEGETKPD PDVTERCSDG WSFDATTLDD NGTMLFFKGE FVWKSHKWDR
60



CAA26382.1
ELISERWKNF PSPVDAAFRQ GHNSVFLIKG DKVWVYPPEK KEKGYPKLLQ DEFPGIPSPL
120




DAAVECHRGE CQAEGVLFFQ GDREWFWDLA TGTMKERSWP AVGNCSSALR WLGRYYCFQG
180




NQFLRFDPVR GEVPPRYPRD VRDYFMPCPG RGHGHRNGTG HGNSTHHGPE YMRCSPHLVL
240




SALTSDNHGA TYAFSGTHYW RLDTSRDGWH SWPIAHQWPQ GPSAVDAAFS WEEKLYLVQG
300




TQVYVFLTKG GYTLVSGYPK RLEKEVGTPH GIILDSVDAA FICPGSSRLH IMAGRRLWWL
360




DLKSGAQATW TELPWPHEKV DGALCMEKSL GPNSCSANGP GLYLIHGPNL YCYSDVEKLN
420




AAKALPQPQN VTSLLGCTH
439





9
gi: 10434804
MEPRAVGVSK QDIREQIWGY MESQNLADFP RPVHHRIPNF KGSYLACQNI KDLDVFARAQ
60



BAB14383.1
EVKVDPDKPL EGVRLLVLQS KKTLLVPTPR LRTGLFNKIT PPPGATKDIL RKCATSQGVR
120




NYSVPIGLDS RVLVDLVVVG SVAASEKGWR IGKGEGYADL EYAMMVSMGA VSKETPVVTI
180




VHDCQVVDIP EELVEEHDIT VDYILTPTRV IATGCKRPKP MGITWFKISL EMMEKIPILR
240




SLRAREQQAG KDVTLQGEHQ HLPEPGCQQT VPLSVGRRPP DTPGPETNSM EAAPGSPPGE
300




GAPLAADVYV GNLPRDARVS DLKRALRELG SVPLRLTWQG PRRRAFLHYP DSAAASRPSP
360




ACRACAWAPT P
371





10
gi: 221044726
MARVLGAPVA LGLWSLCWSL AIATPLPPTS AHGNVAEGET KPDPDVTERC SDGWSFDATT
60



BAH14040.1
LDDNGTMLFF KGEFVWKSHK WDRELISERL KNFPSPVDAA FRQGHNSVFL IKVLLGQNQG
120




QAGKGWNRHW GPFPQMALAW SP
142





11
gi: 215415638
Same as KH3




CAT02161.1







12
gi: 189066554
MAHVRGLQLP GCLALAALCS LVHSQHVFLA PQQARSLLQR VRRANTFLEE VRKGNLEREC
60



BAG35804.1
VEETCSYEEA FEALESSTAT DVFWAKYTAC ETARTPRDKL AACLEGNCAE GLGTNYRGHV
120




NITRSGIECQ LWRSRYPHKP EINSTTHPGA DLQENFCRNP DSSTMGPWCY TTDPTVRRQE
180




CSIPVCGQDQ VTVAMTPRSE GSSVNLSPPL EQCVPDRGQQ YQGRLAVTTH GLPCLAWASA
240




QAKALSKHQD FNSAVQLVEN FCRNPDGDEE GVWCYVAGKP GDFGYCDLNY CEEAVEEETG
300




DGLDEDSDRA IEGRTATSEY QTFFNPRTFG SGEADCGLRP LFEKKSLEDK TERELLESYI
360




DGRIVEGSDA EIGMSPWQVM LFRKSPQELL CGASLISDRW VLTAAHCLLY PPWDKNFTEN
420




DLLVRIGKHS RTRYERNIEK ISMLEKIYIH PRYNWRENLD RDIALMKLKK PVAFSDYIHP
480




VCLPDRETAA SLLQAGYKGR VTGWGNLKET WTANVGKGQP SVLQVVNLPI VERPVCKDST
540




RIRITDNMFC AGYKPDEGKR GDACEGDSGG PFVMKSPFNN RWYQMGIVSW GEGCDRDGKY
600




GFYTHVFRLK KWIQKVIDQF GE
622





13
gi: 194391084
MKLSLTQESQ SEEIDCNDKD LFKAVDAALK KYNSQNQSNN QFVLYRITEA TKTVGSDTFY
60



BAG60660.1
SFKYEIKEGD CPVQSGKTWQ DCEYKDAAKA ATGECTATVG KRSSTKFSVA TQTCQITPAE
120




GPVVTAQYDC LGCVHPISTQ SPDLEPILRH GIQYFNNNTQ HSSLFMLNEV KRAQRQVVAG
180




LNFRITYSIV QTNCSKENFL FLTPDCKSLW NGDTGECTDN AYIDIQLRIA SFSQNCDIYP
240




GKDFVQPPTK ICVGCPRDIP TNSPELEETL THTITKLNAE NNATFYFKID NVKKARVQVV
300




AGKKYFIDFV ARETTCSKES NEELTESCET KKLGQSLDCN AEVYVVPWEK KIYPTVNCQP
360




LGMISLMKRP PGFSPFRSSR IGEIKEETTS HLRSCEYKGR PPKAGAEPAS EREVS
415





14
gi: 158255114
MKLITILFLC SRLLLSLTQE SQSEEIDCND KDFFKAVDAA LKKYNSQNQS NNQFVLYRIT
60



BAF83528.1
EATKTVGSDT FYSFKYEIKE GDCPVQSGKT WQDCEYKDAA KAATGECTAT VGKRSSTKFS
120




VATQTCQITP AEGPVVTAQY DCLGCVHPIS TQSPDLEPIL RHGIQYFNNN TQHSSLFMLN
180




EVKRAQRQVV AGLNFRITYS IVQTNCSKEN FLFLTPDCKS LWNGDTGECT DNAYIDIQLR
240




IASFSQNCDI YPGKDFVQPP TKICVGCPRD IPTNSPELEE TLTHTITKLN AENNATFYFK
300




IDNVKKARVQ AVAGKKYFID FVARETTCSK ESNEELTESC ETKKLGQSLD CNAEVYVVPW
360




EKKIYPTVNC QPLGMISLMK RPPGFSPFRS SRIGEIKEET TSHLRSCEYK GRPPKAGAEP
420




ASEREVS
427





15
gi: 213506121
MKLITILFLC SRLLLSLTQE SQSEEIDCND KDLFKAVDAA LKKYNSQNQS NNQFVLYRIT
60



CAS91511.1
EATKTVGSDT FYSFKYEIKE GDCPVQSGKT WQDCEYKDAA KAATGECTAT VGKRSSTKFS
120




VATQTCQITP AEGPVVTAQY DCLGCVHPIS TQSPDLEPIL RHGIQYFNNN TQHSSLFMLN
180




EVKRAQRQVV AGLNFRMTYS IVQTNCSKEN FLFLTPDCKS LWNGDTGECT DNAYIDIQLR
240




IASFSQNCDI YPGKDFVQPP TKICVGCPRD IPTNSPELEE TLTHTITKLN AENNATFYFK
300




IDNVKKARVQ VVAGKKYFID FVARETTCSK ESNEELTESC ETKKLGQSLD CNAEVYVVPW
360




EKKIYPTVNC QPLGMISLMK RPPGFSPFRS SRIGEIKEET TSHLRSCEYK GRPPKAGAEP
420




ASEREVS
427





16
gi: 213506103
MKLITILFLC SRLLLSLTQE SQSEEIDCND KDLFKAVDAA LKKYNSQNQS NNQFVLYRIT
60



CAS91502.1
EATKTVGSDT FYSFKYEIKE GDCPVQSGKT WQDCEYKDAA KAATGECTAT VGKRSSTKFS
120




VATQTCQITP AEGPVVTAQY DCLGCVHPIS TQSPDLEPIL RHGIQYFNNN TQHSSLFMLN
180




EVKRAQRQVV AGLNFRMTYS IVQTNCSKEN FLFLTPDCKS LWNGDTGECT DNAYIDIQLR
240




IASFSQNCDI YPGKDFVQPP TKICVGCPRD IPTNSPELEE TLTHTITKLN AENNATFYFK
300




IDNVKKARVQ VVAGKKYFID FVARETTCSK ESNEELTESC ETKKLGQSLD CNAEVYVVPW
360




EKKIYPTVNC QPLGMISLMK RPPGFSPFRS SRIGEIKEET TSHLRSCEYK GRPPKAGAEP
420




ASEREVS
427





17
gi: 194376310
MDDDIAALVV DNGSGMCKAG FAGDDAPRAV FPSIVGRPRH QGVMVGIVTN WDDMEKIWHH
60



BAG62914.1
TFYNELRVAP EEHPVLLTEA PLNPKANREK MTQIMFETFN TPAMYVAIQA VLSLYASGRT
120




TGIVMDSGDG VTHTVPIYEG YALPHAILRL DLAGRDLTDY LMKILTERGY SFTTTAEREI
180




VRDIKEKLCY VALDFEQEMA TAASSSSLEK SYELPDGQVI TIGNERFRCP EALFQPSFLG
240




MESCGIHETT FNSIMKCDVD IRKDLYANTV LSGGTTMYPG IADRMQKEIT ALAPSTMKIK
300




IIAPPERKYS VWIGGSILAS LSTFQQMWIS KQEYDESGPS IVHRKCF
347





18
gi: 194388064
MEEEIAALVI DNGSGMCKAG FAGDDAPRAV FPSIVGRPRH QGVMVGMGQK DSYVGDEAQS
60



BAG65416.1
KRGILTLKYP IEHGIVTNWD DMEKIWHHTF YNELRVAPEE HPVLLTEAPL NPKANREKMT
120




QIMFETFNTT GIVMDSGDGV THTVPIYEGY ALPHAILRLD LAGRDLTDYL MKILTERGYS
180




FTTTAEREIV RDIKEKLCYV ALDFEQEMAT AASSSSLEKS YELPDGQVIT IGNERFRCPE
240




ALFQPSFLGM ESCGIHETTF NSIMKCDVDI RKDLYANTVL SGGTTMYPGI ADRMQKEITA
300




LAPSTMKIKI IAPPERKYSV WIGGSILASL STFQQMWISK QEYDESGPSI VHRKCF
356





19
IPI00964149
MQKSEGSGGT QLKNRATGNY DQRTSSSTQL KHRNAVQGSK SSLSTSSPES ARKLHPRPSD
60



gi: 126215685
KLNPKTINPF GEQSRVPSAF AAIYSKGGIP CRLVHGSVKH RLQWECPPES LSFDPLLITL
120



Q8N7B6.2
AEGLRETKHP YTFVSKEGFR ELLLVKGAPE KAIPLLPRLI PVLKAALVHS DDEVFERGLN
180




ALVQLSVVVG PSLNDHLKHL LTSLSKRLMD KKFKEPITSA LQKLEQHGGS GSLSIIKSKI
240




PTYCSICC
248





20
IPI00966721
MASETEKTHA LLQTCSTESL ISSLGLGAFC LVADRLLQFS TIQQNDWLRA LSDNAVHCVI
60



gi: 121940485
GMWSWAVVTG IKKKTDFGEI ILAGFLASVI DVDHFFLAGS MSLKAALTLP RRPFLHCSTV
120



Q0VDI3.1
IPVVVLTLKF TMHLFKLKDS WCFLPWMLFI SWTSHHIRDG IRHGLWICPF GKTSPLPFWL
180




YVIITSSLPH ICSFVMYLTG TRQMMSSKHG VRIDV
215





21
IPI00966826
MDFTAQPKPA TALCGVVSAD GKIAYPPGVK EITDKITTDE MIKRLKMVVK TFMDMDQDSE
60



gi: 121947590
DEKQQYLPLA LHLASEFFLR NPNKDVRLLV ACCLADIFRI YAPEAPYTSH DKLKDIFLFI
120



Q29RF7.1
TRQLKGLEDT KSPQFNRYFY LLENLAWVKS YNICFELEDC NEIFIQLFRT LFSVINNSHN
180




KKVQMHMLDL MSSIIMEGDG VTQELLDSIL INLIPAHKNL NKQSFDLAKV LLKRTVQTIE
240




ACIANFFNQV LVLGRSSVSD LSEHVFDLIQ ELFAIDPHLL LSVMPQLEFK LKSNDGEERL
300




AVVRLLAKLF GSKDSDLATQ NRPLWQCFLG RFNDIHVPVR LESVKFASHC LMNHPDLAKD
360




LTEYLKVRSH DPEEAIRHDV IVTIITAAKR DLALVNDQLL GFVRERTLDK RWRVRKEAMM
420




GLAQLYKKYC LHGEAGKEAA EKVSWIKDKL LHIYYQNSID DKLLVEKIFA QYLVPHNLET
480




EERMKCLYYL YASLDPNAVK ALNEMWKCQN MLRSHVRELL DLHKQPTSEA NCSAMFGKLM
540




TIAKNLPDPG KAQDFVKKFN QVLGDDEKLR SQLELLISPT CSCKQADICV REIARKLANP
600




KQPTNPFLEM VKFLLERIAP VHIDSEAISA LVKLMNKSIE GTADDEEEGV SPDTAIRSGL
660




ELLKVLSFTH PTSFHSAETY ESLLQCLRME DDKVAEAAIQ IFRNTGHKIE TDLPQIRSTL
720




IPILHQKAKR GTPHQAKQAV HCIHAIFTNK EVQLAQIFEP LSRSLNADVP EQLITPLVSL
780




GHISMLAPDQ FASPMKSVVA NFIVKDLLMN DRSTGEKNGK LWSPDEEVSP EVLAKVQAIK
840




LLVRWLLGMK NNQSKSANST LRLLSAMLVS EGDLTEQKRI SKSDMSRLRL AAGSAIMKLA
900




QEPCYHEIIT PEQFQLCALV INDECYQVRQ IFAQKLHKAL VKLLLPLEYM AIFALCAKDP
960




VKERRAHARQ CLLKNISIRR EYIKQNPMAT EKLLSLLPEY VVPYMIHLLA HDPDFTRSQD
1020




VDQLRDIKEC LWFMLEVLMT KNENNSHAFM KKMAENIKLT RDAQSPDESK TNEKLYTVCD
1080




VALCVINSKS ALCNADSPKD PVLPMKFFTQ PEKDFCNDKS YISEETRVLL LTGKPKPAGV
1140




LGAVNKPLSA TGRKPYVRST GTETGSNINV NSELNPSTGN RSREQSSEAA ETGVSENEEN
1200




PVRIISVTPV KNIDPVKNKE INSDQATQGN ISSDRGKKRT VTAAGAENIQ QKTDEKVDES
1260




GPPAPSKPRR GRRPKSESQG NATKNDDLNK PINKGRKRAA VGQESPGGLE AGNAKAPKLQ
1320




DLAKKAAPAE RQIDLQR
1337





22
IPI00760788
MAEEQEFTQL CKLPAQPSHP HCVNNTYRSA QHSQALLRGL LALRDSGILF DVVLVVEGRH
60



gi: 109892504
IEAHRILLAA SCDYFRGMFA GGLKEMEQEE VLIHGVSYNA MCQILHFIYT SELELSLSNV
120



Q53GT1.2
QETLVAACQL QIPEIIHFCC DFLMSWVDEE NILDVYRLAE LFDLSRLTEQ LDTYILKNFV
180




AFSRTDKYRQ LPLEKVYSLL SSNRLEVSCE TEVYEGALLY HYSLEQVQAD QISLHEPPKL
240




LETVRFPLME AEVLQRLHDK LDPSPLRDTV ASALMYHRNE SLQPSLQSPQ TELRSDFQCV
300




VGFGGIHSTP STVLSDQAKY LNPLLGEWKH FTASLAPRMS NQGIAVLNNF VYLIGGDNNV
360




QGFRAESRCW RYDPRHNRWF QIQSLQQEHA DLSVCVVGRY IYAVAGRDYH NDLNAVERYD
420




PATNSWAYVA PLKREVYAHA GATLEGKMYI TCGRRGEDYL KETHCYDPGS NTWHTLADGP
480




VRRAWHGMAT LLNKLYVIGG SNNDAGYRRD VHQVACYSCT SGQWSSVCPL PAGHGEPGIA
540




VLDNRIYVLG GRSHNRGSRT GYVHIYDVEK DCWEEGPQLD NSISGLAACV LTLPRSLLLE
600




PPRGTPDRSQ ADPDFASEVM SVSDWEEFDN SSED
634





23
IPI00917278
MKQLQPQPPP KMGDFYDPEH PTPEEEENEA KIENVQKTGF IKGPMFKGVA SSRFLPKGTK
60



gi:
TKVNLEEQGR QKVSFSFSLT KKTLQNRFLT ALGNEKQSDT PNPPAVPLQV DSTPKMKMEI
120




GDTLSTAEES SPPKSRVELG KIHFKKHLLH VTSRPLLATT TAVASPPTHA APLPAVIAES
180




TTVDSPPSSP PPPPPPAQAT TLSSPAPVTE PVALPHTPIT VLMAAPVPLP VDVAVRSLKE
240




PPIIIVPESL EADTKQDTIS NSLEEHVTQI LNEQADISSK KEDSHIGKDE EIPDSSKISL
300




SCKKTGSKKK SSQSEGIFLG SESDEDSVRT SSSQRSHDLK FSASIEKERD FKKSSAPLKS
360




EDLGKPSRSK TDRDDKYFSY SKLERDTRYV SSRCRSERER RRSRSHSRSE RGSRTNLSYS
420




RSERSHYYDS DRRYHRSSPY RERTRYSRPY TDNRARESSD SEEEYKKTYS RRTSSHSSSY
480




RDLRTSSYSK SDRDCKTETS YLEMERRGKY SSKLERESKR TSENEAIKRC CSPPNELGFR
540




RGSSYSKHDS SASRYKSTLS KPIPKSDKFK NSFCCTELNE EIKQSHSFSL QTPCSKGSEL
600




RMINKNPERE KAGSPAPSNR LNDSPTLKKL DELPIFKSEF ITHDSHDSIK ELDSLSKVKN
660




DQLRSFCPIE LNINGSPGAE SDLATFCTSK TDAVLMTSDD SVTGSELSPL VKACMLSSNG
720




FQNISRCKEK DLDDTCMLHK KSESPFRETE PLVSPHQDKL MSMPVMTVDY SKTVVKEPVD
780




TRVSCCKTKD SDIYCTLNDS NPSLCNSEAE NIEPSVMKIS SNSFMNVHLE SKPVICDSRN
840




LTDHSKFACE EYKQSIGSTS SASVNHFDDL YQPIGSSGIA SSLQSLPPGI KVDSLTLLKC
900




GENTSPVLDA VLKSKKSSEF LKHAGKETIV EVGSDLPDSG KGFASRENRR NNGLSGKCLQ
960




EAQEEGNSIL PERRGRPEIS LDERGEGGHV HTSDDSEVVF SSCDLNLTME DSDGVTYALK
1020




CDSSGHAPEI VSTVHEDYSG SSESSNDESD SEDTDSDDSS IPRNRLQSVV VVPKNSTLPM
1080




EETSPCSSRS SQSYRHYSDH WEDERLESRR HLYEEKFESI ASKACPQTDK FFLHKGTEKN
1140




PEISFTQSSR KQIDNRLPEL SHPQSDGVDS TSHTDVKSDP LGHPNSEETV KAKIPSRQQE
1200




ELPIYSSDFE DVPNKSWQQT TFQNRPDSRL GKTELSFSSS CEIPHVDGLH SSEELRNLGW
1260




DFSQEKPSTT YQQPDSSYGA CGGHKYQQNA EQYGGTRDYW QGNGYWDPRS GRPPGTGVVY
1320




DRTQGQVPDS LTDDREEEEN WDQQDGSHFS DQSDKFLLSL QKDKGSVQAP EISSNSIKDT
1380




LAVNEKKDFS KNLEKNDIKD RGPLKKRRQE IESDSESDGE LQDRKKVRVE VEQGETSVPP
1440




GSALVGPSCV MDDFRDPQRW KECAKQGKMP CYFDLIEENV YLTERKKNKS HRDIKRMQCE
1500




CTPLSKDERA QGEIACGEDC LNRLLMIECS SRCPNGDYCS NRRFQRKQHA DVEVILTEKK
1560




GWGLRAAKDL PSNTFVLEYC GEVLDHKEFK ARVKEYARNK NIHYYFMALK NDEIIDATQK
1620




GNCSRFMNHS CEPNCETQKW TVNGQLRVGF FTTKLVPSGS ELTFDYQFQR YGKEAQKCFC
1680




GSANCRGYLG GENRVSIRAA GGKMKKERSR KKDSVDGELE ALMENGEGLS DKNQVLSLSR
1740




LMVRIETLEQ KLTCLELIQN THSQSCLKSF LERHGLSLLW IWMAELGDGR ESNQKLQEEI
1800




IKTLEHLPIP TKNMLEESKV LPIIQRWSQT KTAVPPLSEG DGYSSENTSR AHTPLNTPDP
1860




STKLSTEADT DTPKKLMFRR LKIISENSMD SAISDATSEL EGKDGKEDLD QLENVPVEEE
1920




EELQSQQLLP QQLPECKVDS ETNIEASKLP TSEPEADAEI EPKESNGTKL EEPINEETPS
1980




QDEEEGVSDV ESERSQEQPD KTVDISDLAT KLLDSWKDLK EVYRIPKKSQ TEKENTTTER
2040




GRDAVGFRDQ TPAPKTPNRS RERDPDKQTQ NKEKRKRRSS LSPPSSAYER GTKRPDDRYD
2100




TPTSKKKVRI KDRNKLSTEE RRKLFEQEVA QREAQKQQQQ MQNLGMTSPL PYDSLGYNAP
2160




HHPFAGYPPG YPMQAYVDPS NPNAGKVLLP TPSMDPVCSP APYDHAQPLV GHSTEPLSAP
2220




PPVPVVPHVA APVEVSSSQY VAQSDGVVHQ DSSVAVLPVP APGPVQGQ
2268





24
IPI00966721
Same as KH 20




gi: 121940485





Q0VDI3.1







25
IPI01012037
MNGEYRGRGF GRGRFQSWKR GRGGGNFSGK WREREHRPDL SKTTGKRTSE QTPQFLLSTK
60



gi: 74735024
TPQSMQSTLD RFIPYKGWKL YFSEVYSDSS PLIEKIQAFE KFFTRHIDLY DKDEIERKGS
120



Q9UHY7.1
ILVDFKELTE GGEVTNLIPD IATELRDAPE KTLACMGLAI HQVLTKDLER HAAELQAQEG
180




LSNDGETMVN VPHIHARVYN YEPLTQLKNV RANYYGKYIA LRGTVVRVSN IKPLCTKMAF
240




LCAACGEIQS FPLPDGKYSL PTKCPVPVCR GRSFTALRSS PLTVTMDWQS IKIQELMSDD
300




QREAGRIPRT IECELVHDLV DSCVPGDTVT ITGIVKVSNA EEGSRNKNDK CMFLLYIEAN
360




SISNSKGQKT KSSEDGCKHG MLMEFSLKDL YAIQEIQAEE NLFKLIVNSL CPVIFGHELV
420




KAGLALALFG GSQKYADDKN RIPIRGDPHI LVVGDPGLGK SQMLQAACNV APRGVYVCGN
480




TTTTSGLTVT LSKDSSSGDF ALEAGALVLG DQGICGIDEF DKMGNQHQAL LEAMEQQSIS
540




LAKAGVVCSL PARTSIIAAA NPVGGHYNKA KTVSENLKMG SALLSRFDLV FILLDTPNEH
600




HDHLLSEHVI AIRAGKQRTI SSATVARMNS QDSNTSVLEV VSEKPLSERL KVVPGETIDP
660




IPHQLLRKYI GYARQYVYPR LSTEAARVLQ DFYLELRKQS QRLNSSPITT RQLESLIRLT
720




EARARLELRE EATKEDAEDI VEIMKYSMLG TYSDEFGNLD FERSQHGSGM SNRSTAKRFI
780




SALNNVAERT YNNIFQFHQL RQIAKELNIQ VADFENFIGS LNDQGYLLKK GPKVYQLQTM
840





26
IPI00940730
MVVLSVPAEV TVILLDIEGT TTPIAFVKDI LFPYIEENVK EYLQTHWEEE ECQQDVSLLR
60



gi:
KQAEEDAHLD GAVPIPAASG NGVDDLQQMI QAVVDNVCWQ MSLDRKTTAL KQLQGHMWRA
120




AFTAGRMKAE FFADVVPAVR KWREAGMKVY IYSSGSVEAQ KLLFGHSTEG DILELVDGHF
180




DTKIGHKVES ESYRKIADSI GCSTNNILFL TDVTREASAA EEADVHVAVV VRPGNAGLTD
240




DEKTYYSLIT SFSELYLPSS T
261





27
IPI00977191
MAMESTATAA VAAELVSADK IEDVPAPSTS ADKVESLDVD SEAKKLLGLG QKHLVMGDIP
60



gi: 23503077
AAVNAFQEAA SLLGKKYGET ANECGEAFFF YGKSLLELAR MENGVLGNAL EGVHVEEEEG
120



P49321.2
EKTEDESLVE NNDNIDEEAR EELREQVYDA MGEKEEAKKT EDKSLAKPET DKEQDSEMEK
180




GGREDMDISK SAEEPQEKVD LTLDWLTETS EEAKGGAAPE GPNEAEVTSG KPEQEVPDAE
240




EEKSVSGTDV QEECREKGGQ EKQGEVIVSI EEKPKEVSEE QPVVTLEKQG TAVEVEAESL
300




DPTVKPVDVG GDEPEEKVVT SENEAGKAVL EQLVGQEVPP AEESPEVTTE AAEASAVEAG
360




SEVSEKPGQE APVLPKDGAV NGPSVVGDQT PIEPQTSIER LTETKDGSGL EEKVRAKLVP
420




SQEETKLSVE ESEAAGDGVD TKVAQGATEK SPEDKVQIAA NEETQEREEQ MKEGEETEGS
480




EEDDKENDKT EEMPNDSVLE NKSLQENEEE EIGNLELAWD MLDLAKIIFK RQETKEAQLY
540




AAQAHLKLGE VSVESENYVQ AVEEFQSCLN LQEQYLEAHD RLLAETHYQL GLAYGYNSQY
600




DEAVAQFSKS IEVIENRMAV LNEQVKEAEG SSAEYKKEIE ELKELLPEIR EKIEDAKESQ
660




RSGNVAELAL KATLVESSTS GFTPGGGGSS VSMIASRKPT DGASSSNCVT DISHLVRKKR
720




KPEEESPRKD DAKKAKQEPE VNGGSGDAVP SGNEVSENME EEAENQAESR AAVEGTVEAG
780




ATVESTAC
788





28
IPI00022434
MKWVTFISLL FLFSSAYSRG VFRRDAHKSE VAHRFKDLGE ENFKALVLIA FAQYLQQCPF
60



gi: 113576
EDHVKLVNEV TEFAKTCVAD ESAENCDKSL HTLFGDKLCT VATLRETYGE MADCCAKQEP
120



P02768.2
ERNECFLQHK DDNPNLPRLV RPEVDVMCTA FHDNEETFLK KYLYEIARRH PYFYAPELLF
180




FAKRYKAAFT ECCQAADKAA CLLPKLDELR DEGKASSAKQ RLKCASLQKF GERAFKAWAV
240




ARLSQRFPKA EFAEVSKLVT DLTKVHTECC HGDLLECADD RADLAKYQKF NQDSISSKLK
300




ECCEKPLLEK SHCIAEVEND EMPADLPSLA ADFVESKDVC KNYAEAKDVF LGMFLYEYAR
360




RHPDYSVVLL LRLAKTYETT LEKCCAAADP HECYAKVFDE FKPLVEEPQN LIKQNCELFE
420




QLGEYKFQNA LLVRYTKKVP QVSTPTLVEV SRNLGKVGSK CCKHPEAKRM PCAEDYLSVV
480




LNQLCVLHEK TPVSDRVTKC CTESLVNRRP CFSALEVDET YVPKEFNAET FTFHADICTL
540




SEKERQIKKQ TALVELVKHK PKATKEQLKA VMDDFAAFVE KCCKADDKET CFAEEGKKLV
600




AASQAALGL
609





29
IPI00022434
Same as KH 28




gi: 113576





P02768.2







30
IPI00219713
MSWSLHPRNL ILYFYALLFL SSTCVAYVAT RDNCCILDER FGSYCPTTCG IADFLSTYQT
60



gi: 20178280
KVDKDLQSLE DILHQVENKT SEVKQLIKAI QLTYNPDESS KPNMIDAATL KSRKMLEEIM
120



P02679
KYEASILTHD SSIRYLQEIY NSNNQKIVNL KEKVAQLEAQ CQEPCKDTVQ IHDITGKDCQ
180




DIANKGAKQS GLYFIKPLKA NQQFLVYCEI DGSGNGWTVF QKRLDGSVDF KKNWIQYKEG
240




FGHLSPTGTT EFWLGNEKIH LISTQSAIPY ALRVELEDWN GRTSTADYAM FKVGPEADKY
300




RLTYAYFAGG DAGDAFDGFD FGDDPSDKFF TSHNGMQFST WDNDNDKFEG NCAEQDGSGW
360




WMNKCHAGHL NGVYYQGGTY SKASTPNGYD NGIIWATWKT RWYSMKKTTM KIIPFNRLTI
420




GEGQQHHLGG AKQVRPEHPA ETEYDSLYPE DDL
453





31
IPI00219713
Same as KH 30




gi: 20178280





P02679







32
IPI00220327
MSRQFSSRSG YRSGGGFSSG SAGIINYQRR TTSSSTRRSG GGGGRFSSCG GGGGSFGAGG
60



gi: 238054406
GFGSRSLVNL GGSKSISISV ARGGGRGSGF GGGYGGGGFG GGGFGGGGFG GGGIGGGGFG
120



P04264.6
GFGSGGGGFG GGGFGGGGYG GGYGPVCPPG GIQEVTINQS LLQPLNVEID PEIQKVKSRE
180




REQIKSLNNQ FASFIDKVRF LEQQNQVLQT KWELLQQVDT STRTHNLEPY FESFINNLRR
240




RVDQLKSDQS RLDSELKNMQ DMVEDYRNKY EDEINKRTNA ENEFVTIKKD VDGAYMTKVD
300




LQAKLDNLQQ EIDFLTALYQ AELSQMQTQI SETNVILSMD NNRSLDLDSI IAEVKAQYED
360




IAQKSKAEAE SLYQSKYEEL QITAGRHGDS VRNSKIEISE LNRVIQRLRS EIDNVKKQIS
420




NLQQSISDAE QRGENALKDA KNKLNDLEDA LQQAKEDLAR LLRDYQELMN TKLALDLEIA
480




TYRTLLEGEE SRMSGECAPN VSVSVSTSHT TISGGGSRGG GGGGYGSGGS SYGSGGGSYG
540




SGGGGGGGRG SYGSGGSSYG SGGGSYGSGG GGGGHGSYGS GSSSGGYRGG SGGGGGGSSG
600




GRGSGGGSSG GSIGGRGSSS GGVKSSGGSS SVKFVSTTYS GVTR
644





33
IPI00029739
MRLLAKIICL MLWAICVAED CNELPPRRNT EILTGSWSDQ TYPEGTQAIY KCRPGYRSLG
60



gi: 158517847
NVIMVCRKGE WVALNPLRKC QKRPCGHPGD TPFGTFTLTG GNVFEYGVKA VYTCNEGYQL
120



P08603.4
LGEINYRECD TDGWTNDIPI CEVVKCLPVT APENGKIVSS AMEPDREYHF GQAVRFVCNS
180




GYKIEGDEEM HCSDDGFWSK EKPKCVEISC KSPDVINGSP ISQKIIYKEN ERFQYKCNMG
240




YEYSERGDAV CTESGWRPLP SCEEKSCDNP YIPNGDYSPL RIKHRTGDEI TYQCRNGFYP
300




ATRGNTAKCT STGWIPAPRC TLKPCDYPDI KHGGLYHENM RRPYFPVAVG KYYSYYCDEH
360




FETPSGSYWD HIHCTQDGWS PAVPCLRKCY FPYLENGYNQ NYGRKFVQGK SIDVACHPGY
420




ALPKAQTTVT CMENGWSPTP RCIRVKTCSK SSIDIENGFI SESQYTYALK EKAKYQCKLG
480




YVTADGETSG SITCGKDGWS AQPTCIKSCD IPVFMNARTK NDFTWFKLND TLDYECHDGY
540




ESNTGSTTGS IVCGYNGWSD LPICYERECE LPKIDVHLVP DRKKDQYKVG EVLKFSCKPG
600




FTIVGPNSVQ CYHFGLSPDL PICKEQVQSC GPPPELLNGN VKEKTKEEYG HSEVVEYYCN
660




PRFLMKGPNK IQCVDGEWTT LPVCIVEEST CGDIPELEHG WAQLSSPPYY YGDSVEFNCS
720




ESFTMIGHRS ITCIHGVWTQ LPQCVAIDKL KKCKSSNLII LEEHLKNKKE FDHNSNIRYR
780




CRGKEGWIHT VCINGRWDPE VNCSMAQIQL CPPPPQIPNS HNMTTTLNYR DGEKVSVLCQ
840




ENYLIQEGEE ITCKDGRWQS IPLCVEKIPC SQPPQIEHGT INSSRSSQES YAHGTKLSYT
900




CEGGFRISEE NETTCYMGKW SSPPQCEGLP CKSPPEISHG VVAHMSDSYQ YGEEVTYKCF
960




EGFGIDGPAI AKCLGEKWSH PPSCIKTDCL SLPSFENAIP MGEKKDVYKA GEQVTYTCAT
1020




YYKMDGASNV TCINSRWTGR PTCRDTSCVN PPTVQNAYIV SRQMSKYPSG ERVRYQCRSP
1080




YEMFGDEEVM CLNGNWTEPP QCKDSTGKCG PPPPIDNGDI TSFPLSVYAP ASSVEYQCQN
1140




LYQLEGNKRI TCRNGQWSEP PKCLHPCVIS REIMENYNIA LRWTAKQKLY SRTGESVEFV
1200




CKRGYRLSSR SHTLRTTCWD GKLEYPTCAK R
1231





34
IPI00384853
QAHGRCSAGAQFVFCRRSAGAACTQQALSR (Sequence 59-88)




gi:
CLVGAQCVLSR (Sequence 100-110)





CTVCTQQALSR (Sequence 125-135)






35
IPI00479708
GSASAPTLFP LVSCENSPSD TSSVAVGCLA QDFLPDSITL SWKYKNNSDI SSTRGFPSVL
60



gi: 193806374
RGGKYAATSQ VLLPSKDVMQ GTDEHVVCKV QHPNGNKEKN VPLPVIAELP PKVSVFVPPR
120



P01871.3
DGFFGNPRKS KLICQATGFS PRQIQVSWLR EGKQVGSGVT TDQVQAEAKE SGPTTYKVTS
180




TLTIKESDWL GQSMFTCRVD HRGLTFQQNA SSMCVPDQDT AIRVFAIPPS FASIFLTKST
240




KLTCLVTDLT TYDSVTISWT RQNGEAVKTH TNISESHPNA TFSAVGEASI CEDDWNSGER
300




FTCTVTHTDL PSPLKQTISR PKGVALHRPD VYLLPPAREQ LNLRESATIT CLVTGFSPAD
360




VFVQWMQRGQ PLSPEKYVTS APMPEPQAPG RYFAHSILTV SEEEWNTGET YTCVAHEALP
420




NRVTERTVDK STGKPTLYNV SLVMSDTAGT CY
452





36
IPI00298497
MKRMVSWSFH KLKTMKHLLL LLLCVFLVKS QGVNDNEEGF FSARGHRPLD KKREEAPSLR
60



gi: 399492
PAPPPISGGG YRARPAKAAA TQKKVERKAP DAGGCLHADP DLGVLCPTGC QLQEALLQQE
120



P02675.2
RPIRNSVDEL NNNVEAVSQT SSSSFQYMYL LKDLWQKRQK QVKDNENVVN EYSSELEKHQ
180




LYIDETVNSN IPTNLRVLRS ILENLRSKIQ KLESDVSAQM EYCRTPCTVS CNIPVVSGKE
240




CEEIIRKGGE TSEMYLIQPD SSVKPYRVYC DMNTENGGWT VIQNRQDGSV DFGRKWDPYK
300




QGFGNVATNT DGKNYCGLPG EYWLGNDKIS QLTRMGPTEL LIEMEDWKGD KVKAHYGGFT
360




VQNEANKYQI SVNKYRGTAG NALMDGASQL MGENRTMTIH NGMFFSTYDR DNDGWLTSDP
420




RKQCSKEDGG GWWYNRCHAA NPNGRYYWGG QYTWDMAKHG TDDGVVWMNW KGSWYSMRKM
480




SMKIRPFFPQ Q
491





37
IPI00021841
MKAAVLTLAV LFLTGSQARH FWQQDEPPQS PWDRVKDLAT VYVDVLKDSG RDYVSQFEGS
60



gi: 113992
ALGKQLNLKL LDNWDSVTST FSKLREQLGP VTQEFWDNLE KETEGLRQEM SKDLEEVKAK
120



P02647.1
VQPYLDDFQK KWQEEMELYR QKVEPLRAEL QEGARQKLHE LQEKLSPLGE EMRDRARAHV
180




DALRTHLAPY SDELRQRLAA RLEALKENGG ARLAEYHAKA TEHLSTLSEK AKPALEDLRQ
240




GLLPVLESFK VSFLSALEEY TKKLNTQ
267





38
IPI00783987
MGPTSGPSLL LLLLTHLPLA LGSPMYSIIT PNILRLESEE TMVLEAHDAQ GDVPVTVTVH
60



gi: 119370332
DFPGKKLVLS SEKTVLTPAT NHMGNVTFTI PANREFKSEK GRNKFVTVQA TFGTQVVEKV
120



P01024.2
VLVSLQSGYL FIQTDKTIYT PGSTVLYRIF TVNHKLLPVG RTVMVNIENP EGIPVKQDSL
180




SSQNQLGVLP LSWDIPELVN MGQWKIRAYY ENSPQQVFST EFEVKEYVLP SFEVIVEPTE
240




KFYYIYNEKG LEVTITARFL YGKKVEGTAF VIFGIQDGEQ RISLPESLKR IPIEDGSGEV
300




VLSRKVLLDG VQNPRAEDLV GKSLYVSATV ILHSGSDMVQ AERSGIPIVT SPYQIHFTKT
360




PKYFKPGMPF DLMVFVTNPD GSPAYRVPVA VQGEDTVQSL TQGDGVAKLS INTHPSQKPL
420




SITVRTKKQE LSEAEQATRT MQALPYSTVG NSNNYLHLSV LRTELRPGET LNVNFLLRMD
480




RAHEAKIRYY TYLIMNKGRL LKAGRQVREP GQDLVVLPLS ITTDFIPSFR LVAYYTLIGA
540




SGQREVVADS VWVDVKDSCV GSLVVKSGQS EDRQPVPGQQ MTLKIEGDHG ARVVLVAVDK
600




GVFVLNKKNK LTQSKIWDVV EKADIGCTPG SGKDYAGVFS DAGLTFTSSS GQQTAQRAEL
660




QCPQPAARRR RSVQLTEKRM DKVGKYPKEL RKCCEDGMRE NPMRFSCQRR TRFISLGEAC
720




KKVFLDCCNY ITELRRQHAR ASHLGLARSN LDEDIIAEEN IVSRSEFPES WLWNVEDLKE
780




PPKNGISTKL MNIFLKDSIT TWEILAVSMS DKKGICVADP FEVTVMQDFF IDLRLPYSVV
840




RNEQVEIRAV LYNYRQNQEL KVRVELLHNP AFCSLATTKR RHQQTVTIPP KSSLSVPYVI
900




VPLKTGLQEV EVKAAVYHHF ISDGVRKSLK VVPEGIRMNK TVAVRTLDPE RLGREGVQKE
960




DIPPADLSDQ VPDTESETRI LLQGTPVAQM TEDAVDAERL KHLIVTPSGC GEQNMIGMTP
1020




TVIAVHYLDE TEQWEKFGLE KRQGALELIK KGYTQQLAFR QPSSAFAAFV KRAPSTWLTA
1080




YVVKVFSLAV NLIAIDSQVL CGAVKWLILE KQKPDGVFQE DAPVIHQEMI GGLRNNNEKD
1140




MALTAFVLIS LQEAKDICEE QVNSLPGSIT KAGDFLEANY MNLQRSYTVA IAGYALAQMG
1200




RLKGPLLNKF LTTAKDKNRW EDPGKQLYNV EATSYALLAL LQLKDFDFVP PVVRWLNEQR
1260




YYGGGYGSTQ ATFMVFQALA QYQKDAPDHQ ELNLDVSLQL PSRSSKITHR IHWESASLLR
1320




SEETKENEGF TVTAEGKGQG TLSVVTMYHA KAKDQLTCNK FDLKVTIKPA PETEKRPQDA
1380




KNTMILEICT RYRGDQDATM SILDISMMTG FAPDTDDLKQ LANGVDRYIS KYELDKAFSD
1440




RNTLIIYLDK VSHSEDDCLA FKVHQYFNVE LIQPGAVKVY AYYNLEESCT RFYHPEKEDG
1500




KLNKLCRDEL CRCAEENCFI QKSDDKVTLE ERLDKACEPG VDYVYKTRLV KVQLSNDFDE
1560




YIMAIEQTIK SGSDEVQVGQ QRTFISPIKC REALKLEEKK HYLMWGLSSD FWGEKPNLSY
1620




IIGKDTWVEH WPEEDECQDE ENQKQCQDLG AFTESMVVFG CPN
1663





39
IPI00878282
MKWVTFISLL FLFSSAYSRG VFRRDAHKSE VAHRFKDLGE ENFKALVLIA FAQYLQQCPF
60



gi: 113576
EDHVKLVNEV TEFAKTCVAD ESAENCDKSL HTLFGDKLCT VATLRETYGE MADCCAKQEP
120



P02768.2
ERNECFLQHK DDNPNLPRLV RPEVDVMCTA FHDNEETFLK KYLYEIARRH PYFYAPELLF
180




FAKRYKAAFT ECCQAADKAA CLLPKLDELR DEGKASSAKQ RLKCASLQKF GERAFKAWAV
240




ARLSQRFPKA EFAEVSKLVT DLTKVHTECC HGDLLECADD RADLAKYICE NQDSISSKLK
300




ECCEKPLLEK SHCIAEVEND EMPADLPSLA ADFVESKDVC KNYAEAKDVF LGMFLYEYAR
360




RHPDYSVVLL LRLAKTYETT LEKCCAAADP HECYAKVFDE FKPLVEEPQN LIKQNCELFE
420




QLGEYKFQNA LLVRYTKKVP QVSTPTLVEV SRNLGKVGSK CCKHPEAKRM PCAEDYLSVV
480




LNQLCVLHEK TPVSDRVTKC CTESLVNRRP CFSALEVDET YVPKEFNAET FTFHADICTL
540




SEKERQIKKQ TALVELVKHK PKATKEQLKA VMDDFAAFVE KCCKADDKET CFAEEGKKLV
600




AASQAALGL
609





40
IPI00784842
GRFTISGDISTNTLYLQMHSLR (Sequence 85-106)




gi:
TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK (Sequence 284-316)





ALPAPIEK (Sequence 355-362)





GQPREPQVYTLPPSRDELTKGFYPSDIAVEWESNGQPENNYK (Sequence 369-420)






41
IPI00022434
Same as KH28





gi: 113576





P02768.2






42
IPI00298497
Same as KH36





gi: 399492





P02675.2






43
IPI00965713
MKRMVSWSFH KLKTMKHLLL LLLCVFLVKS QGVNDNEEGF FSARGHRPLD KKREEAPSLR
60



gi: 399492
PAPPPISGGG YRARPAKAAA TQKKVERKAP DAGGCLHADP DLGVLCPTGC QLQEALLQQE
120



P02675.2
RPIRNSVDEL NNNVEAVSQT SSSSFQYMYL LKDLWQKRQK QVKDNENVVN EYSSELEKHQ
180




LYIDETVNSN IPTNLRVLRS ILENLRSKIQ KLESDVSAQM EYCRTPCTVS CNIPVVSGKE
240




CEEIIRKGGE TSEMYLIQPD SSVKPYRVYC DMNTENGGWT VIQNRQDGSV DFGRKWDPYK
300




QGFGNVATNT DGKNYCGLPG EYWLGNDKIS QLTRMGPTEL LIEMEDWKGD KVKAHYGGFT
360




VQNEANKYQI SVNKYRGTAG NALMDGASQL MGENRTMTIH NGMFFSTYDR DNDGWLTSDP
420




RKQCSKEDGG GWWYNRCHAA NPNGRYYWGG QYTWDMAKHG TDDGVVWMNW KGSWYSMRKM
480




SMKIRPFFPQ Q
491





44
IPI00645363
NSLYLQMNSLRAEDTALYYCAK (Sequence 96-117)




gi:
GPSVFPLAPSSK (Sequence 147-158)





TPEVTCVVVDVSHEDPEVK (Sequence 281-299)





FNWYVDGVEVHNAK (Sequence 300-313)





ALPAPIEK (Sequence 352-359)





GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYK (Sequence 366-417)






45
IPI00219713
Same as KH30




gi: 20178280





P02679







46
IPI00022371
MKALIAALLL ITLQYSCAVS PTDCSAVEPE AEKALDLINK RRRDGYLFQL LRIADAHLDR
60



gi: 123523
VENTTVYYLV LDVQESDCSV LSRKYWNDCE PPDSRRPSEI VIGQCKVIAT RHSHESQDLR
120



P04196.1
VIDFNCTTSS VSSALANTKD SPVLIDFFED TERYRKQANK ALEKYKEEND DFASFRVDRI
180




ERVARVRGGE GTGYFVDFSV RNCPRHHFPR HPNVFGFCRA DLFYDVEALD LESPKNLVIN
240




CEVFDPQEHE NINGVPPHLG HPFHWGGHER SSTTKPPFKP HGSRDHHHPH KPHEHGPPPP
300




PDERDHSHGP PLPQGPPPLL PMSCSSCQHA TFGTNGAQRH SHNNNSSDLH PHKHHSHEQH
360




PHGHHPHAHH PHEHDTHRQH PHGHHPHGHH PHGHHPHGHH PHGHHPHCHD FQDYGPCDPP
420




PHNQGHCCHG HGPPPGHLRR RGPGKGPRPF HCRQIGSVYR LPPLRKGEVL PLPEANFPSF
480




PLPHHKHPLK PDNQPFPQSV SESCPGKFKS GFPQVSMFFT HTFPK
525





47
IPI00022371
Same as KH46





gi: 123523





P04196.1






48
IPI00022463
MRLAVGALLV CAVLGLCLAV PDKTVRWCAV SEHEATKCQS FRDHMKSVIP SDGPSVACVK
60



gi: 313104271
KASYLDCIRA IAANEADAVT LDAGLVYDAY LAPNNLKPVV AEFYGSKEDP QTFYYAVAVV
120



P02787.3
KKDSGFQMNQ LRGKKSCHTG LGRSAGWNIP IGLLYCDLPE PRKPLEKAVA NFFSGSCAPC
180




ADGTDFPQLC QLCPGCGCST LNQYFGYSGA FKCLKDGAGD VAFVKHSTIF ENLANKADRD
240




QYELLCLDNT RKPVDEYKDC HLAQVPSHTV VARSMGGKED LIWELLNQAQ EHFGKDKSKE
300




FQLFSSPHGK DLLFKDSAHG FLKVPPRMDA KMYLGYEYVT AIRNLREGTC PEAPTDECKP
360




VKWCALSHHE RLKCDEWSVN SVGKIECVSA ETTEDCIAKI MNGEADAMSL DGGFVYIAGK
420




CGLVPVLAEN YNKSDNCEDT PEAGYFAIAV VKKSASDLTW DNLKGKKSCH TAVGRTAGWN
480




IPMGLLYNKI NHCRFDEFFS EGCAPGSKKD SSLCKLCMGS GLNLCEPNNK EGYYGYTGAF
540




RCLVEKGDVA FVKHQTVPQN TGGKNPDPWA KNLNEKDYEL LCLDGTRKPV EEYANCHLAR
600




APNHAVVTRK DKEACVHKIL RQQQHLFGSN VTDCSGNFCL FRSETKDLLF RDDTVCLAKL
660




HDRNTYEKYL GEEYVKAVGN LRKCSTSSLL EACTFRRP
698





49
IPI00023006
MCDDEETTAL VCDNGSGLVK AGFAGDDAPR AVFPSIVGRP RHQGVMVGMG QKDSYVGDEA
60



gi: 54036697
QSKRGILTLK YPIEHGIITN WDDMEKIWHH TFYNELRVAP EEHPTLLTEA PLNPKANREK
120



P68032.1
MTQIMFETFN VPAMYVAIQA VLSLYASGRT TGIVLDSGDG VTHNVPIYEG YALPHAIMRL
180




DLAGRDLTDY LMKILTERGY SFVTTAEREI VRDIKEKLCY VALDFENEMA TAASSSSLEK
240




SYELPDGQVI TIGNERFRCP ETLFQPSFIG MESAGIHETT YNSIMKCDID IRKDLYANNV
300




LSGGTTMYPG IADRMQKEIT ALAPSTMKIK IIAPPERKYS VWIGGSILAS LSTFQQMWIS
360




KQEYDEAGPS IVHRKCF
377





50
IPI00021841
Same as KH 37




gi: 113992





P02647.1







51
IPI00023006
Same as KH49




gi: 54036697





P68032.1







52
IPI00930226
MEEEIAALVI DNGSGMCKAG FAGDDAPRAV FPSIVGRPRH QGVMVGMGQK DSYVGDEAQS
60



gi: 54036678
KRGILTLKYP IEHGIVTNWD DMEKIWHHTF YNELRVAPEE HPVLLTEAPL NPKANREKMT
120



P63261.1
QIMFETFNTP AMYVAIQAVL SLYASGRTTG IVMDSGDGVT HTVPIYEGYA LPHAILRLDL
180




AGRDLTDYLM KILTERGYSF TTTAEREIVR DIKEKLCYVA LDFEQEMATA ASSSSLEKSY
240




ELPDGQVITI GNERFRCPEA LFQPSFLGME SCGIHETTFN SIMKCDVDIR KDLYANTVLS
300




GGTTMYPGIA DRMQKEITAL APSTMKIKII APPERKYSVW IGGSILASLS TFQQMWISKQ
360




EYDESGPSIV HRKCF
375





53
gi: 194373497
MEESLPTNPD SSTMGPWCYT TDPTVRRQEC SIPVCGQDQV TVAMTPRSEG SSVNLSPPLE
60



BAG56844.1
QCVPDRGQQY QGRLAVTTHG LPCLAWASAQ AKALSKHQDF NSAVQLVENF CRNPDGDEEG
120




VWCYVAGKPG DFGYCDLNYC EEAVEEETGD GLDEDSDRAI EGRTATSEYQ TFFNPRTFGS
180




GEADCGLRPL FEKKSLEDKT ERELLESYID GRIVEGSDAE IGMSPWQVML FRKSPQELLC
240




GASLISDRWV LTAAHCLLYP PWDKNFTEND LLVRIGKHSR TRYERNIEKI SMLEKIYIHP
300




RYNWRENLDR DIALMKLKKP VAFSDYIHPV CLPDRETAAS LLQAGYKGRV TGWGNLKETW
360




TANVGKGQPS VLQVVNLPIV ERPVCKDSTR IRITDNMFCA GYKPDEGKRG DACEGDSGGP
420




FVMKSPFNNR WYQMGIVSWG EGCDRDGKYG FYTHVFRLKK WIQKVIDQFG E
471





54
gi: 194380034
MNQLRGKKSC HTGLGRSAGW NIPIGLLYCD LPEPRKPLEK AVANFFSGSC APCADGTDFP
60



BAG58369.1
QLCQLCPGCG CSTLNQYFGY SGAFKCLKDG AGDVAFVKHS TIFENLANKA DRDQYELLCL
120




DNTRKPVDEY KDCHLAQVPS HTVVARSMGS KEDLIWELLN QAQEHFGKDK SKEFQLFSSP
180




HGKDLLFKDS AHGFLKVPPR MDAKMYLGYE YVTAIRNLRE GTCPEAPTDE CKPVKWCALS
240




HHERLKCDEW SVNSVGKIEC VSAETTEDCI AKIMNGEADA MSLDGGFVYI AGKCGLVPVL
300




AENYNKSDNC EDTPEAGYFA VAVVKKSASD LTWDNLKGKK SCHTAVGRTA GWNIPMGLLY
360




NKINHCRFDE FFSEGCAPGS KKDSSLCKLC MGSGLNLCEP NNKEGYYGYT GAFRCLVEKG
420




DVAFVKHQTV PQNTGGKNPD PWAKNLNEKD YELLCLDGTR KPVEEYANCH LARAPNHAVV
480




TRKDKEACVH KILRQQQHLF GSNVTDCSGN FCLFRSETKD LLFRDDTVCL AKLHDRNTYE
540




KYLGEEYVKA VGNLRKCSTS SLLEACTFRR P
571





55
gi: 194380034
Same as 54




BAG58369.1









By the present invention it has been shown that purified plasmas containing varying combinations and concentrations of KH proteins have vast implications for the treatment of a large host of diseases, viral infections, and other disorders. As described in more detail below, embodiments of the current invention involve purified plasma constructs, their newly discovered proteins, and their use in the treatment of: HIV 1 and 2; hepatitis B; hepatitis C; influenza; glucose uptake related disorders, e.g. diabetes; atherosclerosis and related cardiovascular diseases; high cholesterol levels; H1N1; arthritis; tumor progression; and parkinson's disease.


Embodiments of the Invention

Study Title: In Vitro Anti-HIV Activity of Human Plasma Derived Proteins on HIV-RT Enzyme


I. Study Objective:


To Analyze Human Plasma Derived Proteins for Anti-HIV Activity on HIV-RT Enzyme


II. Study Protocols:


1. Materials:


1.1 Samples Information:


RAAS provided the test articles in the form of dry powder or liquid (Table 1). Wuxi provided reference compound in DMSO solution.









TABLE 1







Sample information










Name
Protein conc.
Formulation
Diluents





AFOD KH
   10%
Liquid



AFCC KH
  3.50%
Liquid


AFCC RAAS 1
    4%
Lyophilized
AFOD KH 10 mL


AFCC RAAS 4
 0.0020%
Lyophilized
AFOD KH 10 mL


AFCC RDNA
0.00001%
Lyophilized
AFOD KH 10 mL









1.2 Reagents:









TABLE 2







List of reagents









Reagents/Plates
Vendor
Cat.#





HIV-1 Reverse
Merck
382129-500U


Transcriptase wild


type enzyme


Avidin standard plates
MSD
MSD-L15AA-6


RNA template t500
IBA GMBH
Cat. #89142N/S


synthetic piece of RNA


CHAPS
Pierce
Pierce-28300


EGTA
Sigma
Sigma-E3889-10G


DTT
Sigma
Sigma-43815-5G


d-ATP
Sigma
Sigma-D6500-10MG


d-GTP
Sigma
D4010-10MG


d-CTP-Na 2
Sigma
D4635-10MG


Water (DEPC treated)
Invitrogen
Invitrogen-750023


dry biopD500 primer
Shanghai



Shenggong


BSA
Sigma
Sigma-A3294


4x Read buffer T
MSD
MSD-R92TD-1


Ru-d-UTP
MSD
Lot: DG2005245071


96-well round bottom
Costar
Costar-3365


polypropylene plates


PCR tubes
AXYGEN
AXYGEN-PCR-0208-C


PCR tube covers
AXYGEN
AXYGEN-PCR-2CP-RT-C









1.3 Instrument

    • Sector Imager S6000 (MesoScale Discovery MSD)
    • Epmotoin (Eppendorf)
    • Janus (perkinelmer)
    • Orbital shaker


2. Methods


2.1 IC50 Measurement


2.2.1 Drug Treatment:


Human plasma derived protein dilutions are made by using EpMotion with 2-fold serial dilutions for 10 concentrations, each in duplicate.

    • a) Add 30 μL of enzyme solution per well of the Costar 96 well plates.
    • b) Add 5 μL of test article or PBS or DMSO.
    • c) Seal plate and shake for 2 minutes on an orbital shaker
    • d) Incubate for 30 minutes on an orbital shaker at room temperature.
    • e) Add 15 μL of the Master Mix to initiate the reaction.
    • f) Seal plate and shake for 5-10 minutes.
    • g) Incubate at 37 degree for 90 minutes.
    • h) While this is incubating, add 100 pt of 5% BSA in PBS to the wells of the avidin plates.
    • i) Seal the avidin plates and incubate for 1 hour at room temperature.
    • j) After the 90 minute incubation, add 60 μL of quenching buffer to the reaction wells.
    • k) Seal the plates and incubate for 5 minutes on the plate shaker.
    • l) Transfer 50 μL of the well contents to MSD blocked plates (the blocking buffer is simply dumped off. No wash is needed).
    • m) Incubate MSD plates at RT for 60 minutes.
    • n) Freshly dilute the 4× read buffer T to 1× using distilled water (not DEPC-treated)
    • o) Wash MSD plates 3 times with 150 μL of PBS per well per wash.
    • p) Add 150 pt of 1× read buffer T to the wells.
    • q) Read on the Sector Imager Instrument.


2.2.2 Sample or Compound Addition


Test samples were diluted in PBS as 3.5×104 μg/ml stocks. Sample dilutions are made by using Epmotion with 2-fold serial dilutions for 10 concentrations plus PBS (see below for final compound concentrations in the HIV-RT enzyme assay). Reference compound were dissolved in DMSO as 10 mM stocks and dilutions are made by using Epmotion with 3-fold serial dilutions for 10 concentrations plus DMSO (see below for final compound concentrations).









TABLE 3







Sample or compound concentrations for IC50 measurement







Name












Concentration (ug/ml)

















AFOD KH
400
200
100
50
25
12.5
6.25
3.1
1.6
0.8


AFCC KH
400
200
100
50
25
12.5
6.25
3.1
1.6
0.8


AFCC RAAS 1
400
200
100
50
25
12.5
6.25
3.1
1.6
0.8


AFCC RAAS 4
400
200
100
50
25
12.5
6.25
3.1
1.6
0.8


AFCC RDNA
400
200
100
50
25
12.5
6.25
3.1
1.6
0.8









Concentration (nM)

















Reference
100
33.3
11.1
3.7
1.2
0.4
0.1
0.05
0.02
0.01


Compound









2.2.3 Data Analysis:


Percent of HIV-RT inhibition by protein or compound is calculated using the following equation:





% Inh.=[1−(Signal of sample−Signal of control)/(Signal of DMSO or PBS control−Signal of control)]*100.


Dose-response curves are plotted using Prism


III. Assay Results:


3.1 Raw Data from the HIV-RT Enzyme Assay.


3.1.1 HIV-RT Enzyme Assay Plate Map*:


Plate 1


























column
column
column
column
column
column
column
column
column
column
column
column



1
2
3
4
5
6
7
8
9
10
11
12




















raw
A
PBS
AFOD KH
BG


raw
B


raw
C

AFCC KH


raw
D


raw
E
BG
AFCC RAAS 1
PBS


raw
F


raw
G

Reference Compound


raw
H





* BG: background






Plate 2


























column
column
column
column
column
column
column
column
column
column
column
column



1
2
3
4
5
6
7
8
9
10
11
12




















raw
A
PBS
AFCC RAAS 4
BG


raw
B


raw
C

AFCC RDNA


raw
D


raw
E
BG
Reference Compound
PBS


raw
F


raw
G

DMSO


raw
H





* BG: background






3.1.2 Raw Data


Plate 1:


























column
column
column
column
column
column
column
column
column
column
column
column



1
2
3
4
5
6
7
8
9
10
11
12





























raw
A
2439
1596
2113
2160
2304
2448
2214
2152
2307
2360
2357
60


raw
B
2569
1866
2154
2343
2351
2371
2397
2317
2310
2454
2245
64


raw
C
2571
281
329
393
563
805
1157
1683
2011
2304
2384
60


raw
D
2361
267
306
376
518
762
1156
1600
1912
2158
2185
58


raw
E
59
1238
1782
2097
2230
2299
2326
2374
2368
2329
2449
2267


raw
F
52
1248
1812
2166
2300
2406
2462
2398
2369
2346
2353
2366


raw
G
54
87
142
246
469
850
1241
1629
1791
1873
1851
2263


raw
H
53
85
132
241
474
833
1349
1651
1813
1924
1907
2438









Plate 2:


























column
column
column
column
column
column
column
column
column
column
column
column



1
2
3
4
5
6
7
8
9
10
11
12





























raw
A
2491
1713
1940
2168
2411
2358
2378
2459
2289
2262
2038
43


raw
B
2596
1674
2220
2344
2547
2491
2418
2541
2443
2476
2104
45


raw
C
2539
1747
2176
2381
2522
2388
2433
2314
2459
2358
2369
44


raw
D
2544
1689
2123
2305
2453
2385
2400
2426
2204
2049
2168
39


raw
E
44
91
146
270
514
957
1429
1801
1807
1895
1880
2142


raw
F
38
85
139
263
472
946
1377
1614
1708
1850
1853
2292


raw
G
45
2119
2160
2084
2046
2069
1963
1975
2002
1961
1912
2343


raw
H
43
2052
2038
2039
1975
1954
1860
1968
1972
1875
2042
2405









3.2 Activity of the Samples or Compounds.


IC50 values are summarized in Table 4. GraphPad Prism files containing dose-dependent curves are presented in this report, as shown in FIG. 1.


4. Conclusions


The Z factors of the two plate were 0.84 (plate 1), 0.80 (plate 2), which were much better than QC standard of 0.5. Therefore, the assay data met our QC qualification.

    • The IC50s of positive control in this study were 0.9 nM (plate 1), 1.2 nM (plate 2) and these results are consistent with our previous data.


The Results of Neutralization of HIV-1 Env-Pseudotyped Virus


Samples and Control

  • 1. Test samples: 3 in total, AFCC KH, AFCC RAAS and AFOD RAAS110 respectively
  • 2. Positive control compound: 3 in total, AMD 3100 (inhibitor of CXCR4), nifeviroc (inhibitor of CCR5) and Ibalizumab (anti-CD4 monoclonal antibody). All three drugs can inhibit the entry of virus into cells.
    • AMD 3100 was initially developed at the Johnson Matthey Technology Centre for potential use in the treatment of HIV because of its role in the blocking of CXCR4, a chemokine receptor which acts as a co-receptor for certain strains of HIV (along with the virus's main cellular receptor, CD4).
    • NIFEVIROC, a small molecular compound, is a proprietary drug candidate which is developed by TARGETDRUG and holds great promise in inhibiting HIV-1 replication in infected patients. Preclinical studies suggested that NIFEVIROC is a specific CCR5 antagonist determined by multiple receptor functional assays.
    • Ibalizumab (TMB-355 previously known as TNX-355) is a non-immunosuppressive monoclonal antibody that binds CD4, the primary receptor for HIV, and inhibits the viral entry process.
  • 3. Tested virus: 10 strains, they are
    • (1) B′ subtype virus: CNE6 and CNE11;
      • BC recombinant subtype virus: CNE15 and CNE30;
      • CRF01_AE recombinant subtype virus: CNE5 and CNE55;
      • The standard HIV-1 strain virus: sf162, HXB2 and JRFL,
      • All above HIV-1 virus are CCR5 receptor affinity except HXB2 is CXCR4 receptor affinity.
    • (2) Control virus: AMLV.


Test Method

  • 1. Test samples were diluted at 1:20 as start and then 1:60, 1:180, 1:540, 1:1620, 1:4860, 1:14580, 1:43740. It was 3-fold dilution and 8 dilutions in total.
  • 2. Positive control drug was started to dilute from:
    • AMD3100: 10 uM
    • Nifeviroc: 0.05 uM
    • Ibalizumab: 10 ug/ml
    • It was 3-fold dilution and 8 dilutions in total.


Results

  • 1. AFCC KH: no activity in preventing the entry of virus into cells. But it enhances virus invade into cells.
  • 2. AFOD RAAS 110 (AT III): no activity in preventing the entry of virus into cells. But it strongly helps virus invade into cells.
  • 3. AFCC RAAS (15% PCC): it showed 50% inhibition of virus in five strains of virus, which are CNE15, CNE30, CNE55, HXB2 and JRFL, with dose dependent response. It showed weak inhibition in two strains (CNE5 and AMLV) and no effects on the rest 3 strains (CNE6, CNE 11 and sf162)



FIG. 2


The Supplementary Results of Neutralization of HIV-1 Env-Pseudotyped Virus


Samples and Control

  • 4. Test samples: AFCC RAAS
  • 5. Tested virus: 5 strains, they are
    • (3) BC recombinant subtype virus: CNE15 and CNE30;
      • CRF01_AE recombinant subtype virus: CNE55;
      • The standard HIV-1 strain virus: HXB2 and JRFL,
      • All above HIV-1 virus are CCR5 receptor affinity except HXB2 is CXCR4 receptor affinity.
    • (4) Control virus: AMLV.


Test Method

  • 3. Test samples were diluted at 1:1.5 as start and then 1:4.5, 1:13.5, 1:40.5, 1:121.5, 1:364.5, 1:1093.5, and 1:3280.5. It was 3-fold dilution and 8 dilutions in total.


Results

  • 4. It has been shown in FIG. 1 that the inhibition rate of AFCC RAAS in 5 HIV-1 strains and control virus AMLV. The conclusion is that the inhibition rate is about 60% when the dilution was less than 1:40 and the inhibition also was observed in control virus AMLV. Cell toxicity was found in high concentrations via observing cell morphology 48 hours after treatment. Thus cell toxicity test was then conducted.



FIG. 3


  • 4. Cell toxicity test: in this study, we tested the toxicity of AFCCKH, AFOD RAAS 101 and AFCC RAAS. Test samples were diluted at 1:1.5 as start and then 1:4.5, 1:13.5, 1:40.5, 1:121.5, 1:364.5, 1:1093.5, and 1:3280.5. It was 3-fold dilution and 8 dilutions in total. The test kit is cell counting kit 8 (CCK-8). The procedure is according to manufacturer's manual. It has been shown that there is some cell toxicity of RAAS. The inhibition of HIV virus probably is caused by cell toxicity.




FIG. 4


Suggestion for Further Study


To decrease the toxicyte to cell, and ensure the high inhibition of virus at high protein concentration.

    • 1. further increase the protein concentration.
    • 2. Use cell culture medium (DMEM+10% FBS) as the diluent of products when preparing the samples.


Study Title: Test Human Plasma Derived Proteins Against HCV Genotype 1a, 1b and 2a Replicons for Antiviral Activity (EC50)


I. Study Objective


To Analyze Human Plasma Derived Proteins for Anti-HCV Activity (EC50) and Cytotoxicity (CC50) Using HCV 1a, 1b and 2a Replicon Culture Systems


II. Study Protocols


3. Materials:


1.1 Cell Line:


Replicon cell lines 1a and 2a were established following published methods (1,2) using Huh7 by G418 selection. The replicons were assembled using synthetic gene fragments. The GT 1a line is derived from H77 and contains PVIRES-Luciferase-Ubi-Neo, and two adaptive mutations: P1496L, S22041. The 2a line contains no adaptive mutations and encodes a Luciferase reporter. The 1b replicon plasmid is also assembled using synthetic gene fragments. The replicon genome contains PVIRES-Luciferase Ubi-Neo gene segments and harbors 1 adaptive mutation (S22041), and the backbone is Con1.


1.2 Compounds:


The test articles are supplied in the form of dry powder or 10 mM solution, and Ribavirin as control, in duplicate.


1.3 Reagents:









TABLE 1







List of reagents











Reagent
Vendor
Catalog Number







Dimethyl sulfoxide (DMSO)
Sigma
Cat#34869



DMEM
Invitrogen
Cat#11960-044



Fetal Bovine Serum (FBS)
Gibco
Cat#16140



Penicillin-Streptomycin
Invitrogen
Cat#15070063



MEM non-essential amino acids
Invitrogen
cat#11140-050



L-Glutamine
Invitrogen
Cat#25030-081



Trypsin/EDTA
Invitrogen
Cat#25200-072



DPBS/Modified
Hyclone
SH30028.01B



96 well cell plate
Greiner
Cat#655090



CellTiter fluor
Promega
Cat#G6082



Bright-Glo
Promega
Cat#E2650










1.4 Instrument

    • Envision (Perkinelmer)
    • Multidrop (Thermo)
    • Janus (Perkinelmer)


4. Methods


2.1 Cell Addition


T150 flask containing 1a, 1b and 2a replicons cell monolayer is rinsed with 10 ml pre-warmed PBS. Add 3 ml of pre-warmed Trypsin 0.25% and incubate at 5% CO2, 37□ for 3 minutes. Nine milliliters of DMEM complete media are added, and the cells are blown for 30s by pipetting. The cells are counted using hemocytometer.


1a, 1b and 2a replicons cells are resuspended in medium containing 10% FBS to reach a cell density of 64,000 cells/ml (to obtain a final cell plating density of 8000 cells/125 ul/well). Plate cells in Greiner 96 black plate using Multidrop. Incubate plate at 5% CO2, 37□ for 4 hours.


2.2 Compound Addition


RAAS provided the test articles in the form of dry powder or liquid (Table 2). Test samples were diluted in PBS as 3.5×104 μg/ml stocks. Sample dilutions are made by Janus with 2-fold serial dilutions for 10 concentrations plus PBS. Ribavirin is also diluted by Janus with 2-fold for 10 concentrations. The final sample concentrations of the HCV replicon assay are described in Table 3.









TABLE 2







Sample information










Name
Protein conc.
Formulation
Diluents





AFOD KH
   10%
Liquid



AFCC KH
  3.50%
Liquid


AFCC RAAS 1
    4%
Lyophilized
AFOD KH 10 mL


AFCC RAAS 4
 0.0020%
Lyophilized
AFOD KH 10 mL


AFCC RDNA
0.00001%
Lyophilized
AFOD KH 10 mL
















TABLE 3







Sample or compound concentrations for EC50 and CC50 measurement








Name
HCV Genotype












Concentration (μg/ml)


















AFOD KH
1a/1b/2a
400
200
100
50
25
12.5
6.3
3.1
1.6
0.8


AFCC KH

400
200
100
50
25
12.5
6.3
3.1
1.6
0.8


AFCC RAAS 1

400
200
100
50
25
12.5
6.3
3.1
1.6
0.8


AFCC RAAS 4

400
200
100
50
25
12.5
6.3
3.1
1.6
0.8


AFCC RDNA

400
200
100
50
25
12.5
6.3
3.1
1.6
0.8









Concentration (μM)


















Ribavirin

320
160
80
40
20
10
5
2.5
1.3
0.6









2.3 Detection (after 72 Hours of Incubation)


Bright-Glo Luiferase and CellTiter-Fluor™ are prepared and stored in dark while allowing to equilibrate to room temperature. Plates are removed from incubator to allow equilibration to room temperature. Multidrop is used to add 40 ul CellTiter-Fluor™ to each well of compound-treated cells. The plates are incubated for 0.5 hour, and then read on an Envision reader for cytotoxicity calculation. The cytotoxicity is calculates using the equation below.







%





Cytotoxicity

=


(

1
-


Cmpd
-
Background


DMSO
-
Background



)

×
100





100 ul of Bright-Glo are added to each well, incubated for 2 minutes at room temperature, and chemi-luminescence (an indicator of HCV replication) is measured for EC50 calculation.


The anti-replicon activity (% inhibition) is calculated using the equation below







%





Inhibition

=


(

1
-


Cmpd
-
background


DMSO
-
background



)

×
100





Dose-response curves are plotted using Prism.


III. Assay Results


1 Assay Plate Map


2 Raw Data


2.1 Raw Data of Cytotoxicity Assay


2.2 Raw Data of Anti-Replicon Activity Assay


3 Cytotoxicity and Anti-Replicon Activity of the Human Plasma Derived Proteins.


CC50 and EC50 values are summarized in Table 4. GraphPad Prism files containing dose-dependent curves are presented in this report. CC50 and EC50 values are shown in FIG. 1 and FIG. 2 respectively.


IV. Conclusions

    • The Z factors of the cytotoxicity assay plates are 0.83 (1a-plate1), 0.79 (1a-plate2), 0.71 (1b-plate1), 0.68 (1b-plate2), 0.65 (2a-plate1) and 0.83 (2a-plate2), which are better than our QC standard.
    • The Z factors of the anti-replicon assay plates are 0.75 (1a-plate1), 0.70 (1a-plate2), 0.87 (1b-plate1), 0.75 (1b-plate2), 0.58 (2a-plate1) and 0.75 (2a-plate2), which are better than our QC standard.
    • EC50 of the positive control Ribavirin in this study are 57.58 uM (1a), 39.04 uM (1b), and 37.44 (2a), which are consistent with our previous data.


V. References

  • 1. Mutations in Hepatitis C Virus RNAs Conferring Cell Culture Adaptation V. Lohmann et al., 2001 J. Virol.
  • 2. Development of a replicon-based phenotypic assay for assessing the drug susceptibilities of HCV NS3 protease genes from clinical isolates. Qi X et al., Antiviral Res. 2009 February; 81(2:)166-73


In Vitro Anti-HBV Efficacy Test


Method and Materials


1) Cell model: HepG2 cell infected with HBV virus, which is HepG2 2.2.15 cell


2) Cell viability is analyzed by MTT method


3) EIA test to detect the inhibition of HBsAg and HBeAg


4) Positive control drug: Lamivudine


5) RT-PCR detection of HBV-DNA


Procedure


1) Toxicity of Drug to Cell


HepG2 2.2.15 cells are seeded in 96-well plate. Fresh medium with various concentration of drug is added 48 hour later. Cell viability is analyzed 9 days later by MTT method.


2) The Inhibition of HBV Virus


HepG2 2.2.15 cells are seeded in 96-well plate. Fresh medium with various concentration of drug is added 48 hour later. The HBsAg and HBeAg are detected 5 days, 7 days, and 10 days later. RT-PCR detection of HBV-DNA


Results














AFOD
HBsAg
HBeAg











(μg/mL)
OD
Inhibition rate %
OD
Inhibition rate %














10
0.611
47.6
1.020
17.6


5
0.695
40.4
1.059
14.5


2.5
0.775
33.5
1.115
10.0


1.25
0.897
23.1
1.165
5.9


Negative control
1.166
/
1.238
/









Study Title: In Vitro Test of Human Plasma Derived Proteins Against Influenza for Antiviral Activity (EC50)


Influenza Study


I. Study Objective


To Test 2 Compounds from RAAS for Anti-Influenza Activity Against Strains A/Weiss/43 H1N1 in Cell Culture


II. Study Protocols:


3. Materials:


Cell Line:


MDCK cells


1.2 Compounds:


The test articles are supplied in the form of dry powder or 10 mM solution, and Oseltamivir as control, in duplicate.


1.3 Reagents:


The following table designations, such as Table 5.1, refer to tables of a first group of tables in the present application. Other groups of tables in the present application, which will be referred to later in the application, will contain some tables that have the same designations as tables of the first group.









TABLE 5.1







List of reagents and consumable











Reagent
Vendor
Catalog Number







Dimethyl sulfoxide (DMSO)
Sigma
Cat#D8418



SFM
Invitrogen
Cat# 12309-019



Fetal Bovine Serum (FBS)
Gibco
Cat#16140



Penicillin-Streptomycin
Invitrogen
Cat# 15140-122



MEM non-essential amino
Invitrogen
cat# 11140-076



acids



GlutaMAX-I Supplement
Invitrogen
Cat# 35050-061



Trypsin/EDTA
Invitrogen
Cat# 25300-062



PBS
Invitrogen
Cat#10010-049



DPBS/Modified
Hyclone
SH30028.01B



96 well cell plate
Corning
Cat#3599



MTT
sigma
Cat# M2128










1.4 Instrument

    • speterphotemeter (Molecular Devices)
    • Multidrop (Thermo)
    • Janus (perkinelmer)


4. Methods


2.1 Cell Addition


T150 flask containing MDCK cell monolayer is rinsed with 10 ml pre-warmed PBS. Add 3 ml of pre-warmed Trypsin 0.25% and incubate at 5% CO2, 37□ for 3 minutes. Nine milliliters of DMEM complete media are added, and the cells are blown for 30s by pipetting. The cells are counted using hemocytometer. MDCK cells are resuspended in SFM medium to reach a cell density of 50,000 cells/ml (to obtain a final cell plating density of 5000 cells/100 ul/well). Plate cells in 96 well plate using Multidrop. Incubate plate at 5% CO2, 37□ for overnight.


2.2 Compound Addition


RAAS provided the test articles in the form of dry powder or liquid (Table 5.2). Test samples were diluted in PBS as 3.5×104 μg/ml stocks. Sample dilutions are made by Janus with 2-fold serial dilutions for 8 concentrations plus PBS. Osletamivir is diluted with 3-fold for 8 concentrations. The final sample concentrations of the anti-influenza assay are described in Table 5.3.









TABLE 5.2







Sample information










Name
Protein conc.
Formulation
Diluents





AFOD KH
   10%
Liquid



AFCC KH
  3.50%
Liquid


AFCC RAAS 1
    4%
Lyophilized
AFOD KH 10 mL


AFCC RAAS 4
 0.0020%
Lyophilized
AFOD KH 10 mL


AFCC RDNA
0.00001%
Lyophilized
AFOD KH 10 mL
















TABLE 5.3





Sample or compound concentrations for EC50 and CC50 measurement
















Name
Concentration (μg/ml)















AFOD KH
400
200
100
50
25
12.5
6.3
3.1


AFCC KH
400
200
100
50
25
12.5
6.3
3.1


AFCC RAAS 1
400
200
100
50
25
12.5
6.3
3.1


AFCC RAAS 4
400
200
100
50
25
12.5
6.3
3.1


AFCC RDNA
400
200
100
50
25
12.5
6.3
3.1








Osletamivir
Concentration (μM)
















100.00
33.33
11.11
3.70
1.23
0.41
0.14
0.05









2.3 Detection (after 72 Hours of Incubation)


MTT solution is prepared freshly. Plates are removed from incubator to allow equilibration to room temperature. Multidrop is used to add 20 ul MTT to each well of compound-treated cells. The plates are incubated for 4 hour, and then read on a speterphotemeter for EC50 and cytotoxicity calculation.


The anti-influenza activity (% inhibition) is calculated using the equation below







%





Inhibition

=


(

1
-


Cmpd
-
background


DMSO
-
background



)

×
100





The cytotoxicity is calculates using the equation below:





% livability=(Cmpd/PBS control)*100


Dose-response curves are plotted using Prism.


III. Assay Results:


1 Assay Plate Map


2 Raw data


2.1 Raw Data of Anti-Influenza Assay


























1
2
3
4
5
6
7
8
9
10
11
12




























plate 1














A


B

0.93
1.47
1.43
0.24
0.22
0.21
0.18
0.19
0.136
1.504



C

1.032
1.345
1.276
0.455
0.241
0.226
0.203
0.188
0.216
1.439


D

1.348
1.308
1.375
1.485
0.221
0.171
0.197
0.158
0.159
1.506


E

1.362
1.429
1.466
1.386
0.234
0.159
0.173
0.208
0.167
1.565


F

1.486
1.318
0.963
0.264
0.173
0.173
0.185
0.181
0.163
1.477


G

1.584
1.432
0.948
0.322
0.224
0.217
0.205
0.149
0.131
1.468


H


plate 2


A


B

1.48
1.39
0.81
0.27
0.22
0.18
0.14
0.17
0.180
1.279


C

1.464
1.294
0.668
0.236
0.174
0.224
0.176
0.179
0.189
1.261


D

1.411
1.238
0.279
0.183
0.207
0.237
0.175
0.177
0.150
1.262


E

1.418
1.128
0.306
0.211
0.180
0.178
0.231
0.176
0.172
1.238


F

1.290
1.382
1.296
1.266
0.969
0.563
0.544
0.386
0.353
1.319


G

1.292
1.218
1.210
1.295
0.962
0.627
0.431
0.388
0.394
1.397


H









Raw Data of Cytotoxicity Assay


























1
2
3
4
5
6
7
8
9
10
11
12




























plate 1














A


B

1.49
1.61
1.58
1.42
1.03
1.18
1.13
1.10
1.161
1.209


C

1.593
1.550
1.482
1.440
0.995
1.173
1.337
1.043
1.122
1.261


D

1.366
1.332
1.230
1.301
1.321
1.279
1.227
1.322
1.238
1.306


E

1.308
1.323
1.225
1.273
1.268
1.247
1.274
1.357
1.318
1.326


F

1.788
1.718
1.471
1.418
1.406
1.373
1.295
1.340
1.257
1.270


G

1.798
1.741
1.455
1.543
1.471
1.320
1.352
1.367
1.275
1.216


H


plate 2


A


B

1.793
1.799
1.852
1.776
1.796
1.639
1.626
1.650
1.626
1.524


C

1.842
1.870
1.818
1.939
1.773
1.690
1.631
1.649
1.675
1.564


D

1.822
1.897
1.849
1.891
1.688
1.689
1.641
1.637
1.713
1.617


E

1.830
1.944
1.913
1.874
1.812
1.606
1.630
1.652
1.605
1.570


H









3 Cytotoxicity and Anti-Influenza Activity of the Human Plasma Derived Proteins.


CC50 and EC50 values are summarized in Table 5.4. GraphPad Prism files containing dose-dependent curves are presented in this report. CC50 and EC50 values are shown in FIG. 26.17 and FIG. 26.21 respectively.









TABLE 5.4





CC50 and EC50 Summary of the human plasma derived proteins



















cpds
anti H1N1 EC50s (ug/ml)
CC50s (ug/ml)















AFOD KH
69.06
>400



AFCC KH
35.37
>400



AFCC RAAS 1
89.63
>400



AFCC RAAS 4
108.40
>400



AFCC RDNA
154.90
>400







cpds
anti H1N1 EC50s (uM)







Oseltamivir
0.89










IV. Conclusions

    • The EC50 of the positive control Osletamivir in this study is 0.89 uM, which is consistent with our previous data.
    • The human plasma derived proteins showed anti-influenza activity in this study.


Characterization of Cultured Cells for RAAS


Executive Summary


This study is to analyze the cells in culture by flow cytometric analysis. The samples were provided by the client. First, all the samples were counted individually with Vi-CELL Cell Viability Analyzer (Beckman Coulter) for cell number and viability. Then the samples were stained with cellular markers for different lineages including T cells, B cells, granulocytes, natural killer (NK) cells. Normal human peripheral blood sample was used as controls for the staining


Among 59 samples, 30 samples contained cells. Only 10 samples had total cell number above 1×105 and only 5 samples reached viability above 90%. In comparison with forward scatter (FSC)/side scatter (SSC) of distinct subpopulations of human peripheral blood cells, such as lymphocytes, granulocytes, monocytes and macrophages, unknown samples didn't obtain the same distribution shown by FACS Staining and distribution pattern of unknown samples also demonstrated they were not granulocytes, lymphocytes, or NK cells.


List of Abbreviations


















FACS
Flow Cytometry



BSA
Bovine serum albumin



FSC
Forward scatter



SSC
side scatter



NK cells
Natural killer cells










Materials and Methods


Materials


Reagents


FITC, Anti-Human CD66, BD, Cat: 551479


FITC, Anti-Human CD34, BD, Cat: 560942


PE, Anti-Human CD3, BD, Cat: 561803


PE, Anti-Human CD146, BD, Cat: 561013


PE, Anti-Human CD56, BD, Cat: 561903


PE, Anti-Human CD14, BD, Cat: 561707


PE, Anti-Human CD11c, BD, Cat: 560999


PerCP-Cy5.5, Anti-Human CD16, BD, Cat: 560717


APC, Anti-Human CD19, BD, Cat: 561742


PE, Anti-Human CD41a, BD, Cat: 560979


ACK Lysis buffer, Invitrogen, Cat: A10492-01


PBS, Dycent Biotech (Shanghai) CO., Ltd. Cat: BJ141. FBS, Invitrogen Gibco, Cat: 10099141


BSA, Beyotime, ST023


Materials


Cell strainer (70 μm), BD, Cat: 352350


BD Falcon tubes (12×75 mm, 5 ml), BD, Cat: 352054


Equipment


Vi-CELL Cell Viability Analyzer, Beckman Coulter, Cat: 731050


FACSCalibur flow cytometer, BD, Cat: TY1218


Methods


Staining

    • Cells were placed into the 96-well (6×105 cells/well) plate and blocked with 0.08% NaN3/PBS containing 1% FBS, 1% mouse serum and 2% BSA for 15 min at 4° C.
    • Cells were washed once with 1×PBS and resuspended with staining buffer (0.08% NaN3/PBS+1% FBS) with indicated antibodies for 30 min@ 4° C.
    • Cells were washed twice with 0.08% NaN3/PBS (200 μl per well) and resuspended with 400 μl 0.08% NaN3/PBS.
    • Excessive chunk from cell suspension were removed by filtrating through cell strainer. Cells were collected in BD Falcon tubes (12×75 mm, 5 ml) and analyzed by FACSCalibur.


Data Analysis


FACS data were analyzed by flowjo software.


Study Summary


Study Initiation Date and Completion Date


Cell samples were received on Apr. 26, 2012 and analyzed on Apr. 27.


Study Purpose


The purpose of this study was to characterize the unknown cells.


Study Results


Cell Count


59 cell samples were counted individually using Vi-CELL Cell Viability Analyzer (Beckman Coulter). The detailed information was listed in Table 1.









TABLE 1







Cell counting





















Viability


Sample
Denisity ×106/ml
Total cells
Viability
Sample
Denisity ×106/ml
Total cells
(%)


ID
0.00E+00
0.00E+00
(%)
ID
3.60E+04
3.60E+04
50





















3_7
2.40E+04
2.40E+04
20


1_2
0.00E+00

0.00E+00
3_8
2.40E+04
2.40E+04
40


1_3
0.00E+00

0.00E+00
3_9
3.60E+04
3.60E+04
100


1_4
0.00E+00

0.00E+00
3_10
3.60E+04
3.60E+04
60


1_5
0.00E+00

0.00E+00
3_11
9.50E+04
9.50E+04
57.1


1_6
0.00E+00

0.00E+00
3_12
2.40E+04
2.40E+04
40


1_7
0.00E+00

0.00E+00
4_1
9.50E+04
9.50E+04
32


1_8
0.00E+00

0.00E+00
4_2
3.80E+05
3.80E+05
69.6


1_9
0.00E+00

0.00E+00
4_3
3.30E+05
3.30E+05
93.3


1_10
0.00E+00

0.00E+00
4_4
1.20E+05
1.20E+05
35.7


1_11
0.00E+00

0.00E+00
4_5
3.70E+05
3.70E+05
72.1


1_12
0.00E+00

0.00E+00
4_6
2.50E+05
2.50E+05
87.5


2_1
4.80E+04

4.80E+04






66.7



4_7
1.80E+05
1.80E+05
37.5


2_2
0.00E+00

0.00E+00
4_8
2.40E+05
2.40E+05
44.4


2_3
0.00E+00

0.00E+00
4_9
3.30E+05
3.30E+05
96.6


2_4
0.00E+00

0.00E+00
5_1
1.80E+05
1.80E+05
48.4


2_5
0.00E+00

0.00E+00
5_2
2.40E+05
2.40E+05
55.6


2_6
0.00E+00

0.00E+00
5_3
3.00E+05
3.00E+05
92.6


2_7
0.00E+00

0.00E+00
5_4
2.70E+05
2.70E+05
79.3


2_8
0.00E+00

0.00E+00
5_5
2.10E+05
2.10E+05
51.4


2_9
0.00E+00

0.00E+00
5_6
2.40E+04
2.40E+04
66.7


2_10
0.00E+00

0.00E+00
6_1
1.20E+04
1.20E+04
50






6_2
1.20E+04
1.20E+04
50


2_11
0.00E+00

0.00E+00
6_3
1.20E+04
1.20E+04



2_12
0.00E+00

0.00E+00
6_4
0.00E+00
0.00E+00



3_1
4.80E+04

4.80E+04
6_5
0.00E+00
0.00E+00
100


57.1



6_6
0.00E+00
0.00E+00



3_2
2.40E+04
2.40E+04

6_7
0.00E+00
0.00E+00



3_5
2.40E+04
2.40E+04
28.6
6_8






0.00E+00

0.00E+00









Among 59 samples, 30 samples had countable cells. 10 samples had total cell number above 1×105. Only 5 samples reached viability above 90%.


FSC/SSC Analysis by FACS


Among 59 samples, all the samples showed lots of cell debris by FSC/SSC. None of the samples were found to have the same distribution pattern as granulocytes, lymphocytes, monocytes and macrophages, suggesting that there were no visible granulocytes, lymphocytes, monocytes or macrophages in the tested samples (FIG. 1 to FIG. 9).



FIG. 7. FSC/SSC on FACS



FIG. 8. FSC/SSC on FACS



FIG. 9. FSC/SSC on FACS



FIG. 10. FSC/SSC on FACS



FIG. 11. FSC/SSC on FACS



FIG. 1Z FSC/SSC on FACS



FIG. 13. FSC/SSC on FACS



FIG. 14. FSC/SSC on FACS



FIG. 15. FSC/SSC on FACS


Comparison with human T/B cells by FACS


Human peripheral blood and test samples were stained side by side with the same antibodies. B and T cell populations were identified by FACS (FIG. 10 to FIG. 16). The data did not show a convincing population of T or B cells.



FIG. 16. Comparison with human T/B cells on FACS



FIG. 17. Comparison with human T/B cells on FACS



FIG. 18. Comparison with human T/B cells on FACS



FIG. 19. Comparison with human T/B cells on FACS



FIG. 20. Comparison with human T/B cells on FACS



FIG. 21. Comparison with human T/B cells on FACS



FIG. 22. Comparison with human T/B cells on FACS


Comparison unknown samples with granulocytes by FACS


In addition to staining of T and B lymphocytes, human peripheral blood and test samples were stained simultaneously with the same antibodies and granulocytes were further identified by FACS. No granulocytes were found in all the test samples (FIG. 17 to FIG. 24).



FIG. 23. Comparison with human granulocytes on FACS



FIG. 24. Comparison with human granulocytes on FACS



FIG. 25. Comparison with human granulocytes on FACS



FIG. 26. Comparison with human granulocytes on FACS



FIG. 27. Comparison with human granulocytes on FACS



FIG. 28. Comparison with human granulocytes on FACS



FIG. 29. Comparison with human granulocytes on FACS



FIG. 30A. Comparison with human granulocytes on FACS


Comparison unknown samples with NK cells by FACS


None of the samples were found to contain NK cells (FIG. 25).



FIG. 31. Comparison with human NK cells on FACS


Conclusion


The characterization of unknown samples was carried out by staining with different cell surface markers for distinct cell lineages. Normal human peripheral blood cells were used as controls.


Vi-CELL cell viability analysis showed that 30 samples out of 59 samples had cells. Among these, only 10 samples had total cell number above 1×105 and only 5 samples reached viability above 90% (Table 1).


FACS analysis indicated that the test samples may not contain any of the typical cells present in human peripheral blood.


Bioactivity Determination of Protein Samples in Glucose Uptake Assay


Outline

    • 1. Study protocol
    • 2. Data summary
    • 3. Results
    • 4. Conclusions


Study Protocol

    • 1. 3T3-L1 fibroblasts were cultured in DMEM containing 25 mM glucose and 10% bovine calf serum at 37□ with 5% CO2.
    • 2. 3T3-L1 fibroblasts were differentiated into adipocytes 2 days post confluent with the same DMEM medium containing 1 ug/ml insulin (Sigma-Aldrich), 1 uM dexamethasone (Sigma-Aldrich), and 0.5 mM isobutyl-1-methylxanthine (Sigma-Aldrich).
    • 3. Media were replaced with DMEM containing 10% FBS and 1 ug/ml insulin and culture 2 days.
    • 4. Seed 1×105 3T3-L1 adipocytes to 96 wells cell culture plate (DMEM, 10% FBS, 1% PS, 1 ug/ml insulin)
    • 5. Before experiments, adipocytes were incubated in serum free medium for starvation overnight.
    • 6. Dilute each sample 100 folds (2 ul primal solution to 198 ul buffer) in corresponding buffer. Add 2 ul of sample primal solution, 100 folds diluted solution and buffer to 198 ul KRPH buffer.
    • 7. 3T3-L1 adipocytes were washed with KRPH buffer (5 mM Na2HPO4, 20 mM HEPES, pH 7.4, 1 mM MgSO4.1 mM CaCl2, 136 mM NaCl, 47 mM KCl, and 1% BSA) three times, Add 90 ul/well KRPH containing human insulin and samples to assay plate, incubate for 30 min at 37□ and 5% CO2.
    • 8. Add 10 ul KRPH containing 0.25 uCi 1-[3H]-2-deoxyglucose/well and 50 umol/l 2-deoxyglucose and Incubate 10 min in 95% air/5% CO2 at 37□
    • 9. The transport was stopped by rinsing the cells with cold PBS containing 10 mM glucose for three times.
    • 10. The adipocytes were lysed in 50 ul 10% KOH for 5 min
    • 11. Then the aliquots were subjected to scintillation counting using TriCap.


Data Summary
















% Activation

% Activation














Sample ID
First
Second
Average
Sample ID
First
Second
Average

















AFOD 1 (0.1%)
62.1
87.3
74.7
KH 1 (1:100)
15.9
20.89
18.4


AFOD 1 (0.001%)
9.6
28.9
19.2
KH 1 (1:10000)
13.02
23.11
18.1


AFOD RAAS
30.4
36.03
33.2
KH 105 (1)
18.2
9.66
13.9


103 (0.05%)



(1:100)





AFOD RAAS
10.37
3.66
7.0
KH 105 (1)
1.73
1.7
1.7


103 (0.0005%)



(1:10000)





AFOD RAAS
42.4
62.66
52.5
KH 105 (2)
27.76
41.78
34.8


107 (0.05%)



(1:100)





AFOD RAAS
11.87
3.26
7.6
KH 105 (2)
2.07
11.88
7.0


107 (0.0005%)



(1:10000)





AFOD RAAS
23.16
37.37
30.3
AFOD RAAS
3.54
2.97
3.3


108 (0.05%)



101 (0.1%)





AFOD RAAS
4.15
11.49
7.8
AFOD RAAS
17.71
−13.19
2.3


108 (0.0005%)



101 (0.001%)





AFOD RAAS
53.8
72.19
63.0
AFOD RAAS
−6.23
0.27
-3.0


109 (0.1%)



121 (0.1%)





AFOD RAAS
13.02
4.44
8.7
AFOD RAAS
1.98
−1.35
0.3


109 (0.001%)



121 (0.001%)





AFOD RAAS
10.71
15.54
13.1
AFOD KH
44.33
50.07
47.2


110 (0.05%)



(0.1%)





AFOD RAAS
12.9
4.44
8.7
AFOD KH
20.68
11.07
15.9


110 (0.0005%)



(0.001%)





AFOD RAAS
15.21
22.98
19.1






120 (0.05%)









AFOD RAAS
11.41
8.75
10.1






120 (0.0005%)
















The results we obtained in two separated experiments are consistent. Sample AFOD 1, AFOD RAAS 107, AFOD RAAS 109 and AFOD KH show some potency in glucose uptake assay.



FIG. 31A—AFOD 1 results



FIG. 31B—AFOD RAAS 101 results



FIG. 31C—AFOD RAAS 103 results



FIG. 31D—AFOD RAAS 107 results



FIG. 31E—AFOD RAAS 108 results



FIG. 31F—AFOD RAAS 109 results



FIG. 31G—AFOD RAAS 110 results



FIG. 31H—AFOD RAAS 120 results



FIG. 31I—AFOD RAAS 121 results



FIG. 31J—AFOD KH results



FIG. 31K—Kieu Hoang AFCC KH1 results



FIG. 31L—KH 105 (1) results



FIG. 31K—KH 105 (2) results


Conclusions

    • 1. The results we obtained in two times are consistent.
    • 2. Sample AFOD 1, AFOD RAAS 107, AFOD RAAS 109 and AFOD KH show some potency in glucose uptake assay.


Bioactivity Determination of Protein Samples in Glucose Uptake Assay


Outline

    • 1. Study protocol
    • 2. Data summary for dose response assay
    • 3. Results for dose response assay


Study Protocol

    • 1. 3T3-L1 fibroblasts were cultured in DMEM containing 25 mM glucose and 10% bovine calf serum at 37□ with 5% CO2.
    • 2. 3T3-L1 fibroblasts were differentiated into adipocytes 2 days post confluent with the same DMEM medium containing 1 ug/ml insulin (Sigma-Aldrich), 1 uM dexamethasone (Sigma-Aldrich), and 0.5 mM isobutyl-1-methylxanthine (Sigma-Aldrich).
    • 3. Media were replaced with DMEM containing 10% FBS and 1 ug/ml insulin and culture 2 days.
    • 4. Seed 1×105 3T3-L1 adipocytes to 96 wells cell culture plate (DMEM, 10% FBS, 1% PS, 1 ug/ml insulin)
    • 5. Before experiments, adipocytes were incubated in serum free medium for starvation overnight.
    • 6. Dilute each sample 100 folds (2 ul primal solution to 198 ul buffer) in corresponding buffer. Add 2 ul of sample primal solution, 100 folds diluted solution and buffer to 198 ul KRPH buffer.
    • 7. 3T3-L1 adipocytes were washed with KRPH buffer (5 mM Na2HPO4, 20 mM HEPES, pH 7.4, 1 mM MgSO4.1 mM CaCl2, 136 mM NaCl, 47 mM KCl, and 1% BSA) three times, Add 90 ul/well KRPH containing human insulin and samples to assay plate, incubate for 30 min at 37□ and 5% CO2.
    • 8. Add 10 ul KRPH containing 0.25 uCi 1-[3H]-2-deoxyglucose/well and 50 umol/l 2-deoxyglucose and Incubate 10 min in 95% air/5% CO2 at 37□
    • 9. The transport was stopped by rinsing the cells with cold PBS containing 10 mM glucose for three times.
    • 10. The adipocytes were lysed in 50 ul 10% KOH for 5 min
    • 11. Then the aliquots were subjected to scintillation counting using TriCap.


Data Summary for Dose Response Assay















EC50











AFOD RAAS 107
AFOD KH
insulin (nM)
















N1
No fit
0.00596%
3.09



N2
No fit
0.00108%
10.16



N3
No fit
0.00087%
4.173










The difference of EC50 is out of 3 folds between N1 and N2, so we run the N3. The results N2 and N3 are consistent.


For the sample AFOD RAAS 107 we found floccules in the solution



FIG. 31N—Dose response of AFOD RAAS 107 and AFOD KH_N1



FIG. 31O—Dose response of AFOD RAAS 107 and AFOD KH_N2



FIG. 31P—Dose response of AFOD RAAS 107 and AFOD KH_N3


Bioactivity Determination of Protein Samples in Glucose Uptake Assay


Outline

    • 1. Study protocol
    • 2. Data summary for dose response assay
    • 3. Results for dose response assay
    • 4. Conclusions and plan for dose response assay


Study Protocol

    • 1. 3T3-L1 fibroblasts were cultured in DMEM containing 25 mM glucose and 10% bovine calf serum at 37□ with 5% CO2.
    • 2. 3T3-L1 fibroblasts were differentiated into adipocytes 2 days post confluent with the same DMEM medium containing 1 ug/ml insulin (Sigma-Aldrich), 1 uM dexamethasone (Sigma-Aldrich), and 0.5 mM isobutyl-1-methylxanthine (Sigma-Aldrich).
    • 3. Media were replaced with DMEM containing 10% FBS and 1 ug/ml insulin and culture 2 days.
    • 4. Seed 1×105 3T3-L1 adipocytes to 96 wells cell culture plate (DMEM, 10% FBS, 1% PS, 1 ug/ml insulin)
    • 5. Before experiments, adipocytes were incubated in serum free medium for starvation overnight.
    • 6. Dilute each sample 100 folds (2 ul primal solution to 198 ul buffer) in corresponding buffer. Add 2 ul of sample primal solution, 100 folds diluted solution and buffer to 198 ul KRPH buffer.
    • 7. 3T3-L1 adipocytes were washed with KRPH buffer (5 mM Na2HPO4, 20 mM HEPES, pH 7.4, 1 mM MgSO4.1 mM CaCl2, 136 mM NaCl, 47 mM KCl, and 1% BSA) three times, Add 90 ul/well KRPH containing human insulin and samples to assay plate, incubate for 30 min at 37□ and 5% CO2.
    • 8. Add 10 ul KRPH containing 0.25 uCi 1-[3H]-2-deoxyglucose/well and 50 umol/l 2-deoxyglucose and Incubate 10 min in 95% air/5% CO2 at 37□
    • 9. The transport was stopped by rinsing the cells with cold PBS containing 10 mM glucose for three times.
    • 10. The adipocytes were lysed in 50 ul 10% KOH for 5 min
    • 11. Then the aliquots were subjected to scintillation counting using TriCap.


Data Summary for Dose Response Assay















EC50











AFOD 1
AFOD RAAS 109
insulin (nM)
















N1
0.0534%
0.0244%
7.085



N2
0.0205%
0.0090%
8.643











FIG. 31Q—Dose response of AFOD 1 and AFOD RAAS 109_N1



FIG. 31R—Dose response of AFOD 1 and AFOD RAAS 109_N1


Conclusions

    • 1. We have tested the sample AFOD 1 and AFOD RAAS 109 in dose response assay. The results we obtained in two times are consistent.


In Vivo Studies


The Study of APOAI Protein in Preventing Atherosclerosis and Related Cardiovascular Diseases


The current study was designed to investigate the human serum APOAI protein in preventing the atherosclerosis. New Zealand rabbits were adopted in this animal study and divided into 5 groups. They were high dose, medium dose and low dose of treatment, positive and vehicle control. The treatment groups were given APOAI via auricular vein once a week. Vehicle controls received normal saline via auricular vein once a week. Positive controls were given Liptor daily by p.o. with a dose of 0.45 mg/kg body weight. The body weight of animal was determined every week and whole blood was drawn every three weeks. The study duration was 19 weeks. At the end of study, all animals were sacrificed. The important organs like liver, heart, kidney, aorta, and arteria carotis were observed in gross and pathological sections. Lipid content was examined in liver and aorta. And liver index was also determined. Results showed that there was no significant change in body weight. The HDL-C was significantly high in all treatment groups when compared with vehicle control. Although the liver index was lower in treatment group, but there's no statistical difference found. The area of atherosclerosis was significant less in medium group when compared with vehicle control. The pathological examination showed that there was no calcification found in either vehicle control or treatment group. However there was one animal with calcification in positive control group. The pathological change of aorta was better in medium group when considering endothelium swelling, smooth muscle migrating and foam cell formation compared with vehicle control. But there is no significant improvement in low dose group. The cellular swelling and fat degeneration was better in the liver of medium than that of vehicle control. Although the cellular swelling was same in low dose group and vehicle control, but the fat degeneration was better in liver of low dose group than that of vehicle control. The lipid content in aorta was lower in treatment groups than that in vehicle control but there was no statistical significance. The lipid content in liver showed that TG in low and high dose group was significantly lower than that in vehicle control. The TC, TG and LDL-C in medium group were significantly lower than those in vehicle control.


Purpose of the Experiments:


To investigate the human serum APOAI in preventing atherosclerosis and related cardiovascular diseases and provide experimental basis for clinical application.


Methods and Materials


1, Tested Reagent

    • Product name: human Apolipoprotein AI, injection
    • Produced By: Shanghai RAAS Blood Products Co. Ltd.
    • Lot number:
    • Size: 50 mg/mL
    • Appearance: colorless liquid
    • Positive control: Liptor


2. Animal


Strain: New Zealand white rabbit


Vendor: Shanghai JieSiJie Laboratory Animal Co., Ltd


Qualification number:


Sex: male


Body weight: 1.8-2.0 kg


3 High Fat Diet Recipe


1% cholesterol+99% normal diet, provide by Shanghai SiLaiKe Laboratory Animal Center


4 Experimental Design


4.1 Model


Male New Zealand white rabbits were used in this study. The body weight was between 1.8-2.0 kg. The animals were quarantined for 5-10 days with normal diet before study. Blood samples were taken 12 hour after fasting before study to determine the blood lipid parameters.


4.2 Group


Animals were randomly divided into 5 groups including vehicle control, high dose, medium dose, low dose and positive control group. Ten to 14 rabbits were in one group. Each rabbit was fed with 30 gram of high fat diet followed by 120 gram of normal diet with free access to water.


Housing condition: Ordinary Animal Lab with temperature of 24±2□ and humidity of 55%±10%.


4.3 Administration


First dose was given 1 week before high fat diet. The frequency of dosing was once a week. Dose was 80, 40, 20 mg/kg body weight respectively. Drug was given by intravenous injection via auricular vein with the volume of 5 mL.


Liptor was given by intragastric administration


5 Parameters Tested:


5.1 body weight: body weight of each rabbit was determined once a week.


5.2 blood lipid parameters: whole blood was drawn every three weeks Animals were subject to 12 hour fast before taking blood. Resulted blood samples were kept still for 2 hours and then spin with 4,000 rpm for 10 min. The upper layer of serum was then separated and examined for total cholesterol (TC), total triglyceride (TG), low density lipoprotein cholesterin (LDL-C), and high density lipoprotein cholesterin (HDL-C). Test reagents were purchased from Shanghai Rong Sheng Bio-pharmaceutical Co. Ltd.


5.3 Pathological examination


A: The atherosclerosis of aorta (plaque area %)


B: Liver index


C: Aorta, liver, heart, arteria carotis, kidney


Results


1 the Establishment of Animal Model


Animals were fed with high fed diet and treatment as described above. All blood lipid parameters significantly increased. There was no significant difference between vehicle control and treatment groups (data shown below). After 12 weeks of high fat diet, 1 animal in vehicle control or treatment group was sacrificed respectively. The liver of animal in vehicle control showed cream white in color and there was no atherosclerosis observed in aorta. There was no abnormal change in the liver and aorta of animal in treatment group. After 16 weeks of high fat diet, 1 animal of vehicle control was sacrificed and found about 20% of plaque on the inner surface of aortic arch Animal continued to be fed with high fat diet and treatment for 3 more weeks. After 19 weeks of high fat diet, all animals were sacrificed.


2 Animal Procedures and Tissue Sampling


All animals were anesthetized by 20% of ethyl carbamate and then sacrificed with air injection. Abdomen cavity was opened. Whole blood was taken from heart. Heart was harvested along with 7 cm of aorta. Then other organs like liver, kidney and arteria carotis were harvested.


Connective tissue was stripped from resulted organs or tissues followed by washing in normal saline for 3 times. Pictures were taken then.


Aorta was cut from aortic arch, opened longitudinally and taken picture. The aorta was dissected for 0.5 cm from aortic arch, split longitudinally and then kept in cryo-preservation tube for later lipid analysis. One piece of this sample was fixed in formalin for further pathological analysis.


The weight of liver was determined immediately. Two pieces of specimen were cut from hepatic lobe. One was kept in cryo-preservation tube for lipid analysis and another one was fixed in formalin for further pathological analysis.


One piece of kidney sample was taken from renal pelvis and fixed in formalin for further pathological analysis.


Arteria carotis was dissected, cleaned and fixed in Formalin for further pathological examination.


The Formalin solution was replaced by fresh one about 4 hours and sent to pathological department for pathological section.


3 Results


3.1 Change of Body Weight


The body weight of each animal was determined before high fat diet and once a week thereafter. The change of body weight in each group was shown in table 1.









TABLE 1







The change of body weight in different groups











Group
Wk0
Wk 19
Increase
Increase


(animal number)
(kg)
(kg)
(kg)
(%)














Vehicle (n = 9)
1.94 ± 0.231
3.23 ± 0.284
1.29 ± 0.361
66.5%


High dose (n = 8)
1.68 ± 0.078
3.49 ± 0.221
1.81 ± 0.209
107.1%


Medium dose
1.8 ± 0.22
2.99 ± 0.52 
1.18 ± 0.286
65.5%


(n = 9)


Low dose (n = 12)
 2.1 ± 0.174
3.19 ± 0.278
1.09 ± 0.529
51.9%









3.2 Plasma Lipid Parameters


Animals were fast for 12 hours before taking blood samples via auricular vein. Resulted blood samples were kept still for 2 hours. The upper layer of serum was then separated and examined for total cholesterol (TC), total triglyceride (TG), low density lipoprotein cholesterin (LDL-C), and high density lipoprotein cholesterin (HDL-C). Test reagents were purchased from Shanghai Rong Sheng Bio-pharmaceutical Co. Ltd.









TABLE 2







Change of total triglyceride (TG)











Group






(animal
Wk 0
Wk 19
Increase
Increase


number)
(mmol/L)
(mmol/L)
(mmol/L)
(%)





Vehicle
0.823 ± 0.294
1.864 ± 0.871
1.041 ± 0.933
126.5%


(n = 9)


Medium
0.656 ± 0.191
2.144 ± 1.043
1.488 ± 0.988
226.8%


dose


(n = 9)


Low dose
0.786 ± 0.229
1.267 ± 0.772
0.482 ± 0.839
 61.3%


(n = 12)
















TABLE 3







Change of total cholesterol (TC)











Group
Wk0
Wk 19
Increase
Increase


(animal number)
(mmol/L)
(mmol/L)
(mmol/L)
(%)





Control(n = 9)
1.15 ± 0.23
8.049 ± 2.99
6.896 ± 3.03
598.3%


High dose (n = 8)
1.59 ± 0.48
12.49 ± 2.81
10.90 ± 2.66
685.5%


Medium dose
1.77 ± 0.783
10.28 ± 5.82
8.505 ± 5.37
453.0%


(n = 9)


Low dose (n = 12)
1.06 ± 0.27
 9.07 ± 4.92
 8.01 ± 4.87
755.6%
















TABLE 4







Change of high density lipoprotein cholesterin (HDL-C)












Group
Wk 0
Wk 19
Increase
Increase



(animal number)
(mmol/L)
(mmol/L)
(mmol/L)
(%)
Sig















Control(n = 9)
 0.94 ± 0.262
3.527 ± 2.007
2.588 ± 1.918
275.3%



High dose (n = 8)
1.183 ± 0.149
4.993 ± 2.018
 3.81 ± 2.025
322.1%
0.035*


Mediumdose(n = 9)
 0.67 ± 0.207
4.343 ± 2.439
3.674 ± 2.413
548.4%
0.02*


Low dose (n = 12)
0.705 ± 0.246
3.744 ± 2.14 
 3.04 ± 2.019
431.2%
0.028*





P < 0.05













TABLE 5







Change of ligh density lipoprotein cholesterin (LDL-C)











Group






(animal
Wk0
Wk 19
Increase
Increase


number)
(mmol/L)
(mmol/L)
(mmol/L)
(%)














Control
0.872 ± 0.386
5.826 ± 2.909
4.954 ± 2.953
568.1%


(n = 9)


High dose
 0.92 ± 0.324
 14.1 ± 4.188
13.18 ± 4.053
1432.6%


(n = 8)


Medium
 1.06 ± 0.298
6.357 ± 4.475
5.297 ± 4.373
499.7%


dose


(n = 9)


Low dose
0.826 ± 0.279
7.298 ± 4.60
6.472 ± 4.468
783.5%


(n = 12)
















TABLE 6







Liver index











Group






(animal
Body weight
Liver weight
Liver index


number)
(kg)
(g)
(%)
Sig














Control
3.083 ± 0.279
 123.08 ± 22.31
3.984 ± 0.579



(n = 9)


High dose
3.565 ± 0.205
 151.69 ± 18.49
4.257 ± 0.482
0.26


(n = 8)


Medium
3.009 ± 0.554
112.006 ± 25.79
3.708 ± 0.391
0.267


dose (n = 9)


Low dose
 3.3 ± 0.329
128.096 ± 20.43
3.886 ± 0.489
0.571


(n = 12)









3.3 Plaque Area of Aorta


The aorta was dissected and opened for 7.5 cm from aortic arch longitudinally. Pictures were taken and atherosclerosis changing was analyzed. The area of atherosclerosis was graded by clinical standard according to its area to whole area of dissected aorta, by which grade I was less than 25%, grade II was between 25% to 50%, grade III was between 50% to 75% and Grade IV was greater than 75%.









TABLE 7







atherosclerosis change in vehicle control group









Animal number
Plaque area/aorta area
Grade












5
8.62
I


6
16.67
I


7
37.5
II


9
39.47
II


11
1.67
I


12
10
I


17
92.86
IV


18
70.91
II


19
25.17
II





Grade I: 4 animals;


Grade II: 4 animals;


Grade III: 0 animal;


Grade IV: 1 animal













TABLE 8







atherosclerosis change in low dose group









Animal number
Plaque area/aorta area
Grade












31
10
I


32
26
II


36
1.92
I


37
76.79
III


38
11.11
I


39
2.88
I


40
6.67
I


41
2
I


42
92
IV


43
6.67
I


44
0.18
I


48
23.36
I





Grade I: 9 animals;


Grade II: 1 animal;


Grade III: 0 animal;


Grade IV: 2 animals.






Statistical Analysis of Low Dose Group: Mann-Whitney Test

























Grade 0

I





I

I



Level 1
I

I
I
I
I
I

I

I


Theoretic
1
2
3
4
5
6
7
8
9
10
11


level


Level
7
7
7
7
7
7
7
7
7
7
7


Level 0

7





7

7


Level 1
7

7
7
7
7
7

7

7


Grade 0
I

II

II
II
III


IV


Level 1

I

II



III
IV


Theoretic
12
13
14
15
16
17
18
19
20
21


level


Level
7
7
15.5
15.5
15.5
15.5
18.5
18.5
19.5
19.5


Level 0
7

15.5

15.5
15.5
18.5


19.5


Level 1

7

15.5



18.5
19.5





Level sum in Vehicle control: 112.8


Level sum in low dose group: 116.5


T0.05 = 71 T > T0.05 no statistical difference













TABLE 9







atherosclerosis change in medium dose group









Animal number
Plaque area/aorta area
Grade












21
36.53
II


22
1.69
I


23
18.75
I


25
19.17
I


27
11.67
I


28
1.82
I


29
61.67
II


30
1.6
I





Grade I: 6 animals;


Grade II: 2 animals;


Grade III: 0 animal;


Grade IV: 0 animal.






Statistical Analysis of Low Dose Group: Mann-Whitney Test























Grade 0

I


I
I

I



Level 2
I

I
I


I

I


Theoretic
1
2
3
4
5
6
7
8
9


level


Level
5.5
5.5
5.5
5.5
5.5
5.5
5.5
5.5
5.5


Level 0

5.5


5.5
5.5

5.5


Level 2
5.5

5.5
5.5


5.5

5.5


Grade 0

II

II
II
II
III
IV


Level 2
I

II


Theoretic
10
11
12
13
14
15
16
17


level


Level
5.5
13
13
13
13
13
16
17


Level 0

13

13
13
13
16
17


Level 2
5.5

13





Level sum in Vehicle control: 112.8


Level sum in low dose group: 46


T0.05 = 51 T < T0.05 statistical difference













TABLE 10







atherosclerosis change in high dose group









Animal number
Plaque area/aorta area
Grade












50
62.5
II


51
100
IV


52
56.88
II


53
40.13
II


54
100
IV


55
27.19
II


60
68.03
II


62
95.00
IV





Grade I: 0 animal;


Grade II: 5 animals;


Grade III: 0 animal;


Grade IV: 3 animals.






3.4 Pathological Examination


3.4.1 Aorta






















Endo-
Smooth



Animal
Plaque
Plaque

thelium
muscle
Foam


number
(gross)
(section)
calcification
swelling
migrating
cell















Vehicle control













5
+
+


+
+


6
+







7
++
+

+

+


9
++


+

+


11








12
+−







13








17
+++
++

++
+
++


18
+++
+

++

+


19
++
+

+









Medium dose group













21
++
+

+

+


22








23
+







25
+







27








28








29
+++


+




30













Low dose group













31








32
++
+++

+++
+
+++


37
++
+

++

+


38








39








40








41








42
+++
++

+++
++
++


43
+
+


+
+


44








48













High dose group













50
++




+


51
++




++


52
++
++


++
++


53
+







54
++




+


55
+







60
+++
++


++
+++


62
+++












Positive control













65








66
+
++


+
+


68
+

+


+


+2
++


+3
+









The pathological change was better in medium group when considering endothelium swelling, smooth muscle migrating and foam cell formation compared with vehicle control. But there is no significant improvement in low dose group


3.4.2 Liver Gross and Pathological Examination















Anima #
Observation (color, texture and size)
Swelling
Fatty change















Vehicle control










5
dark red, white in some area, soft, left > right
++
+


6
dark red, smooth, soft, left > right
+
+


7
Pink, soft, left > right
+
+


9
pink,, less soft,
+



11
Pink, smooth, soft
++
+


12
pink, rough
+++
+


13
dark red, some area showed pink, smooth,
+




soft


17
Pink, partial rough, less soft
+



18
Partial pink, smooth, soft




19
Partial pink, smooth, soft









Medium dose group










21
dark red, partial pink, soft, less smooth
+
+


22

++



23
dark red smooth, soft, left > right




25
dark red, partial pink, soft, smooth




27
dark red, partial pink, soft smooth
+



28

++



29
dark red, soft, smooth




30
dark red, soft, smooth









Low dose group










31
Partial pink, soft, less smooth
++



32
Pink, soft, less smooth
+
+


36
Partial yellow, rough, less soft
+++
+


37
Partial white, less soft, smooth




38





39
Pink-white color, rough, less smooth
++



40
Pink at Hepatic portal, soft, less smooth
+



41
dark red, soft, smooth




42
Partial pink, soft, smooth
+



43
dark red, soft, smooth




44
dark red, soft, smooth
+



48
dark red, soft, smooth









High dose group










50
Partial yellow, rough surface, less soft
++
++


51
Yellow, rough surface, less soft
++
++


52
dark red, partial pink, rough surface, soft




53
Pink, rough surface, less soft
+++



54
Pink, rough surface, soft
++



55
dark red, partial pink, rough surface, soft
+++



60
Partial yellow, rough surface, less soft
+



62
dark red, partial pink, rough surface, soft
++








Positive control group










65
Yellow, rough surface, less soft
++



66
Yellow-white color, rough surface, less soft
+++



68
Pink-white color at hepatic portal, dark red at





outskirt, rough texture, les soft


+2
Yellow color at hepatic portal, white at outskirt,
+++



rough texture, less soft


+3
Yellow, rough texture, less soft
+++









The cellular swelling and fat degeneration was better in the liver of medium than that of vehicle control. Although the cellular swelling was same in low dose group and vehicle control, but the fat degeneration was better in liver of low dose group than that of vehicle control.


3.4.3 Heart, Arteria Carotis and Kidney
















Heart/Coronary
Arteria carotis
kidney














Lipid

Lipid

Perirenal
Pathological


Animal number
infiltration
plaque
infiltration
plaque
adipose capsule
change
















5




Full, thick



6




Full, thin



7




Full, relatively








thick



9




Full, relatively








thick



11




Full, thin



12




Full, relatively








thick



13




Full, a little thick



17




Full, a little thick



18




Full, a little thick



19




Full, relatively








thick








Medium dose group













21




Full, thin



22








23




Spots, thin



25




Full, very thin



27




Full, very thin



29




Full, very thin



30




Full, very thin








Low dose group













31




Full, very thin



32




Full, very thin



36




Full, very thin



37




Full, thin



38




Full a little thick



39




Full a little thick



40




Full, relatively








thick



41




Full, a little thick



42




Full, relatively








thick



43








44




Full, very thin








High dose group













50




Full relatively








thick



51




Full relatively








thick



52




Full relatively








thin



53




Full, relatively








thin



54




Full, relatively








thick



55




Full, relatively








thin



60




Full, relatively








thin



62




Full, relatively








thin








Positive control group













65




Less full, thin



66




Full, thin



68




Full, thin



+2




Full, thin



+3




Less full, thin









There was no pathological change found in heart and kidney either in vehicle control or treatment groups. There was no atherosclerosis change found in Arteria carotis.


3.4.3 Lipid Content in Tissues


1) Lipid Content in Liver


















Control
Low dose
Middle
High




















TC
3.056 ± 0.775
 2.95 ± 0.809
2.214 ± 0.515
2.841 ± 0.298


TG
1.817 ± 0.446
1.369 ± 0.251
1.081 ± 0.31 
 1.3 ± 0.171


HDL-
0.712 ± 0.244
0.803 ± 0.236
0.815 ± 0.249
0.825 ± 0.129


C


LDL-
2.035 ± 0.328
1.857 ± 0.559
1.407 ± 0.418
2.302 ± 0.054


C









Statistics Analysis of Lipid Content in Liver

















Low dose
Medium
High





















TC
0.775
0.022
0.564



TG
0.022
0.011
0.009



HDL-C
0.81
0.74
0.684



LDL-C
0.436
0.011
0.989










The lipid content in liver showed that TG in low and high dose group was significantly lower than that in vehicle control. The TC, TG and LDL-C in medium group were significantly lower than those in vehicle control.


2) Lipid Content in Aorta


















Control
Low dose
Middle
High




















TC
0.331 ± 0.097
 0.28 ± 0.047
0.332 ± 0.135
 0.29 ± 0.098


TG
0.406 ± 0.178
0.337 ± 0.055
0.388 ± 0.124
0.402 ± 0.101


HDL-
0.065 ± 0.032
0.092 ± 0.066
0.128 ± 0.064
0.111 ± 0.057


C


LDL-
0.323 ± 0.116
0.254 ± 0.078
0.307 ± 0.043
0.318 ± 0.05 


C









Statistics Analysis of Lipid Content in Aorta

















Low dose
Medium
High





















TC
0.387
0.879
0.483



TG
0.341
0.80
0.952



HDL-C
0.416
0.065
0.171



LDL-C
0.138
0.73
0.912










The lipid content in aorta was lower in treatment groups than that in vehicle control but there was no statistical significance.


Summary:


This study was designed to investigate the prevention efficacy of APOAI in atherosclerosis. The test article was given along with high fat diet which caused no significant decrease in blood lipid parameters. However the treatment significantly increased the HDL-C level in all treated groups. There was no dose escalation effect found in three treatment groups upon anatomic, pathological and biochemistry examination. It has been showed that the atherosclerosis in medium dose group was significantly less than that in vehicle control. The pathological change was better in medium group when considering endothelium swelling, smooth muscle migrating and foam cell formation in aorta compared with vehicle control. But there is no significant improvement in low dose group. The cellular swelling and fat degeneration was better in the liver of medium than that of vehicle control. Although the cellular swelling was same in low dose group and vehicle control, but the fat degeneration was better in liver of low dose group than that of vehicle control. The lipid content in aorta was lower in treatment groups than that in vehicle control but there was no statistical significance. The lipid content in liver showed that TG in low and high dose group was significantly lower than that in vehicle control. The TC, TG and LDL-C in medium group were significantly lower than those in vehicle control.


Appendix 1: Pictures of Aorta



FIG. 32—Vehicle control



FIG. 33—Low dose group



FIG. 34—Medium dose group



FIG. 35—High dose group



FIG. 36E—Positive control (Liptor)



FIG. 36A—Liver fatty change of control and treated animals



FIG. 36B—Fat deposit on heart of control and treated animals



FIG. 36C—Atherosclerosis on control and treated animals



FIG. 36D—Atherosclerosis change on control and treated animals.


Experimental Design and Results of Pilot Scale


Pre-Clinical Animal Test of Apo-AI


For the Antiatherogenic and Cholesterol-Lowing Properties


Apolipoprotein A-I (APOAI) is the major protein component of high density lipoprotein (HDL) in human plasma. The protein promotes cholesterol efflux from tissues to the liver for excretion and also helps to clear cholesterol from arteries Human APOAI protein was purified from pooled normal human plasma via chromatography with 98% of purity. Rabbit model with atherosclerosis was established in order to examine the efficacy of the resulted APOAI protein. APOAI was given intravenously to rabbits with dose escalation. Plasma lipid concentration was determined at indicated time point and the change of fatty streak lesions and liver tissue were also examined Our results showed that there was a decrease in all plasma lipid concentrations like total cholesterol (TC), tri-gliceride (TG), low density lipoprotein-cholesterol (LDL-C), very low density lipoprotein-cholesterol (VLDL-C), high density lipoprotein-cholesterol (HDL-C) in all animals received APOAI when compared to animal without APOAI treatment. There was also a significant change in fatty liver appearance. Although APOAI didn't stop the progress of fatty streak lesions, but it inhibited the growth of fatty streak lesions by 38% and 29% in two cohorts of animals respectively. Meanwhile the lipid content in aorta decreased in APOAI treated animals as well. Our results show that administration of APOAI can decrease the plasma lipid concentrations and inhibit the progress of fatty streak lesions in rabbits.


1. Purpose of the Experiments:


1.1 To establish an animal model of atherosclerosis


1.2 To investigate the efficacy of Apo-AI for the suppression of fatty streak lesions.


1.3 To investigate a dose escalation of APOAI in treating fatty streak lesions


2. Methods and Materials


2.1 Animal Procedures


Male New Zealand white-ear or other strain healthy rabbits (2.0 kg body weight, 4 in each group) were adopted. The rabbits were fed with normal diet under regular lab conditions for 5-10 days. The rabbits were fasted for 12 hrs before the beginning of the experiments. Blood parameters were then tested as the normal level of plasma indicators.


A total of 52 rabbits were purchased at different time, four of them were used as normal control and fed with normal diet the whole time during the experiments. There rest of the animals was switched to high fat diet for 10-11 weeks. When animal developed obvious fatty streak lesions in blood vessels, animals were randomly divided into 4 groups as following (FIG. 1).

  • 1. Control group (without APOAI and Atorvastatin): n=4, animals were given normal diet after wk 10.
  • 2. APOAI group 1: n=7, Apo-AI was administered once a week and 100 mg/each from wk 11 to wk 14; 50 mg/each were administered twice a week from wk 14 to wk 18.
  • 3. APOAI group 2: n=7, Apo-AI was administered once a week and 100 mg/each from wk 11 to wk 14; 50 mg/each were administered twice a week from wk 14 to wk 18; 100 mg/each were administered once a week from wk 18 to wk 21.
  • 4. Atorvastatin group: n=4, Atorvastatin was given to animal once a week from wk 10 to wk 14.



FIG. 1. Schematic Display of Animal Procedure.


2.2 Determination of Plasma Lipid Concentrations


Plasma concentrations of total cholesterol (TC), tri-gliceride (TG), low density lipoprotein-cholesterol (LDL-C), very low density lipoprotein-cholesterol (VLDL-C), high density lipoprotein-cholesterol (HDL-C) were quantified by enzymatic assays. TC/HDL-C or (LDL-C+VLDL-C)/HDL-C ratios were then determined.


2.3 Morphometric Evaluation of Atherosclerotic Lesions


Animals were sacrificed under deep anesthesia with sodium pentobarbital 25 mg/kg IV. The entire aorta from the aortic valve to the iliac bifurcation was removed from each rabbit and opened longitudinally. The vessel was fixed with 10% buffered formaldehyde (pH 7.4). Atheromatous lesions were measured without staining Atheromatous lesions were manually traced in the photographs, and the percent area of the atheromatous lesions was calculated.


2.4 Determination of Lipid Content in Dissected Aorta


Sample of aorta from animals were homogenated. And the content of lipid was determined.


3. Results


3.1 Establishment of Animal Model


At week 4, one of the lab animals were sacrificed and showed limited amount of fatty streak lesions. At week 10 and week 11, five lab animals were sacrifices and aortas were dissected. Obvious fatty streak lesions can be observed on the inner surface of the aorta. Fat deposit can also be observed on the liver tissues. During the animal model construction, 7 animals died during the first 4 weeks of high-fat diet due to stomach symptoms. Between week 7-10, 6 more lab animals died because of high-fat. The mortality rate is 16.7%. These animals were also dissected and 90% of them the aorta tissue showed fatty streak lesions occupied 20% of the total area (see FIG. 2).



FIG. 37. The fatty streak lesions and liver change of animal fed with high fat diet and sacrificed on week 10. A) the lesions felt tender and hard as touched y hand. The fatty streak lesions of aorta were about 24.3% of the whole aorta. B) the surface of animal liver showed abnormal white colored spots which indicated a fat liver appearance


3.2 Successful Rate for Model Construction


During the animal model construction, 7 animals died during the first 4 weeks of high-fat diet due to stomach symptoms. Between week 7-10, 6 more lab animals died because of high-fat. The mortality rate is 16.7%. These lab animals were also dissected and 90% of them the aorta tissue showed fatty streak lesions occupied 20% of the total area (shown in FIG. 3).



FIG. 38. The plaque area change with normal diet after 10 weeks' high fat diet in control group. Control group were given normal diet after establishing fatty streak lesions in aortas (refer to methods and materials, animal procedure). A) the fatty streak lesions of aorta were about 45.3% of the whole aorta at Wk 14. B) and C) the fatty streak lesions of aorta were about 98.5% and 78.9.5% of the whole aorta in two animals at Wk 18.


3.3 Plasma Lipid Concentrations


1) APOA1 group1: The weight and plasma lipid concentrations of animals in APOA1 group 1 were determined at wk 0 (start of high fat diet), wk 10 (before APOA1 administration) and wk 18 (end of APOA1 administration).


























TC/



Weight
TG
TCH
VLDL-C
HDL-C
LDL-C
HDL-C







Wk 0
2.164
0.967
 1.152
 0.870
0.748
 0.282
1.938


Wk 10
2.7
5.191
36.153
14.996
8.261
21.157
6.560


Wk 18
2.79
1.17
 3.69
 1.09
1.46
 2.60
3.000









2) APOA1 group 2: The weight and plasma lipid concentrations of animals in APOA1 group 2 were determined at wk 0 (start of high fat diet), wk 10 (before APOA1 administration) and wk 21 (end of APOA1 administration).


























TC/



Weight
TG
TCH
VLDL-C
HDL-C
LDL-C
HDL-C







Wk 0
2.2
0.93
 1.430
 0.958
 0.432
 0.472
4.185


Wk 10
2.45
4.507
34.683
15.443
10.168
19.24
3.667


Wk 21
2.65
1.94
 3.322
 1.14
 1.17
 2.19
3.844









3) Atorvastatin group: The weight and plasma lipid concentrations of animals in Atorvastatin group were determined at wk 0 (start of high fat diet), wk 10 (before Atorvastatin administration) and wk 18 (end of Atorvastatin administration).


























TC/



Weight
TG
TCH
VLDL-C
HDL-C
LDL-C
HDL-C







Wk 0
2.25
0.450
 0.946
0.509
0.539
 0.437
1.844


Wk 10
2.85
9.122
20.339
9.710
8.404
10.911
4.511


Wk 18
3.1
0.474
 8.535
3.675
1.25
 4.86
6.811









4) control group: The weight and plasma lipid concentrations of animals in control group were determined at wk 0 (start of high fat diet), wk 10 (before normal) and wk 18 (before sacrifice).


























TC/



Weight
TG
TCH
VLDL-C
HDL-C
LDL-C
HDL-C







Wk 0
2.113
0.843
 1.444
0.885
0.684
 0.559
2.108


Wk 10
2.742
2.666
32.42
7.467
5.657
24.953
9.459


Wk 18
3.1
1.207
 5.277
1.961
0.759
 3.316
6.458









5) changes of plasma lipid concentrations in APOAI treated animals compared to control group animals (FIG. 4).


In APOAI group 1 and APOAI group 2, the main concentrations of plasma lipid decreased after 8 or 11 weeks of Apo-AI treatment meanwhile the decrease in control group was also observed. There is a significant decrease in VLDL-C and TC/HDL-C of APOAI group 1 when compared to control group (P<0.05). There is no significant change in the rest of values.



FIG. 39. The change of weight and plasma lipid concentrations were compared between APOAI treated and control animals. Data were obtained on wk 18, wk 21 and wk 18 from APOAI group 1 (gray solid column), APOAI group 2 (dark solid column) and control group (white column) respectively. X axis stands for the change of each value at the end of experiment compared to wk 10. Y axis stands for the parameters examined. * represents P<0.05


6) Changes of HDL-C in Animals


The change of HDL-C was determined between wk 0 and at the end of experiment (that is wk 18 in APOAI group 1 and control group and wk 21 in APOAI group 2) in APOAI treated and control animals Since HDL-C represents the lipid concentration carried by HDL which is formed by APOAI and phospholipids, so this result indicated that administration of Apo-AI could lower blood cholesterol through the formation of HDL.



















Wk 18 or wk
Concentration




Wk 0
21
increased
Fold increased




















APOAI Group 1
0.748
1.464
0.716
1.436


APOAI Group 2
0.432
1.423
0.992
3.078


Control group
0.684
0.759
0.074
0.102









3.4 Change of Liver Tissue


1) Pathological Examination of Liver Tissue


The gross change of liver surface is white colored spots observed at wk 10 of establishing the animal model. The surface of the liver feels harder than normal tissue. Histological analysis showed fatty liver change. The liver samples taken from the APOAI treated group showed less fatty change in gross specimen and the surface is not as hard as that at wk 10. The control group also showed relief in its gross chance. The probable reason is that the high cholesterol and atherosclerosis model is established in a relative short period of time, switch to normal diet also helped to alleviate the symptoms.


2) Liver Index


There is no difference in liver index between APOAI treated and control group.
















Weight (g)
Liver index




















APOAI Group 1
0.09
0.033



APOAI Group 2
0.117
0.044



Control group
0.111
0.036










3.5 Fatty Streak Lesions


The fatty streak lesions were examined in all animals at the end of the experiment. The area of the atherosclerosis was determined and then compared to wk 10 and wk 18 of control group respectively.




















Fatty








streak
Compare

Compare





lesions
to wk 10

to wk 18




Time
area
of control
Increase
of control
Decrease



point
(%)
group
%
group
%





















APOAI
Wk 18
43.84
19.03
77
−27.36
38.43


Group 1








APOAI
Wk 21
50.51
25.71
104
−20.69
29.05


Group 2








Control
Wk 18
71.20
46.39
187




group








Control
Wk 10
24.81






group









1) Gross examination of Fatty streak lesion: there was accumulation and swelling on aortic wall. The tissue was tender and hard when touched with hand. Dissection of the blood vessels showed fat deposit in the cross-section of the tissue. The fatty streak lesion decreases as the aorta desends. Compared with the control group, there were no bumps on the inner surface of aorta in APOAI treated group. The tissue feels soft.


2) Area measurement of the fatty streak lesion: the area of the fatty streak lesion increased 77% or 104% in APOAI treated animals and 187% in control group. Compared to control group at wk 10, the area of fatty streak lesion decreased by 38.43% and 29.05% in APOAI group 1 and APOAI group 2 respectively (shown in FIGS. 5, 6, and 7).



FIG. 40. Normal rabbit aorta without fatty streak lesion.



FIG. 41. the area of fatty streak lesion in aorta from APOAI group 1.



FIG. 42. the area of fatty streak lesion in aorta from APOAI group 2


3) Analysis of Lipid Content at Dissected Aorta


The lipid content in aorta was determined in all groups. The triglyceride content at dissected aorta of the APOAI group is significantly lower to that in control group (P<0.05).

















P value (compared to



lipid con. (umol/mg)
control group)


















APOAI Group 1 (n = 7)
 0.025 ± 0.0095
0.006


APOAI Group 2 (n = 4)
 0.0267 ± 0.0054
0.015


Atorvastatin group (n = 4)
0.0274 ± 0.006
0.046


Control group (n = 4)
0.0736 ± 0.014









4 Summary


The purpose of this preclinical animal is to test the dose and efficacy of APOAI in inhibiting the development of fatty streak lesion in rabbits.


Based on data from the experiments, it takes 4-5 weeks to establish a high cholesterol rabbit model need and 10 weeks to form atherosclerosis fatty streak lesion with high-fat diet (the average surface area of fatty streak lesion is 24% at week 10). 60% of animals will develop fatty streak lesion in aorta.


After intravenous infusion of human APOAI at various doses, the hypercholesterolemia and liver lesion improved dramatically, but APOAI inhibits rather than stop the progress of fatty streak lesion in aorta.


The experiment shows that the administration of APOAI to hypercholesterolemia lab animals reduces the surface area of fatty streak lesion in aorta and decreases the triglyceride content in the lesion tissue, thus, APOAI is a candidate of anti-atherogenic and anti-cholesterol medicine.


Report Title: Antiviral Efficacy of AFCC in an Influenza H1N1-Infected Mouse Model


Part 1 Pilot Study


Content


Objective


Infection with human influenza virus (IFV) causes respiratory tract illness in human and animals including mice. Mouse model infected Intranasally with IFV H1N1 is well recognized for anti-IFV compound screening. This study is designed to evaluate in vivo anti-IFV activity of a blood-derived product AFCC from RAAS in the mouse model and to identify appropriate dosages for the in vivo efficacy study.


Study Method


Study RAAS-20120216B was executed in the following steps:


1) Treat mice with RAAS blood product AFCC-KH.


1) Infect mice with IFV by intranasal inoculation.


2) Observe mice for 26 days.


3) Sacrifice mice in the end of the study.


Result Summary


In the pilot study all mice that were injected q.o.d. iv/ip in turn with AFCC survived through whole period of observation time (14 days) and their body weights varied in the normal range without significant loss. This indicates that the dosage and regimen of AFCC administration were well tolerated by the treated mice. Thereafter the 14-day-treated mice, together with an additional group of untreated mice as vehicle were challenged intranasally with IFN WSN. The AFCC treatment for 2 weeks before IFN WSN infection significantly decreased the mouse mortality and prolonged mouse survival time.


Report for RAAS-20120216B


I. Method


Animals:


Female BALB/c mice (6-8 weeks, 17-22 g) were divided into defined study groups after a visual examination and a 3 to 5-day acclimation upon arrival.


Solution Preparation:


1. Sodium Pentobarbital: Freshly dissolved in saline for injection at 8 mg/ml prior to using.


2. Test article: human plasma derived protein AFCC in sterile solutions for vein injection provided by the client.


Experimental Procedure:


IFV Infection and Test Article Administration:


1. From day 1 to day 14, AFCC KH 1 is intravenously and/or intraperitoneally administrated for 14 days.


2. On day 15, mice are anesthetized by intraperitoneal injection of sodium pentobarbital (80 mg/kg). Mice are inoculated with 5×10̂3 pfu of Influenza H1N1 A/WSN/33 via the intranasal route in SFM medium.


3. From day 1 through day 40 mice are observed two times a day. Mortality and body weight are recorded daily.


4. On day 40, the experiment is terminated by sacrificing survived mice.


II. Groups and Schedules:









TABLE 1







Action summary of Study WX-IFV02162012















IFV
AFCC,



Study Day
Date
Weighing
challenge
iv/ip
mouse sacrifice





Day 1
02162012






Day 2
02172012



Day 3
02182012





Day 4
02192012



Day 5
02202012





Day 6
02212012



Day 7
02222012





Day 8
02232012



Day 9
02242012





Day 10
02252012



Day 11
02262012





Day 12
02272012



Day 13
02282012





Day 14
02292012



Day 15
03012012





Day 16
03022012



Day 17
03032012



Day 18
03042012



Day 19
03052012



Day 20
03062012



Day 21
03072012



Day 22
03082012



Day 23
03092012



Day 24
03102012



Day 25
03112012



Day 26
03122012



Day 27
03132012



Day 28
03142012



Day 29
03152012



Day 30
03162012



Day 31
03172012



Day 32
03182012



Day 33
03192012



Day 34
03202012



Day 35
03212012



Day 36
03222012



Day 37
03232012



Day 38
03242012



Day 39
03252012



Day 40
03262012









✓ indicates that the action was taken.













TABLE 2







Experimental Design for the pilot experiment












AFCC-KH





Day
(ml/mouse)
animal number

H1N1 WSN














1
iv, 0.2
5




3
ip, 0.6
5


5
iv, 0.2
5


7
ip, 0.6
5


9
iv, 0.2
5


11
ip, 0.6
5


13
iv, 0.2
5


15
ip, 0.6*
5
5
in, 5 × 10{circumflex over ( )}3






pfu/mouse


17

5
4


19

5
4


21

5
4


23

5
4


25

5
4


27

5
4


29

5
4


31

5
4


33

5
4


35

5
4


37

5
4


39

5
4


40

5
4





*ip mice 4 h post H1N1 challenge.






III Adverse Events and Tolerability of Compounds:

  • 1. In the AFCC treatment group, one mouse died of severe face and neck damages on Mar. 3, 2012 (experimental day 17) due to serious fight among mice. This mouse was eliminated for final data analysis.


Results and Discussion


To serve the purpose to identify appropriate dosages for the efficacy study a small scale pilot experiment was carried out. In the pilot study all mice that were injected q.o.d. iv/ip in turn with AFCC survived through whole period of observation time (14 days) and their body weights varied in the normal range without significant loss. This indicates that the dosage and regimen of AFCC administration were well tolerated by the treated mice.


Thereafter the 14-day-treated mice, together with an additional group of untreated mice as vehicle were challenged intranasally with IFN WSN. The result indicated that all 5 mice in the H1N1-challenged vehicle control group died (FIG. 2). In comparison to the vehicle group, 50% mice pre-treated with AFCC survived to the end of experiment (FIG. 2) and their body weights started to recover within 9-24 days post-IFV WSN challenge after severe body weight loss of first a few days post WSN challenge (FIG. 3, FIG. 4). Treatment with AFCC significantly prolonged the survival time of the other 50% pre-treated mice by 18 days, compared with the vehicle group although the AFCC treatment didn't prevent their body weight loss (FIG. 4). In conclusion the AFCC treatment for 2 weeks before IFN WSN infection significantly decreased the mouse mortality and prolonged mouse survival. time.



FIG. 43. Body weight changes caused with AFCC treatment in mice









TABLE 3







Effect of AFCC on mean day to death of mice infected with H1N1


A/WSN/33













Mean day to death ±



Treatment
Survivor/total
S.D.







H1N1 + AFCC
2/4
23.8 ± 3.0**



H1N1 + Vehicle
0/5
 5.8 ± 0.8







**P < 0.01 compared to the H1N1 + vehicle control







FIG. 44. Efficacy of AFCC on H1N1 WSN-caused mouse death



FIG. 45. Body weight changes caused by AFCC in mice infected with H1N1 (WSN) influenza



FIG. 46. Body weight change caused with AFCC treatment in mice infected with H1N1 (WSN) influenza



FIG. 47. Body weight change caused with Vehicle treatment in mice infected with H1N1 (WSN) influenza


Report: Antiviral Efficacy of AFOD RAAS-2 in an Influenza H1N1-Infected Mouse Model


Summary of the Report


Objective


Infection with human influenza virus (IFV) causes respiratory tract illness in human and animals including mice. Mouse model intranasally infected with IFV H1N1 is well recognized for antiviral compound screening against IFV infection. This study is designed to evaluate the compound AFOD RAAS2 from RAAS for its in vivo anti-IFV efficacy.


Study Method


This study was performed in the following steps:

  • 1) Infect mice with IFV by intranasal inoculation.
  • 2) Treat the mice pre or post INF infection by iv/ip dosing of the AFOD RAAS2. 3) Daily record body weight of the mice.
  • 4) Sacrifice survived mice and inspect their major organs in the end of the study.


Result Summary


One-week preventive treatment with RAAS-2 fully protected H1N1-challenged mice from death and body weight loss although one-week therapeutic treatment with RAAS-2 led to one mouse, out of 5 mice survived in this group to the end of the experiment. In the H1N1-challenged vehicle control group all mice died and their body weights dramatically dropped by 20% to 30% within 4-7 days post-IFV H1N1 challenge. In contrast with the vehicle group, all mice treated therapeutically with oseltamivir survived although their body weights dropped and recovered to some extent. This indicated that the mouse model worked successfully in current study.


For Study Protocol: RAAS-20120428.v.2


I. Method


Animals:


Female BALB/c mice (6-8 weeks, 17-22 g) were divided into defined study groups after a visual examination and a 3 to 5-day acclimation upon arrival.


Solution Preparation:


1. Sodium Pentobarbital: Freshly dissolved in saline for injection at 7.5 mg/ml prior to using.


2. Test article: human plasma derived protein 29% AFOD RAAS2 in sterile solutions for vein injection provided by the client.


3. Vehicle: PBS


4. Oseltamivir phosphate (prodrug): aqueous solution in PBS, 0.1 mg/ml


Experimental Procedure:


IFV Infection and Test Article Administration:


1. From day −7 through day −1, 5 mice from group 4 are intravenously or intraperitoneally (iv/ip) administrated daily for 7 days.


2. On the day of Influenza administration, mice are anesthetized by intraperitoneal injection of sodium pentobarbital (80 mg/kg).


3. Anesthetized mice are inoculated with 5×10″3 pfu/mouse of Influenza H1N1 A/WSN/33 via the intranasal route in SFM medium.


4. Test article or vehicle is intravenously or intraperitoneally (iv/ip) administrated daily for 7 days. Oseltamivir (1 mg/kg) is orally given twice daily for 8 days. First dosing for oseltamivir or test article is executed 4 h pre H1N1 inoculation.


5. From day 1 through day 14 the infected mice are observed two times a day. Mortality and body weight are recorded daily.


6. On day 14, all living mice are sacrificed and dissected for the inspection of organ appearances.


II. Groups and Schedules:









TABLE 1







Action summary of the Study
















IFV
AFOD,
Oseltamivir, po















Study


challenge,
iv/ip,
10:00-
19:50-
mouse


Day
Date
Weighing
14:00-16:00
10:00-12:00
10:20
20:10
sacrifice





Day −7
May 22, 2012








Day −6
May 23, 2012








Day −5
May 24, 2012








Day −4
May 25, 2012








Day −3
May 26, 2012








Day −2
May 27, 2012








Day −1
May 28, 2012








Day 0
May 29, 2012








Day 1
May 30, 2012








Day 2
May 31, 2012








Day 3
Jun. 1, 2012








Day 4
Jun. 2, 2012








Day 5
Jun. 3, 2012








Day 6
Jun. 4, 2012








Day 7
Jun. 5, 2012








Day 8
Jun. 6, 2012








Day 9
Jun. 7, 2012








Day 10
Jun. 8, 2012








Day 11
Jun. 9, 2012








Day 12
Jun. 10, 2012








Day 12
Jun. 11, 2012








Day 13
Jun. 12, 2012








Day 14
Jun. 13, 2012











√ indicates that the action was taken.













TABLE 2







Experimental Design for the efficacy study





















H1N1






Vol
Treatment
1st treatment
(PFU/mo


Group
Mice
Compound
Dose
(ml/kg)
Schedule
time
use)





1
5
Vehicle#
0.2/0.4

Iv/ip, QD*
4 hrs
5 × 10{circumflex over ( )}3





ml/mouse


pre-infection



2
5
AFOD RAAS
0.2/0.4

Iv/ip, QD*
4 hrs
5 × 10{circumflex over ( )}3




2
ml/mouse


pre-infection



3
5
Oseltamivir
1 mg/kg
10
po, BID**
4 hrs
5 × 10{circumflex over ( )}3




phosphate



pre-infection



4
5
AFOD RAAS
0.2/0.4

Iv/ip, QD*
7 days
5 × 10{circumflex over ( )}3




2
ml/mouse


pre-infection





Iv/ip, QD*: Iv/ip means that iv injection is carried out with the volume indicated in “dose” column on day 0, 1, 2, 4 and ip injection is carried out on day 3; QD: daily (QD) for 4 days after H1N1 inoculation;


**BID, twice daily.


Vehicle#: PBS






III Adverse Events and Tolerability of Compounds:

  • 1. On day 5 post H1N1 infection, hematuria occurred in group 2 of AFOD RAAS2 treatment. We stopped AFOD RAAS2 medication on the sixth day post H1N1 infection.
  • 2. One mouse in the oseltamivir group died day 3 post H1N1 challenge. Its body dissection indicated that its esophagus was damaged probably due to harsh oral gavage. Therefore this mouse was ruled out from the experiment.


Result and Discussion


In the H1N1-challenged vehicle control group all 5 mice died and their body weights dramatically dropped by 20% to 30% within 4-8 days post-IFV H1N1 challenge (FIG. 1, FIG. 2, and Table 3). In contrast with the vehicle group, 4 out of 5 mice in the oseltamivir group survived to the end of experiment (FIG. 1, FIG. 2, and Table 3) although one mouse died accidentally of harsh oral gavage, which should be ruled out from the experiment as suggested early (see Part III, 2 in this report). The body weights in this group dropped by <15% days 5 to 8 post H1N1 challenge and recovered thereafter to some extent (FIG. 2). This indicated that the mouse model worked successfully in current study.


Impressively one-week preventive treatment with 0.2 ml/0.4 ml/mouse iv/ip QD of RAAS-2 totally protected H1N1-challenged mice from death and body weight loss till the end of this study (FIG. 1, FIG. 2 and Table 3). The protection of body weight loss by the preventive treatment of RAAS-2 is even better than that by oseltamivir treatment (FIG. 2). However the therapeutic treatment with 0.2 ml/0.4 ml iv/ip QD of RAAS-2 only protected one mouse out of 5 mice in the group from death and partial body weight loss of all 5 mice days 2 to 5 post H1N1 infection. Other 4 mice in this group died days 4 to 6 post H1N1 infection. In addition, some of the mice in the RAAS-2 therapeutic group (G2) had hematuria day 5 post H1N1 challenge and afterward, indicating that the dose used in this group was beyond mouse tolerance in H1N1 challenge status.


We don't understand why the RAAS-2 displayed such significant preventive efficacy on mouse death and body weight loss caused by H1N1 challenge. We have a number of suggestions to fully establish and understand this efficacy. First, we need to expand the efficacy experiment using a few more mice each group to confirm the data due to the small experiment scale (5 mice each group only) in the current study. In addition, a longer term study should be designed to fully know how long the preventive efficacy of the blood-derived product RAAS-2 could last. For example the mice should be challenged with H1N1 two weeks, three weeks, four weeks and even longer, respectively, post one week of preventive treatment of the RAAS-2. Some well designed mechanism studies should be carried out, such as in vivo H1N1 replication in infected mouse lungs in the preventive treatment and control groups, detection of immunological markers to reflect immune system activation and other biomarker assays post preventive treatment and H1N1 challenge. Finally a dose-dependent observation should be carried out for the RAAS-2 preventive treatment.



FIG. 48. Effect of AFOD RAAS2 on H1N1-caused mouse mortality









TABLE 3







Effect of AFOD RAAS2 or Oseltamivir on mean day to death (MDD) of


mice infected with H1N1 A/WSN/33













Mean





day to death ±


Treatment
Dose
Survivor/total
S.D.














H1N1 + Vehicle
0.2/0.4
ml/mouse
0/5
 4.8 ± 1.3


H1N1 + AFOD
1
mg/kg
1/5
 6.2 ± 4.4


RAAS2


H1N1 +
0.2/0.4
ml/mouse
4/4
>14 ± 0.0***


Oseltamivir


AFOD RAAS2 +
0.2/0.4
ml/mouse
5/5
>14 ± 0.0***


H1N1





***P < 0.001 compared to the H1N1 + vehicle control







FIG. 49. The average body weight change in mice infected with H1N1 influenza


Study Report


Efficacy of RAAS-8 in the HBV Mouse Hydrodynamic Injection Model


1 Introduction


Hydrodynamic injection (HDI) is an in vivo gene delivery technology. It refers to transiently transfect the mouse liver cells with a foreign gene via tail vein injection of a large volume saline containing plasmid within a few seconds. Taking the advantage of the liver-targeting manner of hydrodynamic injection, a single hydrodynamic injection of a replication-competent HBV DNA, could result in HBV replication in mouse liver shortly. This HBV hydrodynamic injection model on immunocompetent mice is a convenient and reproducible animal model for anti-HBV compound screening in vivo, which has been successfully established in WuXi ID department.


The purpose of this study is to evaluate in vivo anti-HBV efficacy of RASS 8 using the mouse hydrodynamic injection model.


2 Materials and Reagents


2.1. Animal:


Female BALB/c mice, age 6-8 weeks, between 18˜22 g.


2.2. Test article:


Vehicle: normal saline.


Entecavir (ETV): supplied as powder by custom-charactercustom-character dissolved in normal saline prior to dosing.


AFOD-RAAS 8 (RAAS 8): provided by RAAS, 25% (blood-derived proteins) solution.


2.3. Reagent:


HBV plasmid DNA:


pcDNA3.1/HBV, prepared with Qiagen EndoFree Plasmid Giga Kit; QIAamp 96 DNA Kit, Qiagen 51162; Universal PCR Master Mix, ABI 4324020; HBV DIG DNA probe, prepared by PCR DIG Probe Synthesis Kit, Roche 11636090910; DIG Wash and Block Buffer Set, Roche 11585762001; HBsAg ELISA kit, Kehua.


3 Experimental Procedure


3.1 Hydrodynamic Injection and Compound Administration

    • 3.1.1. From day −7 to day 0, all 5 mice in group 4 were administrated i.p./i.v. with test article daily for 8
      • days according to Table 2.
    • 3.1.2. On day 0, all groups of mice were hydrodynamicly injected via tail vein with pcDNA3.1/HBV plasmid DNA in a volume of normal saline equal to 8% of a mouse body weight. The plasmid DNA solution for injections was prepared one day before injection and then stored in 4E until injection.
    • 3.1.3. From day 0 to day 5, mice in groups 1-3 were weighed and treated with compounds or vehicle according to the regimen in Table 2. For groups 1 and 3, the first dosing was executed 4 hours pre HDI. For groups 2, the first dosing was executed 4 hours post HDI. For group 4, the last dosing was carried out 4 hours post HDI.
    • 3.1.4. All mice were submandibularly bled for plasma preparation according to the design in Table 1.
    • 3.1.5. All mice were sacrificed and dissected to obtain livers (two pieces of left lobe, one piece of middle lobe and one piece of right lobe) according to the regimen in table 1. Isolated livers were snap frozen in liquid nitrogen immediately upon collected.









TABLE 1







Experimental Design for the pilot experiment























plasmid DNA injection























Vol
Treatment

μg/
treatment




Group
Mice
CPD
Dose
(ml/kg)
Schedule
1st treatment time
mouse
schedule
bleeding
liver dissection





1
5
Vehicle#
See Tab 2

See Table 2
4 hrs pre-injection
20
tail vein HDI
days
day 7


2
5
RAAS 8
See Tab 2

See Table 2
4 hrs

of pcDNA
1, 3, 4,
day 7








post-injection

3.1 HBV,
5, 7



3
5
ETV
0.1 mg/kg
10
PO, QD*,
4 hrs


day 0, q.d.
day 5







days 0-4
pre-injection






4
5
RAAS 8
See Tab 2

See Table 2
last dosing,



day 7








4 hrs












post-injection





QD*: once a day;


Vehicle#: normal saline













TABLE 2







Schedule for Compound administration























group
Day
−7
−6
−5
−4
−3
−2
−1
0
1
2
3
4
5
6
7





4
am
0.2
0.4
0.2
0.4
0.2
0.4
0.4
HDI*,
No
No
No
No
No
No
No




ml,
ml
ml,
ml
ml
ml
ml
IV











IV
IP
IV
IP
IV
IP
IP











pm
No
No
No
No
No
No
No
0.5 ml
No
No
No
No
No
No
No











IP









2
am
No
No
No
No
No
No
No
HDI,
0.2
0.5
0.2
0.5
No
No
No











IV
ml
ml
ml
ml















IV
IP
IV
IP






pm
No
No
No
No
No
No
No
0.5 ml
0.3
No
0.3
No
No
No
No











IP
ml

ml
















IP

IP






1
am
No
No
No
No
No
No
No
0.5 ml
0.2
0.5
0.2
0.5
No
No
No











IP
ml
ml
ml
ml















IV
IP
IV
IP






pm
No
No
No
No
No
No
No
HDI,
0.3
No
0.3
No
No
No
No











IV
ml

ml
















IP

IP





HDI*: hydrodynamic injection






3.2 Sample Analysis


3.2.1 Detect HBV DNA Replication Level in Plasma

    • 3.2.1.1 Isolate DNA from 50 μl plasma using QIAamp 96 DNA Blood Kit. DNA was eluted with 120 μl ddH2O.
    • 3.2.1.2. Run qPCR for HBV DNA quantification.
      • a) Dilute HBV plasmid standard by 10-fold from 107 copies/μl to 10 copies/μl.
      • b) Prepare qPCR mix as shown below.














PCR reagents
Volume
Volume for 100 Reactions







DEPC Water
1.1 μl
 110 μl


Taqman Universal Master Mix
12.5 μl 
1250 μl


(2X)


HBV Primer Forward (50 μM)
0.2 μl
 20 μl


HBV Primer Reverse (50 μM)
0.2 μl
 20 μl


HBV Probe (5 μM)
  1 μl
 100 μl


Total
 15 μl
1500 μl













      • c) Add 15 μl/well PCR mix to 96-well optical reaction plates.

      • d) Add 10 μl of the diluted plasmid standard.

      • e) Transfer 10 μl of the extracted DNA to the other wells. Seal the plates with optical adhesive film. Mix and centrifuge.

      • f) Place the plates into qPCR machine and run the program according to the table blow.





















50° C.
 2 min
1 cycle


95° C.
10 min
1 cycle


95° C.
15 s
40 cycle 


60° C.
60 s









To eliminate the influence of input HBV plasmid, primers and probe targeting HBV sequence which detect newly replicated HBV DNA and input HBV plasmid DNA and targeting pcDNA3.1 plasmid backbone sequence which only detect the input plasmid DNA were used to do real-time PCR, respectively.


HBV DNA quantity=DNA determined by HBV primer-DNA determined by plasmid primer.


3.2.2 Detect HBsAg Level in Plasma


Dilute the plasma 500 fold;


Detect HBsAg level in 50 μl diluted plasma by using HBsAg ELISA kit.


3.2.3 Detect HBV Intermediate DNA Level in Livers


3.2.3.1 Liver DNA Isolation

    • a) Homogenize the liver tissue with Qiagen Tissue Lyser in 10 mM Tris.HCl, 10 mM EDTA, pH7.5.
    • b) Spin samples. Transfer the supernatant to a new tube containing equal volume of 2× proteinase K digestion buffer. Incubate at 50□ for 3 hours.
    • c) Extract with phenol: chloroform: Isoamyl alcohol.
    • d) Transfer the upper phase to new tubes, add RNase A and incubate at 37° C. for 30 min.
    • e) Extract with phenol: chloroform: Isoamyl alcohol.
    • f) Transfer the upper phase to new microfuge tubes, add 0.7-1 volume of isopropanol, add GlycoBlue Coprecipitant to 50 μg/mL, incubate at −20□ for 30 min.
    • g) Centrifuge (12000 g, 10 min) to precipitate DNA.
    • h) Wash the precipitate with 70% ethanol. Dissolve it in 25 μl ddH2O. Store DNA at −20□ until use.


3.2.3.2 qPCR for HBV DNA Quantification with Total Liver DNA.


The total liver DNA was diluted to 10 ng/μl. Use 10 μl diluted sample to run real-time PCR.


HBV DNA quantity=DNA determined by HBV primer-DNA determined by plasmid primer.


3.2.3.3 Southern Blot to Detect HBV Intermediate DNA Level in Livers.

    • a) Load 50 μg DNA for each sample. Run 1.2% agarose gel in 1×TAE.
    • b) After denaturing the gel with 0.25 M HCl at RT, neutralize the gel with neutralizing buffer.
    • c) Transfer the DNA form the gel to a pre-wet positively charged nylon membrane by upward capillary transfer overnight.
    • d) Remove the nylon membrane from the gel transfer assembly, UV cross-link the membrane (700 Microjoules/cm2), then wash it in 2×SSC for 5 min. Place the membrane at RT until dry.
    • e) Prehybridize membrane for 1 hour with hybridization buffer.
    • f) Pour off hybridization solution, and add the hybridization/pre-heated probe mixture, overnight
    • g) After hybridization and stringency washes, rinse membrane briefly in washing buffer.
    • h) Incubate the membrane in blocking solution, then in Antibody solution.
    • i) After wash in washing buffer, equilibrate in Detection buffer.
    • j) Place membrane with DNA side facing up on a development folder (or hybridization bag) and apply CDP-Star, until the membrane is evenly soaked. Immediately cover the membrane with the second sheet of the folder to spread the substrate evenly and without air bubbles over the membrane.
    • k) Squeeze out excess liquid and seal the edges of the development folder. Expose to X-ray film.
    • l) Expose to X-ray film at 15-25° C.


4 Results and Discussion


To investigate the effect of tested compounds on HBV replication in hydrodynamic model, the level of HBV DNA in plasma was analyzed by real-time PCR method (FIG. 1). Because the injected HBV plasmid DNA can also be detected by the primers targeting to HBV sequence, the primers and probe targeting the backbone sequence of pcDNA3.1 vector were designed and used for real-time PCR to eliminate the influence of residual plasmid in blood. The HBV quantity was calculated by the quantity determined by primers targeting HBV sequence subtracted by quantity determined by primers targeting the plasmid backbone sequence.


The results indicated that RASS 8 significantly inhibited the HBV replication by therapeutic or prophylactic treatment in a time-dependent manner post HDI. On day 1, RASS 8 therapeutic treatment showed ˜23% inhibition and RASS 8 prophylactic treatment showed ˜37% inhibition to HBV replication. On day 3 and day 4, the inhibition percentage to HBV replication by RASS 8 therapeutic, or prophylactic treatment was >99%, which is statistically significant. On day 5, RASS 8 therapeutic treatment caused ˜93% inhibition while its prophylactic treatment made almost 100% inhibition. The HBV level in both RAAS 8 prophylactic and therapeutic groups recovered a little on day 7 compared to the data on day 5. As a reference compound for the HBV HDI model, entecavir had significant inhibition to the HBV replication in the therapeutically-treated mice from day 3 post HDI to the end of experiment.



FIG. 50. Efficacy of therapeutic treatment or prophylactic treatment of RAAS 8 or ETV on in vivo HBV replication in HBV mouse HDI model. The total DNA was isolated from plasma by QIAamp 96 DNA Blood Kit. The HBV viral load in plasma during the course of the experiment was quantified by real-time PCR. Data is expressed as mean±SE. * P<0.05, ** P<0.01 by Student's t-test.


Secreted HBV surface proteins are also important index for HBV replication. HBsAg level in plasma was


detected by ELISA method (FIG. 2). Both RASS 8 therapeutic and prophylactic treatment had a significant inhibitory effect on HBsAg level in plasma within 5 days post HBV HDI while ETV didn't have significant inhibition to the HBsAg generation, suggesting that the in vivo effect of RAAS 8 on the in vivo HBV replication may be through a different mechanism from the entecavir.



FIG. 51. Effect of prophylactic treatment or therapeutic treatment of RAAS 8 or ETV on the HBsAg in mouse blood. The HBsAg level in plasma during the course of the experiment was determined by HBsAg ELISA kit. Data is expressed as mean±SE. * P<0.05, ** P<0.01 by Student's t-test.


Hepatitis B virus is a member of the hepadnavirus family, which replicates in livers and depends on liver specific factors. Thus, the existence of intermediate DNA in livers is a direct evidence for HBV replication in livers. To quantify the intermediate HBV DNA in livers, the total DNA was isolated from liver and HBV DNA level was determined by real-time PCR (FIG. 3). ETV, as a positive control, significantly decreased the HBV intermediate DNA in liver on day 5 Similar to ETV, RASS 8 prophylactic treatment had a significant inhibition on the replication of HBV intermediate DNA in livers on day 7. In comparison to the prophylactic treatment of RAAS 8, its therapeutic treatment caused significant but to less extent inhibition to the liver HBV replication by real time PCR (FIG. 3).


The HBV quantity determined by real-time PCR is total copy number of rcDNA, dsDNA and ssDNA. To separate and visualize rcDNA, dsDNA and ssDNA, southern blot was performed (FIG. 4). The major form of HBV replication intermediate DNA was ssDNA, which was consistent with report in literatures. Due to the limitation of DIG DNA probe sensitivity, we were not able to detect rcDNA or dsDNA. ssDNA decreased dramatically after RASS 8 prophylactic treatment or ETV treatment (FIG. 4), which confirms the result by real-time PCR (FIG. 3).



FIG. 52. Effect of prophylactic treatment or therapeutic treatment of RAAS 8 or ETV on the intermediate HBV replication in the mouse livers by qPCR. Mice in ETV group were sacrificed on day 5 and mice in the other three groups were sacrificed on day 7 post HDI. Liver DNA was isolated and subjected to real-time PCR to quantify the level of HBV replication intermediate DNA. Data is expressed as mean±SE. **P<0.01 by Student's t-test.



FIG. 53. Southern blot determination of intermediate HBV DNA in mouse livers. 50 μg total DNA each was subjected to southern blot. Lane 1 is 3.2 kb fragment of HBV plasmid (100 pg). Lane 2 and lane 19 are DNA makers. Lanes 3 to 18 are samples.



FIG. 54. The body weights of mice treated with vehicle or indicated compounds during the course of experiment


In summary, the RAAS 8 significantly inhibited HBV DNA replication by prophylactic or therapeutic treatment in the current study with the mouse HDI model. Impressively the prophylactic treatment with RAAS 8 displayed stronger inhibition to the HBV replication than its therapeutic treatment although we need more experiment to understand this phenomenon. In this study only 5 mice were used in each group. Thus the result may need to be confirmed by using more animals. In addition a well-designed mechanism study may be required to clarify how the RAAS 8 protein functions against HBV infection.


Efficacy of a Human Plasma Derived Protein AFOD RAAS 105 in Inhibition of the HBV Replication in the Mouse Hydrodynamic Injection Model


1.0 Introduction


Hydrodynamic injection (HDI) is an in vivo gene delivery technology. It refers to transiently transfect the mouse liver cells with a foreign gene via tail vein injection of a large volume saline containing plasmid within a few seconds. Taking the advantage of the liver-targeting manner of HDI, a single HDI of a replication-competent HBV DNA, could result in HBV replication in mouse liver. This HBV HDI model on immunocompetent mice is a convenient and reproducible animal model for anti-HBV compound screening in vivo. The purpose of this study is to evaluate in vivo anti-HBV efficacy of RAAS 105 using the mouse HDI model.


Materials and Reagents


Animal:


Female BALB/c mice (6-8 weeks old), between 18˜22 g, specific pathogen free, are purchased from SLAC (Shanghai Laboratory Animal Center of Chinese Academy of Sciences) and housed in an animal care facility in individually ventilated cages. Guidelines are followed for the care and use of animals as indicated by WuXi IACUC (Institutional Animal Care and Use Committee). The study was approved by WuXi IACUC (IACUC protocol 20120104-mouse). Mice are allowed to acclimate to the new environment for 3-5 days and are grouped according to the experimental set up.


Test Article:


Vehicle: normal saline.


Entecavir (ETV): supplied as powder by Rongda Pharm & Chem Co. Ltd., dissolved in normal saline prior to dosing.


AFOD RAAS 105: 25% protein concentrations (human plasma derived protein), provided by RAAS.


Reagents:


HBV Plasmid DNA:


pcDNA3.1/HBV, prepared with Qiagen EndoFree Plasmid Giga Kit, supplied as 850 ng/μl solution, diluted in normal saline.


QIAamp 96 DNA Kit, Qiagen 51162; Universal PCR Master Mix, ABI 4324020; HBV DIG DNA probe, prepared by PCR DIG Probe Synthesis Kit, Roche 11636090910; DIG Wash and Block Buffer Set, Roche 11585762001; HBsAg ELISA kit, Kehua.


Experimental Procedure


HDI and Compound Administration


From day −7 to day 0, mice in groups 6-7 are administrated IP/IV by turn with test article daily for 8 days according to Table 3.


On day 0, mice from group 1 to group 7 are hydrodynamically injected through tail vein with pcDNA3.1/HBV plasmid DNA in a volume of normal saline equal to 8% of a mouse body weight within 5 seconds. The plasmid DNA solution for injections is prepared before injection.


From day 0 to day 4, mice in groups 3-5 are administrated IP/IV by turn with test article daily for 5 days according to Table 2. From day 0 to day 4, mice in groups 1 and 2 are administrated PO with vehicle or ETV daily for 5 days according to Table 1. For groups 1-5, the first dosing is executed 4 hours post hydrodynamic injection. For groups 6-7, the last dosing is executed 4 hours pre hydrodynamic injection.


Mice from group 1 to group 7 are submandibularly bled for plasma preparation according to the design in Table 1. The blood samples will be collected in a tube containing heparin sodium and centrifuged at 7000×g and 4° C. for 10 min to obtain plasma samples.


On day 5, all mice in groups 1-7 are sacrificed and dissected to obtain livers (two pieces of left lobe, one piece of middle lobe and one piece of right lobe) according to the regimen in table 1. Livers are snap frozen in liquid nitrogen immediately upon collected.









TABLE 1







Grouping and Treatment














Test article
Plasmid DNA
























1st
injection

























or last
Dose






Mice/
Group

In vol

dosing
μg/
Treatment

sacrifice


Group
group
ID
Dose
(ml/kg)
schedule
time
mouse
schedule
Bleeding
time




















1
10
vehicle

10
PO, QD,
1st, 4 hrs
~20
HDI
0.2
Day 5







days 0-4
post-HDI

pcDNA
ml/mouse,



2

ETV
0.1
10
PO, QD,
1st, 4 hrs

3.1/HB
days






mg/kg

days 0-4
post-HDI

V,
1, 3, 4, 5



3

Therapeutic
0.15

See
1st, 4 hrs

day 0,






RAAS
ml/

Table 2
post-HDI

QD






105
mouse









4


0.25

See
1st, 4 hrs









ml/

Table 2
post-HDI









mouse









5


0.35

See
1st, 4 hrs









ml/

Table 2
post-HDI









mouse









6

Prophylactic
0.35

See
last, 4 hrs








vehicle
ml/

Table 3
pre-HDI









mouse









7

Prophylactic
0.35

See
last, 4 hrs








RAAS
ml/

Table 3
pre-HDI








105
mouse
















TABLE 2







Dosing schedule for therapeutic groups














group
compound
day
0
1
2
3
4





3
Therapeutic
am
HDI IV
0.15 ml
0.15 ml
0.15 ml
0.15 ml



RAAS 105


IP
IV
IP
IV




pm
0.15 ml









IP






4

am
HDI IV
0.25 ml
0.25 ml
0.25 ml
0.25 ml






IP
IV
IP
IV




pm
0.25 ml









IP






5

am
HDI IV
0.35 ml
0.35 ml
0.35ml
0.35 ml






IP
IV
IP
IV




pm
0.35 ml









IP
















TABLE 3







Dosing schedule for prophylactic groups

















Group
Compound
Day
−7
−6
−5
−4
−3
−2
−1
0





6
Prophylactic
am
0.35
0.35 ml
0.35
0.35 ml
0.35
0.35 ml
0.35
0.35



vehicle

ml IP
IV
ml IP
IV
ml IP
IV
ml IP
ml IP




pm







HDI












IV


7
Prophylactic
am
0.35
0.35 ml
0.35
0.35 ml
0.35
0.35 ml
0.35
0.35



RAAS

ml IP
IV
ml IP
IV
ml IP
IV
ml IP
ml IP



105
pm







HDI












IV









Sample Analysis


Detect HBV DNA Replication Level in Plasma


Isolate DNA from 50 μl plasma using QIAamp 96 DNA Blood Kit. DNA was eluted with 120 μl ddH2O.


Run qPCR for HBV DNA quantification.

    • a. Dilute HBV plasmid standard by 10-fold from 10′ copies/μl to 10 copies/μl.
    • b. Prepare qPCR mix as shown below.



















Volume for



PCR reagents
Volume
100 Reactions









DEPC Water
1.1 μl
110 μl



Taqman Universal Master
12.5 μl 
1250 μl 



Mix (2X)



HBV Primer Forward (50 μM)
0.2 μl
 20 μl



HBV Primer Reverse (50 μM)
0.2 μl
 20 μl



HBV Probe (5 μM)
  1 μl
100 μl



Total
 15 μl
1500 μl 












    • c. Add 15 μl/well PCR mix to 96-well optical reaction plates.

    • d. Add 10 μl of the diluted plasmid standard.

    • e. Transfer 10 μl of the extracted DNA to the other wells. Seal the plates with optical adhesive film. Mix and centrifuge.

    • f. Place the plates into qPCR machine and run the program according to the table blow.



















50° C.
 2 min
1 cycle


95° C.
10 min
1 cycle


95° C.
15 s
40 cycle 


60° C.
60 s









To eliminate the influence of input HBV plasmid, primers and probe targeting HBV sequence which detect newly replicated HBV DNA and input HBV plasmid DNA and targeting pcDNA3.1 plasmid backbone sequence which only detect the input plasmid DNA were used to do real-time PCR, respectively.


HBV DNA quantity=DNA determined by HBV primer-DNA determined by plasmid primer.


Detect HBsAg Level in Plasma

    • Dilute the plasma 200 fold;
    • Detect HBsAg level in 50 μl diluted plasma by using HBsAg ELISA kit.


Detect HBV Intermediate DNA Level in Livers


Liver DNA Isolation

    • a. Homogenize the liver tissue with Qiagen Tissue Lyser in 10 mM Tris.HCl, 10 mM EDTA, pH7.5.
    • b. Spin samples. Transfer the supernatant to a new tube containing equal volume of 2× proteinase K digestion buffer. Incubate at 50□ for 3 hours.
    • c. Extract with phenol: chloroform: Isoamyl alcohol.
    • d. Transfer the upper phase to new tubes, add RNase A and incubate at 37° C. for 30 min.
    • e. Extract with phenol: chloroform: Isoamyl alcohol.
    • f. Transfer the upper phase to new microfuge tubes, add 0.7-1 volume of isopropanol, add GlycoBlue Coprecipitant to 50 μg/mL, incubate at −20□ for 30 min.
    • g. Centrifuge (12000 g, 10 min) to precipitate DNA.
    • h. Wash the precipitate with 70% ethanol. Dissolve it in 25 μl ddH2O. Store DNA at −20□ until use.


qPCR for HBV DNA quantification with total liver DNA.


The total liver DNA was diluted to 10 ng/μl. Use 10 μl diluted sample to run real-time PCR.


HBV DNA quantity=DNA determined by HBV primer-DNA determined by plasmid primer.


Results and Discussion


To investigate the effect of the test compound on HBV replication in the mouse hydrodynamic injection model, the level of HBV DNA in plasma was analyzed by real-time PCR method (FIG. 1, FIG. 2). Because the injected HBV plasmid DNA can also be detected by the primers targeting to HBV sequence, the primers and probe targeting the backbone sequence of pcDNA3.1 vector were designed and used for real-time PCR to eliminate the influence of residual plasmid in blood. Thus the in vivo replicated HBV quantity was determined by subtracting DNA quantity amplified by primers targeting the plasmid backbone sequence from the DNA quantity amplified by primers targeting HBV sequence.


The results in FIG. 1 indicated that on day 3 and day 4 post HDI, the therapeutic treatment with 0.15, 0.25 or 0.35 ml RAAS 105/mouse inhibited the HBV DNA replication in plasma. The inhibition on day 4 was statistically significant compared to the vehicle group although the data on day 3 was not statistically significant due to the variation of HBV DNA replication. On day 5, after stopped dosing with the RAAS 105 the HBV DNA replication inhibition in plasma rebounded a little to a small extent.



FIG. 55 Efficacy of therapeutic treatment of RAAS 105 on in vivo HBV replication in plasma in mouse HDI model. The total DNA was isolated from mouse plasma obtained at time points as indicated in the figure by QIAamp 96 DNA Blood Kit. The HBV viral load in plasma during the course of the experiment was quantified by real-time PCR. Data is expressed as mean±SE. * P<0.05 by Student's t-test.


The results in FIG. 2 indicated that the prophylactic treatment with high dose (0.35 mL) of RAAS 105 didn't inhibit the HBV replication in plasma. In the other way around, the prophylactic treatment with 0.35 ml RAAS 105/mouse for a week pre HDI resulted in a significant enhancement of HBV level in mouse plasma on days 4 and 5 post HDI. Currently the mechanism of this effect remains to be figured out.



FIG. 56 Efficacy of prophylactic treatment of RAAS 105 on in vivo HBV replication in plasma in mouse HDI model. The total DNA was isolated from mouse plasma obtained at time points as indicated in the figure by QIAamp 96 DNA Blood Kit. The HBV viral load in plasma during the course of the experiment was quantified by real-time PCR. Data is expressed as mean±SE. ##P<0.01 by Student's t-test.


Secreted HBV surface proteins are also important index for HBV replication. HBsAg level in plasma was


detected by ELISA method (FIG. 3, FIG. 4).


The therapeutic treatment with 0.15, 0.25 or 0.35 ml RAAS 105/mouse had a very significant inhibitory effect on HBsAg level in plasma on days 1, 3, and 4. In contrast, oral 0.1 mg/kg ETV displayed similar pattern for the HBsAg generation to the vehicle group and ETV even caused significant increase on day 3, perhaps due to the data variation, suggesting that the in vivo effect of RAAS 105 on the in vivo HBV replication may be through a different mechanism from the entecavir.



FIG. 57. Effect of therapeutic treatment of RAAS 105 on the HBsAg in mouse plasma. The HBsAg level in plasma during the course of the experiment was determined by HBsAg ELISA kit. Data is expressed as mean±SE. ** P<0.01 by Student's t-test.


The prophylactic treatment with 0.35 ml RAAS 105/mouse caused decrease of HBsAg level in plasma on day 1 and day 3 although the inhibitory effect on day 3 was not statistically significant, perhaps due to the data variation (FIG. 4). On days 4 and 5, however, the prophylactic treatment with high dose of RAAS 105 increased the HBsAg level in plasma compared with the vehicle (FIG. 4), which is somewhat consistent with the in vivo effect of RAAS 105 on the HBV load in mouse plasma (FIG. 2).



FIG. 58. Effect of prophylactic treatment of RAAS 105 on the HBsAg in mouse plasma. The HBsAg level in plasma during the course of the experiment was determined by HBsAg ELISA kit. Data is expressed as mean±SE. ##P<0.01 by Student's t-test.


Hepatitis B virus is a member of the hepadnavirus family, which replicates in livers and depends on liver specific factors. Thus, the existence of intermediate DNA in livers is a direct evidence for HBV replication in livers. To quantify the intermediate HBV DNA in livers, the total DNA was isolated from liver and HBV DNA level was determined by real-time PCR (FIG. 5, FIG. 6).


As a reference compound for the HBV HDI model, entecavir, after 4-day oral treatment, significantly inhibited HBV replication in livers compared to the vehicle-treated group. However, RAAS 105 neither therapeutic nor prophylactic treatment showed statistically significant effect on HBV replication in livers (FIG. 5, FIG. 6), which is consistent with the in vivo effect of RAAS 105 on the HBV load in mouse plasma on day 5 (FIG. 1, FIG. 2).



FIG. 59. Effect of therapeutic treatment of RAAS 105 on the intermediate HBV replication in the mouse livers by qPCR. Liver DNA was isolated and subjected to real-time PCR to quantify the level of HBV replication intermediate DNA. Data is expressed as mean±SE. * P<0.05 by Student's t-test.



FIG. 60. Effect of prophylactic treatment of RAAS 105 on the intermediate HBV replication in the mouse livers by qPCR. Mouse livers were dissected in the end of experiment (day 5), liver DNA was isolated and subjected to real-time PCR to quantify the level of HBV intermediate DNA. Data is expressed as mean±SE.


Mouse body weight in all groups was monitored daily over the study period. During the time course, mice in endpoint assessment control groups and experimental groups did not differ significantly in terms of changes in body weight from baseline values (FIG. 7), indicating that no toxicity presented in mice treated with vehicle or test compound doses as indicated in the FIG.



FIG. 61. The body weights of mice in all groups treated with vehicle or indicated compounds during the course of experiment (n=10). * P<0.05 by Student's t-test.


Conclusion


The therapeutic treatment with RAAS 105 displayed significant inhibition on HBV DNA replication in plasma and a very significant inhibitory effect on HBsAg level in plasma. The prophylactic treatment with 0.35 ml/mouse RAAS 105 didn't have any inhibition on HBV DNA replication in plasma on days 1 and 3 post HDI, and instead increased HBV DNA level in mouse plasma on days 4 and 5 post HDI. In addition, the prophylactic treatment showed some inhibition on HBsAg level in plasma in early time points, such as day 1 post HDI.


Neither RAAS 105 therapeutic treatment nor high dose of prophylactic treatment showed any inhibition on HBV DNA replication in livers although therapeutic treatment with oral 0.1 mg/kg ETV significantly reduced the HBV DNA replication in livers on day 5 post HDI.


After the AFOD RAAS 8 and AFOD RAAS 105 have successfully inhibited the replication of Hepatitis B in mice immediately from day 1 to day 5 and the most impressive result is that AFOD RAAS 8 as well as AFOD RAAS 105 have completely eliminated the presence of Hepatitis B surface antigen in these mice. While the positive control drug ETV can only stop the replication of Hepatitis B virus. However ETV cannot kill the virus like AFOD RAAS 8 and AFOD RAAS 105. In order to know the population of cells to help these infected mice to recover a further study on the immune cell test was conducted on these three group of mice. Amazingly we discovered a new found cell like non B and non T cell.


Final Report


Characterization of Lymphoid Tissues and Peripheral Blood in HBV Infected BALB/c Mice Treated with RAAS 105


1 Executive Summary


This study was to investigate the effects of RAAS 105 on multiple cell lineages in lymphoid tissues and peripheral blood in HBV infected BALB/c mice. HBV infection and RAAS 105 treatment were performed by ID unit at Wuxi. At the termination, blood samples and lymphoid tissues were provided to us for analysis of various cell lineages by FACS.


Two independent experiments were performed. One experiment was to test therapeutic effects of RAAS 105 and the other experiment was to test prophylactic effects of RAAS 105.


Compared with the vehicle group, the differences observed in the animals treated with RAAS 105 therapeutically include: 1) percentages of T cells and B cells in peripheral blood, spleen and lymph nodes were decreased significantly; 2) CD62L was greatly downregulated on both CD4+ and CD8+ T cells in the spleen and lymph nodes; 3) granulocytes and monocytes/macrophages in peripheral blood and lymph nodes increased significantly; 4) the percentages of regulatory T cells (CD4+CD25+Foxp3+) in the spleen and lymph nodes were increased significantly.


However, prophylactic treatment with RAAS 105 led to somewhat different results. In the group treated with RAAS 105, T- and B-lymphocytes were also decreased. The percentages of monocytes and macrophages were increased albeit to a less degree.


These results suggested that administration of RAAS 105 had significant effects on the frequencies of immune cell lineages. However, it is not clear whether the effects were contributed by the high concentration of proteins in RAAS 105.


2 List of Abbreviations


















FACS
Flow Cytometry



mDC
Myeloid dendritic cell



pDC
Plasmacytoid dendritic cell










3 Materials and Methods


3.1 Materials


3.1.1 Reagents


FITC, Rat Anti-Mouse CD4, BD, Cat: 557307


PerCP-Cy5.5, Rat Anti-Mouse CD4, BD, Cat: 550954


FITC, Rat Anti-MouseCD3 molecular complex, BD, Cat: 561798


PerCP-Cy5.5, Rat Anti-Mouse CD3, BD, Cat: 560527


PerCP-Cy5.5, Rat Anti-Mouse CD8a, BD, Cat: 551162


PE, Rat Anti-MouseCD8a, BD, Cat: 553032


PE, Rat Anti-Mouse B220/CD45R, BD, Cat: 553089


APC, Rat Anti-Mouse CD11b, BD, Cat: 553312


APC, Ar Ham Anti-Mouse CD11c, BD, Cat: 550261


PE, Rat Anti-Mouse CD62L, BD, Cat: 553151


APC, Rat Anti-Mouse CD44, BD, Cat: 559250


PE, Rat Anti-Mouse Gr-1(Ly-6G and Ly-6C), BD, Cat: 553128


Alexa Fluor® 647, Rat Anti-Mouse Foxp3, BD, Cat: 560401


PerCP-Cy5.5, Rat Anti-Mouse CD19, BD, Cat: 551001


PE, Rat Anti-Mouse CD25, BD, Cat: 553075


ACK Lysing buffer, Invitrogen, Cat: A10492-01


RPMI 1640 medium, Invitrogen Gibco, Cat: 22400105


Dulbecco's Phosphate Buffered Saline, Thermo. Cat: SH30028.01B.


Fetal bovine serum, Invitrogen Gibco, Cat: 10099141


3.1.2 Materials


Cell strainer (70 μm), BD, Cat: 352350


BD Falcon tubes (12×75 mm, 5 ml), BD, Cat: 352054


3.1.3 Equipments


Vi-CELL Cell Viability Analyzer, Beckman Coulter, Cat: 731050


FACS Caliburflow cytometer, BD, Cat: 342975


3.2 Methods


Peripheral blood was collected through cardiac puncture. After removing red blood cells with lysis buffer followed by two rounds of washing using 1×PBS, mononuclear cells (monocytes, macrophages, dendritic cells, and lymphocytes) and granulocytes were obtained. Spleen and lymph nodes cell suspension were obtained after filtering through 70 μm cell strainer. Cell viability and number were analyzed by Vi-CELL Cell Viability Analyzer followed by cell surface staining Cells were centrifuged and resuspended in staining buffer (0.08% NaN3/PBS+1% FBS) containing appropriate fluorescent-conjugated antibodies. After 30 min incubation at 4° C. in the dark, cells were washed twice with 0.08% NaN3/PBS (200 μl per sample), and resuspended with 400 μl 0.08% NaN3/PBS in BD Falcon tubes (12×75 mm, 5 ml) followed by FACS analysis.


4 Data Analysis


FACS data were analyzed by flowjo software.


5 Study Summary


5.1 Study Initiation Date and Completion Date


To investigate the therapeutic and prophylactic effect of RAAS 105 on the immune system in mice infected with HBV, the study had divided into two parts. The individual part was performed on Sep. 11 and 19, 2012, respectively.


5.2 Study Purpose


The purpose of this study was to investigate the effect of RAAS 105 on cellular composition in lymphoid tissues and peripheral blood of HBV infected mice treated with RAAS 105.


5.3 Study Results


5.3.1 Effect of Therapeutic Treatment with RAAS 105


1) Mice Information


Total 10 female BALB/c mice including 2 naïve mice at the same age were transferred from Infectious Disease (ID) Group of WuxiApptec. The group and the regimen information were shown by Table 1.









TABLE 1







The experimental group and dosing regimen of the 1st part of the study
















1st or last



Groups
N
Group ID
Dose
dosing
Analysis















1
4
Therapeutic

1st, 4 hrs
Day 5




vehicle

post-HDI


3
4
Therapeutic
0.4 ml/mouse
1st, 4 hrs
Day 5




RAAS 105

post-HDI


11
2
Naive












2) Cell Populations in Peripheral Blood


After removing red blood cells, T cell lineages, B cells, DCs, granulocytes, and monocytes/macrophages in peripheral blood were analyzed by FACS analysis.


Total T cells and B cells were characterized by CD3 and CD19, respectively. HBV infection did not change the percents of CD3+ T cells compared with naïve mice. Therapeutic treatment of RAAS 105 reduced the percents of both CD3+ T cells and CD19+B cells significantly (FIG. 1). The representative FACS profiles from each group were illustrated in FIG. 2.



FIG. 62. Percents of T and B lymphocytes in peripheral blood. Total lymphocytes were gated. After therapeutic treated by RAAS 105, percents of T/B cells significantly decreased in peripheral blood. (by t test)



FIG. 63. Percent of T cells and B cells in peripheral blood. Total lymphocytes were gated.


Further analysis of the percents of CD4+ and CD8+ (non-CD4+) T cell lineages were performed gating on total CD3+ T cells. The results showed there were no differences in the percents of CD4+ and CD8+T cells among all the groups (FIG. 3). The representative FACS profiles from each group were illustrated in FIG. 4.



FIG. 64. Percents of CD4 and CD8 T cells in peripheral blood. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages.



FIG. 65. Percents of CD4 and CD8 T cells in peripheral blood. Total CD3 T cells were gated.


Percents of total CD11c+ dendritic cells (DC) and Gr-1+ granulocytes in peripheral blood were investigated. HBV infection reduced the percents of CD11c+DCs, a phenomenon which also be observed in human patients, whereas the percents of Gr-1+ granulocytes were not affected. Therapeutic treatment of RAAS 105 did not show any effect on CD11c+DCs, but increased the percents of Gr-1+ granulocytes significantly (FIG. 5). The representative FACS profiles from each group were illustrated in FIG. 6.



FIG. 66. Percents of Dendritic cells and Granulocytes in peripheral blood. Total live cells were gated. After therapeutic treatment, percents of granulocytes increased in peripheral blood (by T test)



FIG. 67. Percents of Granulocytes/Dendritic cells in peripheral blood. Total live cells were gated.


Percents of Monocytes were examined using surface marker CD11b. It increased significantly as same as Gr1+ granulocytes compared with the vehicle group (FIG. 7). The representative FACS profiles from each group were illustrated in FIG. 8.



FIG. 68. Percents of Monocytes in peripheral blood. Total live cells were gated. After treatment, percents of monocytes in peripheral blood significantly increased (t test)



FIG. 69. Percents of monocytes in peripheral blood. Total live cells were gated.


3) Cell Populations in Spleen


Cell lineages in spleen including T cell lineages (CD4+/CD8+ T cells, naïve T cells, memory T cells and regulatory T cells), B cells, mDCs, pDCs, granulocytes and macrophages were characterized by cell surface and intracellular markers.


Percents of total T cells and B cells in spleen were investigated. Therapeutic treatment of RAAS 105 reduced the percents of both CD3+ T cells and CD19+B cells significantly (FIG. 9). The representative FACS profiles from each group were illustrated in FIG. 10.



FIG. 70. Percents of T and B lymphocytes in spleen. Total lymphocytes were gated. After therapeutic treatment by RAAS 105, percents of T cells and B cells significantly decreased in spleen.



FIG. 71. Percents of T cells and B cells in spleen. Total lymphocytes were gated.


Further analysis of the percents of CD4+ (non-CD8+) and CD8+T cell lineages were performed gating on total CD3+ T cells. There were no differences in the percents of CD4+ and CD8+ T cells among all the groups (FIG. 11). The representative FACS profiles from each group were illustrated in FIG. 12.



FIG. 72. Percents of CD4 and CD8 T cells in spleen. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages.



FIG. 73. Percents of CD4 and CD8 T cells in spleen. Total CD3 T cells were gated.


Three T cell lineages, naïve T cells (CD44lowCD62Lhigh), central memory T cells (TCMs, CD44highCD62Lhigh) and Effector memory T cells (TEMs, CD44highCD62Llow), were characterized by surface markers CD44 and CD62L. Percents of these T cell lineages in CD4+ or CD8+ T cells were analyzed respectively. Both in CD4+ and CD8+ T cells, percents of naïve T cells and TCMs decreased and TEMs increased after the therapeutic treatment of RAAS 105, suggesting the compound may have effect to promote the transformation of T cells from naïve T cells to memory T cells in spleen (FIGS. 13 and 15). The representative FACS profiles from each group were illustrated in FIGS. 14 and 16.



FIG. 74. T cell subsets percentages in spleen. Total CD4 T cells were gated and T cell subsets were determined.



FIG. 75. CD4 T cell subsets percentages in spleen. Total CD4 T cells were gated and T cell subsets were determined.



FIG. 76. T cell subsets percentages in spleen. Total CD8 T cells were gated and T cell subsets were determined.



FIG. 77. CD8 T cell subsets percentages in spleen. Total CD8 T cells were gated and T cell subsets were determined.


Regulatory T cells (Tregs) were analyzed by cell surface staining of anti-CD4 and anti-CD25 antibodies followed by intracellular staining of anti-Foxp3 antibody. Percents of Tregs in spleen increased compared with the vehicle group (FIG. 17). The representative FACS profiles from each group were illustrated in FIG. 18.



FIG. 78. Percents of Foxp3 regulatory T cells in spleen. Foxp3 regulatory T cells were analyzed by intracellular staining After treatment, the percentage of T regulate cells is increased.



FIG. 79. Percents of regulatory T cells in spleen. Total CD4 T cells were gated.


Dendritic cells, including myeloid dendritic cells (mDC, B220CD11c+) and plasmacytoid dendritic cells (pDC, B220+CD11c+) in spleen were analyzed. No significant differences of mDCs and pDCs were observed among all groups (FIG. 19). The representative FACS profiles from each group were illustrated in FIG. 20.



FIG. 80. Percents of pDcs and mDcs in spleen. Total live cells were gated. There were no significant differences after compound treatment. (by t test)



FIG. 81. Percents of mDc and pDcs in spleen. Total live cells were gated.


CD11b+ macrophages and Gr-1+ granulocytes in spleen were analyzed. There were no significant alterations among all groups in the percents of these cell lineages in spleen, as shown in FIG. 21. The representative FACS profiles from each group were illustrated in FIG. 22.



FIG. 82. Percents of Macrophages and Granulocytes in Spleen. Total live cells were gated. There were no significant differences after compound treatment. (by t test)



FIG. 83. Percents of macrophages/Granulocytes in spleen. Total live cells were gated.


4) Cell Populations in Draining Lymph Nodes


Cell lineages in draining lymph nodes including T cell lineages (CD4+/CD8+ T cells, naïve T cells, memory T cells and regulatory T cells), DCs, granulocytes and macrophages were characterized by cell surface and intracellular markers.


Percents of total T cells in lymph nodes were analyzed. HBV infection did not affect the percents of CD3+ T cells but therapeutic treatment of RAAS 105 reduced it significantly compared with vehicle group (FIG. 23). The representative FACS profiles from each group were illustrated in FIG. 24.



FIG. 84. Percents of T cells in lymph nodes. Total lymphocytes were gated. After the treatment, the percentage of T cells in the lymph nodes were significantly decreased (t test)



FIG. 85. Percents of CD3 T cells in lymph nodes. Total lymphocytes were gated.


Further analysis of the percents of CD4+ and CD8+T cell lineages were performed gating on total CD3+ T cells. Percents of CD4+ T cells tended to decrease while CD8+ T cells tended to increase, suggesting that therapeutic treatment of RAAS 105 may have effect on the ratio of CD4+/CD8+ T cells in lymph nodes (FIG. 25). The representative FACS profiles from each group were illustrated in FIG. 26.



FIG. 86. Percents of CD4 and CD8 T cells in lymph nodes. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages. After therapeutic treatment, the percentage of CD4 T cells decreased. (by t test)



FIG. 87. Percents of CD4 and CD8 T cells in lymph nodes. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages.


Three T cell lineages, naïve T cells, TCMs and TEMs were characterized by surface markers CD44 and CD62L. Percents of these T cell lineages in CD4+ or CD8+ T cells were analyzed respectively. The results in lymph nodes were comparable to those in spleen. Both in CD4+ and CD8+ T cells, percents of naïve T cells and TCMs decreased and TEMs increased after the therapeutic treatment of RAAS 105, suggesting the compound also have effect to promote the transformation of T cells from naïve T cells to memory T cells in lymph nodes (FIGS. 27 and 29). The representative FACS profiles from each group were illustrated in FIGS. 28 and 30.



FIG. 88. CD4 T cell subsets percentages in lymph nodes. Total CD4 T cells were gated and T cell subsets were determined. No significant differences were found in all the groups compared to vehicle group.



FIG. 89. CD4 T cell subset percents in lymph nodes. Total CD4 T cells were gated and T cell subsets were determined.



FIG. 90. CD8 T cell subsets percents in lymph nodes. Total CD8 T cells were gated and T cell subsets were determined.



FIG. 91. CD8 T cell subsets percents in lymph nodes. Total CD8 T cells were gated and T cell subsets were determined.


Regulatory T cells (Tregs) were analyzed. Percents of Tregs in lymph node slightly increased without significant differences (FIG. 30B). The representative FACS profiles from each group were illustrated in FIG. 32.



FIG. 92. Percents of Foxp3 regulatory T cells in lymph nodes. There were no significant alterations after compound treatment



FIG. 93. Percents of regulatory T cells in lymph nodes. Total CD4 T cells were gated. One representative profile from each group is shown.


Total dendritic cells in lymph nodes were analyzed. Therapeutic treatment of RAAS 105 may reverse the reduction of DCs induced by HBV infection (FIG. 33). The representative FACS profiles from each group were illustrated in FIG. 34.



FIG. 94. Percents of DCs in lymph nodes. Total live cells were gated. After treatment, percents of DCs increased significantly (by t test)



FIG. 95. Percents of DCs in lymph nodes. Total live cells were gated.


CD11b+ macrophages and Gr-1+ granulocytes in lymph nodes were analyzed. Both percents of CD11b+ macrophages and Gr-1+ granulocytes increased significantly (FIG. 35). The representative FACS profiles from each group were illustrated in FIG. 36E.



FIG. 96. Percents of Macrophages and Granulocytes in lymph nodes. Total live cells were gated. Percents of macrophages and granulocytes significantly increased in lymph node. (by t test)



FIG. 97. Percents of Macrophages/Granulocytes in lymph nides. Total live cells were gated.


5.3.2 Effect of Prophylactic Treatment with RAAS 105


1) Mice Information


Total 14 female BALB/c mice including 2 naïve mice at the same age were transferred from Infectious Disease (ID) Group of Wuxi Apptec. The group and the regimen information were shown by Table 2.









TABLE 2







The experimental group and closing regimen of the 2nd part of the study
















1st or last



Groups
N
Group ID
Dose
dosing
Analysis















5
4
Prophylactic

last, 4 hrs
Day 5




vehicle#

pre-HDI


7
4
Prophylactic
0.4 ml/mouse
last, 4 hrs
Day 5




RAAS 105

pre-HDI


10
4
ETV
0.1 mg/kg
1st, 4 hrs
Day 5






pre-HDI


11
2
Naive












2) Cell Populations in Peripheral Blood


After removing red blood cells, T cell lineages, B cells, DCs, granulocytes, and monocytes/macrophages in peripheral blood were analyzed by FACS analysis.


Total T cells and B cells were characterized. Unlike therapeutic treatment, prophylactic treatment of RAAS 105 had no effect on percents of CD3+ T cells but reduced the percents of CD19+B cells although the statistical significance was not found (FIG. 37). The representative FACS profiles from each group were illustrated in FIG. 38.



FIG. 98. Percents of T and B lymphocytes in peripheral blood. Total lymphocytes were gated.



FIG. 99. Percents of T cells and B cells in peripheral blood. Total lymphocytes were gated.


Further analysis of the percents of CD4+ and CD8+ (non-CD4+) T cell lineages were performed gating on total CD3+ T cells. Unlike therapeutic treatment, prophylactic treatment reduced percents of CD4+ T cells and increased percents of CD8+ T cells, suggesting the potential effect of RAAS 105 to reduce the ratio of CD4+/CD8+ T cells in peripheral blood (FIG. 39). The representative FACS profiles from each group were illustrated in FIG. 40.



FIG. 100. Percents of CD4 and CD8 T cells in peripheral blood. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages. After prophylactic treated by RAAS 105, percents of CD4 T cells decreased while CD8 T cells increased (by t test)



FIG. 101. Percents of CD4 and CD8 T cells in peripheral blood. Total CD3 T cells were gated.


Results of total CD11c+ dendritic cells (DC) and Gr-1+ granulocytes in peripheral blood were also different from those in therapeutic treatment. Prophylactic treatment of RAAS 105 reversed the reduction of DCs induced by HBV infection, but had no significant effect on granulocytes in peripheral blood (FIG. 41). The representative FACS profiles from each group were illustrated in FIG. 42.



FIG. 102. Percents of Dendritic cells and Granulocytes in peripheral blood. Total live cells were gated. After prophylactic treated, percents of dendritic cells increased in peripheral blood.



FIG. 103. Percents of Granulocytes/Dendritic cells in peripheral blood. Total live cells were gated.


Percents of Monocytes were examined. There were no significant differences among all groups (FIG. 43). The representative FACS profiles from each group were illustrated in FIG. 44.



FIG. 104. Percents of Monocytes in peripheral blood. Total live cells were gated.



FIG. 105. Percents of monocytes in peripheral blood. Total live cells were gated.


3) Cell Populations in Spleen


Cell lineages in spleen including T cell lineages (CD4+/CD8+ T cells, naïve T cells, memory T cells and regulatory T cells), B cells, mDCs, pDCs, granulocytes and macrophages were characterized by cell surface and intracellular markers.


Percents of total T cells and B cells in spleen were investigated. Unlike therapeutic treatment, prophylactic treatment did not show effects on percents of CD3+ T cells and CD19+B cells (FIG. 45). The representative FACS profiles from each group were illustrated in FIG. 46.



FIG. 106. Percents of T and B lymphocytes in spleen. Total lymphocytes were gated.



FIG. 107. Percents of T and B cells in spleen. Total lymphocytes were gated.


Further analysis of the percents of CD4+ (non-CD8+) and CD8+T cell lineages were performed gating on total CD3+ T cells. Percents of CD4+ T cells slightly decreased and CD8+T cells slightly increased in spleen (FIG. 47). The representative FACS profiles from each group were illustrated in FIG. 48.



FIG. 108. Percents of CD4 and CD8 T cells in spleen. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages. After prophylactic treated by RAAS 105, the percentage of CD4 T cells slightly decreased while CD8 T cells slightly increased (by t test)



FIG. 109. Percents of CD4 and CD8 T cells in spleen. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages.


Naïve T cells, central memory T cells and Effector memory T cells were investigated. Percents of these T cell lineages in CD4+ or CD8+ T cells in spleen were analyzed respectively. Both in CD4+ and CD8+ T cells, percents of naïve T cells decreased and TEMs increased significantly after the prophylactic treatment of RAAS 105 (FIGS. 49 and 51). The representative FACS profiles from each group were illustrated in FIGS. 50 and 52.



FIG. 110. T cell subset percents in spleen. Total CD4 T cells were gated and T cell subsets were determined.



FIG. 111. T Cell subsets percents in spleen. Total CD4 T cells were gated and T cell subsets were determined.



FIG. 112. T cell subsets percents in spleen. Total CD8 T cells were gated and T cell subsets were determined.



FIG. 113. T cell subsets percents in spleen. Total CD8 T cells were gated and T cell subsets were determined.


Results of regulatory T cells (Tregs) were comparable with those in therapeutic treatment. Percents of Tregs in spleen increased compared with the vehicle group by prophylactic treatment of RAAS 105 (FIG. 53). The representative FACS profiles from each group were illustrated in FIG. 54.



FIG. 114. Percents of Foxp3 regulatory T cells in spleen. Foxp3 regulatory T cells were analyzed by intracellular staining



FIG. 115. Percents of regulatory T cells in spleen. Total CD4 T cells were gated.


Dendritic cells, including mDCs and pDCs in spleen were analyzed. No significant differences of mDCs and pDCs were observed among all groups after prophylactic treatment (FIG. 55). The representative FACS profiles from each group were illustrated in FIG. 56.



FIG. 116. Percents of pDCs and mDC in spleen. Total live cells were gated. There were no significant differences after compound treatment (by t test)



FIG. 117. Percents of mDCs and pDCs in spleen. Total live cells were gated.


CD11b+ macrophages and Gr-1+ granulocytes in spleen were analyzed. Percents of macrophages and granulocytes increased, but no statistical differences were observed, as shown in FIG. 57. The representative FACS profiles from each group were illustrated in FIG. 58.



FIG. 118. Percents of Macrophages and Granulocytes in spleen. Total live cells were gated. There were no significant differences after compound treatment. (by t test)



FIG. 119. Percents of macrophages/granulocytes in spleen. Total live cells were gated.


4) Cell Populations in Draining Lymph Nodes


Cell lineages in draining lymph nodes including T cell lineages (CD4+/CD8+ T cells, naïve T cells, memory T cells and regulatory T cells), DCs, granulocytes and macrophages were characterized by cell surface and intracellular markers.


Percents of total T cells in lymph nodes were analyzed. Similar with therapeutic treatment, HBV infection did not affect the percents of CD3+ T cells but prophylactic treatment of RAAS 105 reduced it significantly compared with vehicle group (FIG. 59). The representative FACS profiles from each group were illustrated in FIG. 60.



FIG. 120. Percents of T cells in lymph nodes. Total lymphocytes were gated. After the treatment, percents of T cells in the lymph nodes were significantly decreased. (t test)



FIG. 121. Percents of CD3 T cells in lymph nodes. Total lymphocytes were gated.


Further analysis of the percents of CD4+ and CD8+T cell lineages were performed gating on total CD3+ T cells. Percents of CD4+ T cells tended to decrease while CD8+ T cells tended to increase after prophylactic treatment, as was seen in therapeutic treatment (FIG. 61). The representative FACS profiles from each group were illustrated in FIG. 62.



FIG. 122. Percents of CD4 and CD8 T cells in lymph nodes. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages. After prophylactic treatment, percents of CD4 T cells decreased (by t test)



FIG. 123. Percents of CD4 and CD8 T cells in lymph nodes. Total CD3 T cells were gated and further analyzed for CD4/CD8 percentages.


Results of naïve T cells, central memory T cells and Effector memory T cells were totally difference with those in therapeutic treatment. Prophylactic treatment did not show significant effects on naïve T cells and TCMs, but increased percents of TEMs (FIGS. 63 and 65). The representative FACS profiles from each group were illustrated in FIGS. 64 and 66.



FIG. 124. T cell subsets percents in lymph nodes. Total CD4 cells were gated and T cell subsets were determined. No significant differences were found except effector memory T cells compared to vehicle group.



FIG. 125. T cell subsets percents in lymph nodes. Total CD4 T cells were gated and T cell subsets were determined.



FIG. 126. T cell subsets percents in lymph nodes. Total CD8 T cells were gated and T cell subsets were determined. No significant differences were found in all the groups compared to vehicle group.



FIG. 127. T cell subsets percents in lymph nodes. Total CD8 T cells were gated and T cell subsets were determined.


Regulatory T cells were analyzed. There were no significant differences among all groups (FIG. 67). The representative FACS profiles from each group were illustrated in FIG. 68.



FIG. 128. Percents of Foxp3 regulatory T cells in lymph nodes. Foxp3 regulatory T cells were analyzed by intracellular staining There were no significant alterations after compound treatment. (by t test)



FIG. 129. Percents of regulatory T cells in lymph nodes. Total CD4 T cells were gated.


Results of total dendritic cells in lymph nodes were similar with those in therapeutic treatment. Prophylactic treatment of RAAS 105 also increased the percents of DCs significantly compared with vehicle group (FIG. 69). The representative FACS profiles from each group were illustrated in FIG. 70.



FIG. 130. Percents of DCs in lymph nodes. Total live cells were gated. After the treatment, percents of the DCs increased significantly (by t test)



FIG. 131. Percents of DCs in lymph nodes. Total live cells were gated.


CD11b+ macrophages and Gr-1+ granulocytes in lymph nodes were analyzed. Both macrophages and granulocytes increased significantly (FIG. 71). The representative FACS profiles from each group were illustrated in FIG. 72.



FIG. 132. Percents of Macrophages and Granulocytes in lymph nodes. Total live cells were gated. After therapeutic treated by RAAS 105, percents of macrophages and granulocytes significantly increased. (by t test)



FIG. 133. Percents of Macrophages/Granulocytes in lymph nodes. Total live cells were gated.


7 Conclusions


The effects of RAAS 105 on different cell lineages in lymphoid tissues and peripheral blood in HBV infected mice were investigated by FACS analysis. T cell lineages (including CD4+/CD8+ T cells, naïve T cells, memory T cells and regulatory T cells), B cells, dendritic cells (including mDCs, pDCs), granulocytes and monocytes/macrophages were analyzed. RAAS 105 was administered in two different time schedules for therapeutic and prophylactic treatment.


Therapeutic treatment revealed some interesting findings. The animals treated with RAAS 105 exhibited alterations in multiple immune cells and various lineages compared with that in the vehicle group, including reduction of lymphocytes and increase of granulocytes and monocytes. Prophylactic treatment led to less dramatic alterations in the immune cells.


Final Report


Efficacy of Eight RAAS Test Articles on Adjuvant-Induced Arthritis (AIA) in Lewis Rats Executive Summary


















AIA
Adjuvant-induced arthritis



Dex
Dexamethasone



i.p.
intraperitoneal



HPMC
(Hydroxypropyl) methyl cellulose



p.o.
Per oral



b.i.d.
Twice a day



q.d.
Once a day



N/A
Not available










This study has evaluated the efficacy of eight RAAS test articles in the treatment of Adjuvant-Induced Arthritis (AIA) in Lewis rats. Male Lewis rats were immunized with Mycobacterium tuberculosis H37Ra to elicit AIA. On day 11 after immunization, when all the animals developed arthritis, the rats were administered with saline, Dexamethasone (Dex, positive control), and eight RAAS test articles for various durations, according to the sponsor's requests. The detailed treatment regimen is described below.


The data from this study showed that after the onset of the disease, the treatment with all eight RAAS products did not significantly affect the disease progression. After treatments, all the groups maintain 100% incidence rate. However, the group of animals treated with Dex had very mild disease, demonstrating dramatic inhibitory effects on the arthritic response. On the contrary, all the groups of rats treated with different RAAS products showed severe arthritis. The arthritic scores are similar among all the groups treated with RAAS products compared to that of vehicle group. Nevertheless, the measurement of paw swelling indicated that the paw volumes of the animals treated with AFCC KH and AFOD 101 decreased but the differences were not significant statistically at the most of the times compared to the vehicle group.


A. List of Abbreviations


B. Materials and Methods


a. Experimental Groups


The original study was planned to do the treatment for 10 days after disease onset. Table 1 was the group setting and dosing regimen.









TABLE 1







Grouping and Dosing Regimen for Day 11 to 20.

















Conc.
Dose vol.



Group
Test Article
N
Route
mg/ml
ml/rat
Frequency
















1
Normal
5
N/A
N/A
N/A
N/A


2
Vehicle (Saline)
8
i.p.
N/A
3
q.d.


3
Dex a
8
p.o.
0.02
5 ml/kg
q.d.


4
AFCC KH
8
i.p.
 18%
3
q.d.


5
AFOD KH
8
i.p.
 20%
3
q.d.


6
AFOD 101
8
i.p.
 20%
3
q.d.


7
AFOD 102
8
i.p.
  5%
3
q.d.


8
AFOD 103
8
i.p.
  5%
3
q.d.


9
AFOD 107
8
i.p.
  1%
3
q.d.


10
AFOD 108
8
i.p.
2.5%
3
q.d.


11
AFOD 1
8
i.p.
  5%
3
q.d.






a 0.5% HPMC/0.02% Tween 80 made with MilliQ water as vehicle







After the completion of 10-day treatment, the sponsor requested to continue the treatment for 15 more days and to increase dosing volumes (from 3 ml/rat/day q.d., to 2.5 ml/rat/day b.i.d.) as indicated in Table 2.









TABLE 2







Grouping and Dosing Regimen for Day 21 to 35

















Conc.
Dose vol.



Group
Test Article
N
Route
mg/ml
ml/rat
Frequency
















1
Normal
5
N/A
N/A
N/A
N/A


2
Vehicle (Saline)
8
i.p.
N/A
2.5
b.i.d.


3
Dex a
8
p.o.
0.02
5 ml/kg
q.d.


4
AFCC KH
8
i.p.
 18%
2.5
b.i.d.


5
AFOD KH
8
i.p.
 20%
2.5
b.i.d.


6
AFOD 101
8
i.p.
 20%
2.5
b.i.d.


7
AFOD 102
8
i.p.
  5%
2.5
b.i.d.


8
AFOD 103
8
i.p.
  5%
2.5
b.i.d.


9
AFOD 107
8
i.p.
1-2%
2.5
b.i.d.


10
AFOD 108
8
i.p.
2.5%
2.5
b.i.d.


11
AFOD 1
8
i.p.
  5%
2.5
b.i.d.






a 0.5% HPMC/0.02% Tween 80 made with MilliQ water as vehicle







After the completion of 25-day treatment, the sponsor requested additional 7 days treatment for five groups—Saline, Dex, AFCC KH, AFOD 101 and AFOD 102, as listed in Table 3. Please note that there was a two-day gap (Day 36 and 37) without treatment, before starting this 7-day period of treatment.









TABLE 3







Grouping and Dosing Regimen for Day 38 to Day 45:

















Conc.
Dose vol.



Group
Test Article
N
Route
mg/ml
ml/rat
Frequency





1
Normal
5
N/A
N/A
N/A
N/A


2
Vehicle (Saline)
8
i.p.
N/A
2.5
b.i.d.


3
Dex a
8
p.o.
0.02
5 ml/kg
q.d.


4
AFCC KH
8
i.p.
18%
2.5
b.i.d.


6
AFOD 101
8
i.p.
20%
2.5
b.i.d.


7
AFOD 102
8
i.p.
28%
2.5
b.i.d.






a 0.5% HPMC/0.02% Tween 80 made with MilliQ water as vehicle







b. Material


i. Reagents



Mycobacterium tuberculosis H37Ra: Difico (Detroit, Mich., USA), Cat: 231141


Paraffin oil: China National Medicine Corporation Ltd, Cat: 30139828


Hydroxypropyl Methyl Cellulose: Sigma, Cat: C5135


Tween 80: Sigma, Sigma-Aldrich. (St. Louis, Mo., USA), Cat: P-4780


Saline: Jiangsu Kang Bao Pharmaceutical Co., Ltd. Cat: H32026295


Dexamethasone (Dex): Xinyi Pharmaceutical Co., Ltd, H31020793


ii. Dose Formulation and Storage


All test articles were provided by the sponsor and storage at 4° C. before use.


iii. Equipment


Plethysmometer, Italy UGO BASJLE, Biological Research Apparatus 21025


iv. Animals and Testing Facility

  • Species: Rat
  • Strain: Lewis
  • Vendor: Beijing Vital Rivers Laboratories
  • Sex: Male
  • Body Weight when study started 180-200 g
  • Test Facility: WuXi AppTec Vivarium
  • Food: Free access to food (irradiated, Shanghai SLAC Laboratory Animal Co. Ltd., China)
  • Water: Free access to water (municipal tap water filtered by Mol Ultrapure Water System)
  • Total number of animals 85
  • Animal housing: 4 Rats/cage by treatment group
  • Identification Each rat was identified by ear tag and cage card
  • Adaptation: At least 7 days
  • Room: SPF Room
  • Room temperature: 20-26° C.
  • Room relative humidity: 40-70%
  • Light cycle: Fluorescent light for 12-hour light (6:00-18:00) and 12-hour dark (18:00-6:00)
  • Allocation to treatment groups: Randomization into 11 groups to achieve similar mean body weight, minimizing bias (See Table 1).


NOTE: All of the experimental procedures carried out within this study were approved by IACUC at WuXi AppTec.


v. Test Article Preparation


Dex: Dex was dissolved with 0.5% HPMC/0.02% Tween 80 into a final concentration of 0.02 mg/ml. The dosing volume is 5 ml/kg. Sonicate the suspension in an ice water bath for 10 minutes. Four 12 ml aliquots were stored in 4° C. refrigerator before use.


RAAS test article: Right before each dosing, a 50 ml of aliquot of each test article was prepared and warmed to room temperature.


vi. Immunization


Adjuvant Preparation

    • Weigh 100 mg of heat-killed Mycobacterium tuberculosis, ground suspended in Paraffin oil to final concentration of 10 mg/ml.
    • Sonicate the suspension in an ice water bath for 15 minutes.


Immunization Procedure

    • Shake the suspension of heat-killed/VI tuberculosis in Paraffin oil (to ensure even distribution of bacterial particles), then draw suspension into a 1 ml glass syringe attached to a 20-G needle. Replace the needle on the glass syringe with a 25-G needle. Re-suspend material in glass syringe by rolling between hands.
    • Anesthetize the rats with isoflurane, then inject 0.1 ml M. tuberculosis suspension subcutaneously in the left hind foot pad.
    • For the normal group (n=5), mineral oil was injected subcutaneously in the left hind foot pad.
    • 80 rats were randomly allocated to 10 groups (Table 1). The day of the injection was considered as day 0.


vii. Treatment

    • The treatment started at Day 11 as instructed by the sponsor. The incidence rates were 100%. The original planned treatment was 10 days (Day 11 to 20), with the dosage and dosing routes indicated in Table 1.
    • Per sponsor's request, all eight test articles were continued treated for additional 15 days (Day 21 to 35), with increased dosage. The detailed dosage and regimen was listed in Table 2.
    • The sponsor requested another additional 7 days (Day 38 to 45) of treatment for Saline, Dex, AFCC KH, AFOD 101 and AFOD 102 groups (Table 3). There was a two days gap (Day 36 and 37) before this segment.


viii. Endpoints

    • Body weight: Body weight of each animal was recorded every two days.
    • Paw swelling: The volume of right hind paw was pre-measured before immunization, and the right hind paw was measured once every two days, from Day 7 with plethysmometer.
    • Arthritic score: Start from Day 7 to 45, evaluate disease development by macroscopic inspection every two days. Assess walking ability, and screen for skin redness and swelling at the site of ankle and wrist joints and small interphalangeal joints. The left hind foot (the injected paw) will be excluded, the highest score is 12. See the criteria in table 4.









TABLE 4







Scoring system for evaluate arthritis severity








Score
Clinical signs





0
No erythema or swelling


1
Slight erythema and swelling in one of the toes or fingers


2
Erythema and swelling in more than one toe or finger or mild



swelling extending from the ankle to the mid-foot


3
Eryghema and severe swelling in the ankle or wrist


4
Complete erythema and swelling in toe or fingers and ankle or



wrist, and inability to bend the ankle or wrist









C. Data Analysis


Data were presented as mean±SEM. The body weight and paw volume were analyzed with two-way repeated ANOVA and the arthritis scores with Kruskal-Wallis test, by Graph Pad Prism 5. The statistical significance was noted when p<0.05.


D. Study Summary


a. Study Initiation Date and Completion Date


The study was initiated on Aug. 10, 2012, and ended on Sep. 24, 2012


b. Study Purpose


The goal of this project is to examine eight RAAS products in an autoimmune arthritis model, adjuvant induced arthritis (AIA) in rats. The study is to determine whether the products have therapeutic effects on AIA.


c. Study Results


The results of eight test articles are presented in two sections, according to their treatment durations: 1) 35 days treatment for AFOD KH, AFOD 103, AFOD 107, AFOD 108 and AFOD 1; 2) 45 days treatment for AFCC KH, AFOD 101 and AFOD 102.


i. Body Weight


Except Dex group, there was no significant difference for the body weight of all the treatment groups, when compared with saline group, in both 35 days and 45 days treatment sections (FIGS. 1 and 2). The reduction of body weight in Dex group was due to the side effect of Dex treatment.



FIG. 134. Effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108 and AFOD 1 on body weight (A) and body weight change (B) in AIA model till Day 35 (*p<0.05, **p<0.01, ***p<0.001, treatment groups v.s. saline group, two-way repeated or one-way ANOVA).



FIG. 135. Effects of AFCC KH, AFOD 101 and AFOD 102 on body weight (A) and body weight change (B) in AIA model till Day 45 (**p<0.01, ***p<0.001, treatment groups v.s. saline group, two-way repeated or one-way ANOVA).


ii. Paw Volume


The measurement of the paw volume indicated that the paw swelling was slightly reduced in the groups of animal treated with AFCC KH and AFOD 101. Statistical analysis showed that at the most of the times, the reduction was not significant statistically. However, the animals treated with AFCC KH showed significantly reduced paw volume on Day 22 and 35, compared to that of saline group (FIG. 4A). The animals treated with AFOD 101 showed significantly reduced paw swelling on day 22 (FIG. 4A). All other groups treated with the other six RAAS products didn't show any significant reduction in the paw swelling (FIGS. 3B & 4B).



FIG. 136. Effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108 and AFOD 1 on delta paw (right hind paw) volume (A) in AIA model till Day 35. AUC of delta paw volume curves were also presented (B). The delta paw volume of Dex group was significantly lower than saline group, from day 14 (***p<0.001, v.s. saline group, two-way repeated or one-way ANOVA).



FIG. 137. Effects of AFCC KH, AFOD 101 and AFOD 102 on delta paw (right hind paw) volume (A) in AIA model till Day 45. AUC of delta paw volume curves were also presented (B). The delta paw volume of Dex group was significantly lower than saline group, from day 14 (***p<0.001, v.s. saline group, two-way repeated or one-way ANOVA).


iii. Arthritic Score


The arthritic scores in all the groups treated with the eight test articles were similar to that of vehicle group (FIGS. 5 & 6). Dex treatment significantly inhibited the disease development (FIGS. 5 & 6).



FIG. 138. Effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108 and AFOD 1 on arthritic score in AIA model till day 35. The arthritic score of Dex group was significantly lower than saline group, from day 14 (p<0.01 for day 14, p<0.001 for day 16 to 35, Kruskal-Wallis test).



FIG. 139. Effects of AFCC KH, AFOD 101 and AFOD 102 on arthritic score in AIA model till Day 45. The arthritic score of Dex group was significantly lower than saline group, from day 14 (p<0.01 for day 14, p<0.001 for day 16 to 45, Kruskal-Wallis test).


iv. Incidence Rate


All the animals immunized with adjuvant developed arthritis at day 11 after immunization, when the treatment started, per sponsor's request. The incidence rates of all the groups remained 100% throughout the study period (FIGS. 7 & 8).



FIG. 140. Effects of AFOD KH, AFOD 103, AFOD 107, AFOD 108 and AFOD 1 on incidence rate in AIA model till day 35. The incidence rate reached 100%, 11 days after immunization. There was no chance of incidence rate afterward, for all the treatment



FIG. 141. Effects of AFCC KH, AFOD 101 and AFOD 102 on incidence rate in AIA model till day 45. The incidence rate reached 100%, 11 days after immunization. There was no change of incidence rate afterward. for all the treatment groups.


E. Conclusion

    • The treatment of eight test articles did not significantly affect the body weight changes compared to the saline group. The body weight of Dex group was lower than the other groups after treatment from Day 11.
    • Overall, the treatment of eight test articles did not inhibit paw swelling significantly after 25-day or 32-day treatments. However, the group of animals treated with AFCC KH and AFOD 101 showed reduced paw swelling. Statistical analysis showed significant difference for AFCC KH and AFOD 101, but only on Day 22, 35 and Day 22 respectively, by comparing to vehicle group.
    • Based on the arthritic scores, all the treatments did not show significant impacts on the disease progression. Dex treatment significantly inhibited the disease development.
    • The incidence rate reached 100% after day 11, before the treatment started, demonstrating successful setup of the model. During the treatment from day 11 to day 45, the incidence rates in all the groups remained 100%.


F. Reference


Debra M Meyer, Michael I Jesson, Xiong Li. Anti-inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor CP-690,550, in rat adjuvant-induced arthritis 2010.7.1


Study Title: Efficacy Study of AFOD RAAS 1 (APOA1) on Atherosclerosis Model in ApoE Mice


1. Abbreviations and Definitions


kg kilogram


g gram


Mg milligram


ng Nano gram


ml Milliliter


microliter


h hours


min minutes


Cpd Compound


BW Body Weight


BG Blood Glucose


FBG Fasting Blood Glucose


DOB Date of Birth


TC Total Cholesterol


TG Triglyceride


LDL Low Density Lipoprotein


HDL High Density Lipoprotein


FBW Fasting Blood Glucose


SD Standard Deviation


SE Standard error


i.p Intraperitoneal injection


PFA paraformaldehyde


2. Introduction


The study described in this report evaluated in vivo efficacy of RAAS antibody APOA I on atherosclerotic model in ApoE knockout mice.


3. Purpose


To evaluate the efficacy effect of RAAS antibody APO AI on plasma lipid profile, plaque lesion of inner aorta and related parameters in atherosclerotic model.


4. Materials

  • 4.1. Test article: RAAS Apo A I; Atorvastatin (reference compound)
  • 4.2. Animal: ApoE knock out (ko) mouse
    • Sex: male
    • Strain: C57BL/6
    • Vender: Beijing Vitol River
    • Age: 8 weeks (arrived on 23 Dec. 2011)
    • Number: 60
  • 4.3. Lipid profile test: Shanghai DaAn Medical Laboratory, Roche Modular automatic biochemistry analyzer
  • 4.4. Heparin Sodium Salt: TCI, H0393
  • 4.5. Capillary: 80 mm, 0.9-1.1 mm
  • 4.6. Ophthalmic Tweezers and scissors: 66 vision-Tech Co., LTD, Suzhou, China. Cat#53324A, 54264TM
  • 4.7. High Fat diet: TestDiet, Cat#58v8(35% kcal fat 1% chol)
  • 4.8. Glycerol Jelly Mounting Medium: Beyotime, Cat# C0187.
  • 4.9. Glucose test strips: ACCU-CHEK Performa: ROCHE (Lot#470396)
  • 4.10. Image analyse: Aperio ScanScope system; Image-Proplus 6.0 software; Aperio image scope version 11.0.2.725 software.
  • 4.11. Aorta staining: Oil Red O (Alfa Aesar) Isopropanol (Lab partner)


5. Experiment Method


5.1. Grouping Mice:


10 ApoE ko mice were fed with regular chow diet and used as negative control group. 50 ApoE ko mice were fed with high fat diet (35% kcal fat, 1% cholesterol) for 8 weeks, and then the plasma samples were collected for lipid profile measurement before the treatment. 50 ApoE ko mice were assigned into 5 groups based on the fasting overnight plasma TC and HDL level. The group information is shown in the table below.









TABLE 1







Information of groups













ApoE ko


Conc. Of



Group
mice
Diet
Solution
CPD
Formulation





Negative Control
n = 10
Normal diet





Vehicles (saline)
n = 10
High fat diet
0.9%





NaCL


ApoA1 High Dose: 0.1 ml i.p q.o d
n = 10
High fat diet

5% Protein


ApoA1 Mid Dose: 0.075 ml i.p q.o d
n = 10
High fat diet

5% Protein


ApoA1 Low Dose: 0.0.05 m i.p q.o
n = 10
High fat diet

5% Protein


Positive Control (Atorvastatin)
n = 10
High fat diet
0.5% CMC
2 mg/mL
20 mg + 10 ml


20 mg/kg (increased to 40 mg/kg)




0.5% CMC









5.2. Study Timeline:

  • 23 Dec. 2011: 60 ApoE mice arrived at chempartner and were housed in the animal facility in the building #3 for the acclimation.
  • 6 Jan. 2012: Measured the body weight for each mouse. 50 mice were fed with high fat diet and 10 mice were fed with normal chow diet.
  • 2 Mar. 2012: All mice were fasted over night and plasma samples (about 300 ul whole blood) were collected for lipid profile measurement before treatment with RAAS antibody.
  • 19 Mar. 2012 to 6 Apr. 2012: Group the mice based on the TC and HDL level and start the treatment with 3 doses of antibody APOA1 by i.p daily on the weekday (The first dose was administered by iv injection via the tail vein. The reference compound atorvastatin was administered by oral dosing every day.
  • 7 Apr. 2012 to 12 Apr. 2012: Stop dosing for 5 days. After 15 doses treatment with the antibody, several mice died in the treatment groups. The client asked for stopping treatment for a while.
  • 13 Apr. 2012-6 Jul. 2012: The treatment with antibody APOA1 was changed to i.p injection every two days (Monday, Wednesday, and Friday) per client's instruction.
  • 14 May 2012: All mice were fasted over night and plasma sample for each mouse (about 300 ul whole blood) was collected for lipid profile measurement after 8 weeks treatment.
  • 9 Jul. 2012: All mice were fasted over night and plasma sample for each mouse (about 300 ul whole blood) was collected for lipid profile measurement after 16 weeks treatment. Blood glucose was also measured for each mouse.
  • 9 Jul. 2012: The study was terminated after 16 weeks treatment. Measure BW, sacrificed each mouse, dissected the aorta, heart, liver and kidney and fixed them in 4% PFA.


5.3. Route of Compound Administration:


Antibody products were administrated by intraperitoneal injection every two days (Monday, Wednesday, and Friday). and the positive compound was administered by p.o every day.


5.4. Body Weight and Blood Glucose Measurement:


The body weight was weighed weekly during the period of treatment. The fasting overnight blood glucose was measured at the end of study by Roche glucometer.


5.5 24 h Food Intake Measurement:


24 hours food intake for each cage was measured weekly


5.6. Plasma Lipid Profile Measurement:


About 300 ul of blood sample was collected from the orbital vein for each mouse and centrifuged at 7000 rpm for 5 min at 4E and the plasma lipid profile was measured by Roche Modular automatic biochemistry analyzer in DaAn Medical Laboratory


5.7. Study Taken Down:


After RAAS antibody products treatment for 16 weeks, all mice were sacrificed. Measured body weight and collected blood sample for each mouse. Weighed liver weight and saved a tiny piece of liver into 4% paraformaldehyde (PFA) fixation solution for further analysis. At same time, take the photos with heart, lung, aortas and two kidneys.


5.8. Oil Red Staining Procedure:

    • 1. Sacrificed the mice and dissected the heart, aorta, and arteries under dissecting microscope.
    • 2. Briefly wash with PBS and fixed in 4% paraformaldehyde (PFA) overnight at 4□.
    • 3. Rinse with 60% isopropanol
    • 4. Stain with freshly prepared Oil Red O working solution 10 min.
      • 1). Oil red O stock stain: 0.5% powder in isopropanol
      • 2). Working solution: dilute with distilled water (3:2) and filter with membrane (0.22 um)
    • 5. Rinse with 60% isopropanol 10 second.
    • 6. Dispel the adherent bit fat outside of the aorta under the dissecting microscope.
    • 7. Cut the vascular wall gently and keep the integrated arteries using the micro scissors.
    • 8. Unfold the vascular inner wall with the cover slides and fix it by water sealing tablet.


5.9. Image Scanning and Analysis:


Scanning the glasses slides with the Aperio ScanScope system and analyze with the image proplus software to measure the area of atherosclerotic plaque lesion. The results were expressed as the percentage of the total aortic surface area covered by lesions. The operation procedure of software was briefly described as follow: Converted the sys version photos into JPG version, then calibrated it and subsequently selected the red regions and then calculate the total area automatically by image proplus software.


5.10. Clinic Observation:


The information of dead animals was shown in the table as below.


6. Data Analysis


The results were expressed as the Mean±SEM and statistically evaluated by student's t-test. Differences were considered statistically significant if the P value was <0.05 or <0.01.


7. Results


7.1. Effect of APOA 1 on Body Weight



FIG. 142. Body weight


The body weight in Apo E knockout mice fed with HFD significantly increased after 6 weeks treatment compared with the mice in negative control group that were fed with normal diet. There is no significant difference between the treatment groups and vehicle group.


7.2. Effect of HFD on Lipid Profile in ApoE Ko Mice



FIG. 143. Plasma lipid profile of ApoE mice fed with normal diet and high fat diet.


The lipid profile was measured in Apo E ko mice fed with high fat diet for 8 weeks. As shown above, plasma TC, TG, LDL as well as HDL in Apo E ko mice fed with high fat/high cholesterol for 8 weeks were significantly increased compared to Apo E KO mice fed with normal chow diet.


7.3. Effect of RAAS Antibody on Plasma Total Cholesterol (TC)



FIG. 144, Plasma TC



FIG. 145. Net change of plasma TC


As shown in the figure above, positive control atorvastatin can significantly lower total cholesterol level after 16 week treatment in ApoE ko mice but not reduce the TC net change.


7.4. The Effect of RAAS Antibody on Plasma Triglyceride (TG)



FIG. 146. Plasma TG


As shown in figure above, positive control atorvastatin and RAAS antibody had no effect on plasma TG level in Apo E ko mice fed with HFD after 16 weeks treatment.


7.5. The Effect of RAAS Antibody on High Density Lipoprotein (HDL)



FIG. 147. Plasma HDL



FIG. 148. Plasma HDL net change


As shown in figure above, positive control atorvastatin can significantly lower high density lipoprotein in Apo E ko mice fed with HFD after 16 week treatment and RAAS antibody had a mild trend to decrease the HDL level in ApoE ko mice after 16 weeks treatment.


7.6. The Effect of RAAS Antibody on Low Density Lipoprotein (LDL)



FIG. 149. Plasma LDL level



FIG. 150. Plasma HDL net change


As shown in figure above, positive control atorvastatin can significantly decrease low density lipoprotein in Apo E ko mice fed with HFD after 16 week treatment and there is no significant difference in net change of LDL.


7.7. The Effect of RAAS Antibody on Atherosclerosis Plaque Lesion



FIG. 151. Illustrated by negative control group animal #10



FIG. 152. Illustrated by negative control group animal #10 As shown in the above diagram, we calculated all the plaque area stained by oil red and divided by total inner vascular area





Area percent (%)=Sum area of atherosclerotic plaque (mm2)/whole area of vascular inner wall (mm2)



FIG. 153. Percent of plaque area in total inner vascular area


No significant difference between the vehicle and treatment groups in plaque area and percentage of plaque area although Atorvastatin showed a mild trend to decrease percentage of plaque area after 16 weeks oral administration.



FIG. 154. Illustrated Analysis of arterial arch area


The total area of aorta from the aortic root to the thoracic aorta was measured (bracketed area).


As shown in the left panel, because the total lumen area in arterial arch is very difficult to identify in en face vessel, we measured the total area at the length of about 2 mm from aortic root down to the thoracic artery (bracketed area).



FIG. 155. Percent of plaque area in arterial arch area


The plaque lesion was more severe in mice fed with HFD than mice in the normal diet (negative) group. No significant difference between the vehicle and treatment groups in plaque area and percentage of plaque area.



FIG. 156. Illustrated Analysis of from root to right renal artery


As shown in the left panel, the total area from the aortic root to the right renal artery were measured (bracketed area)



FIG. 157. Percent of plaque area from root to right renal artery


There is no significant difference between vehicle and treatment groups in plaque area and percentage of plaque area.


7.8. The Effect of RAAS Antibody on Liver Weight



FIG. 158. Diagram of liver weight



FIG. 159. Diagram of liver index


Atorvastatin at 20 mg/kg reduced the ratio of liver/body weight significantly after 16 weeks treatment, which is consistent with the 8 weeks treatment result in study 2.


7.9 Comparison of Percentage of Plaque Area in Study 1, 2, 3



FIG. 160. Comparison of percentage of plaque area in study 1, 2, 3


We also compared percent of plaque area in the study 1, 2 and 3. In study 1, all ApoE ko mice were fed with HFD for 4 weeks and mice were sacrificed at 14 weeks of age. In study 2, all ApoE ko mice were fed with HFD for 19 weeks except the mice in negative control group and all mice were sacrificed at 29 weeks of age. In study 3, the ApoE ko mice were fed with HFD for 27 weeks and sacrificed at 37 weeks. It is apparent that:


1. The plaque area increased steadily with HFD feeding time or aging.


2. The aorta atherosclerosis model in ApoE ko mouse was established successfully.


3. HFD feeding for 10 weeks plus 8 weeks Rx gave best result.


7.10 Comparison of TC Level in Study 1, 2, 3



FIG. 161. Comparison of TC level in study 1, 2, 3



FIG. 162. Comparison of percentage of plaque area in study 1, 2, 3


The TC and LDL values from study 1, 2 and 3 in vehicle and reference groups peaked at week 10, and deceased subsequently during 27 weeks high fat diet feeding. This phenomenon was also observed in relevant literature reports (details can be seen in the report on ppt. version).


7.11. Image of Aorta with Red Oil Staining


One image of aorta stained by oil red from each group was selected and showed below. The branches of artery and the lipid plaques could be observed clearly and the plaques mainly distribute in the aortic root and principal branches of the abdominal aorta. It is consistent with the reference literatures.



FIG. 164, Images of aorta plaque lesions after 16 weeks treatment


8. Summary and Interpretation

  • 1). Atorvastatin at 40 mg/kg significantly reduced liver/BW ratio, plasma TC, HDL and LDL, but did not affect the plaque lesion area of aorta in ApoE KO mice after 16 weeks treatment.
  • 2). RAAS APOA1 did not affect the lipid profile in ApoE KO mice after 16 weeks treatment.
  • 3). RAAS APOA1 did not reduce the plaque lesion area of aorta in ApoE KO mice after 16 weeks treatment.


Interpretation:

  • 1). The % athero-plaque lesion area reached 50% at the end of 16 week treatment. The 26 week HFD feeding might have made the mice too sick for the test drugs to reverse.
  • 2). Seems 8 weeks treatment gave optimal athero-plaque reduction, as shown by RAAS Study 2 as well as by literature reports.
  • 3). If repeat, suggest to reduce the HFD feeding duration before drug treatment to <6 weeks, and keep the treatment duration to 8 weeks.


9. Conclusion:

  • 1). Atorvastatin at 40 mg/kg significantly reduced plasma TC, HDL and LDL level, liver weight and the ratio of liver/BW, but did not affect the plaque lesion area of aorta in ApoE KO mice after 16 weeks treatment.
  • 2). RAAS antibody APOA1 didn't affect the lipid profile and reduce the plaque lesion of aorta in ApoE KO mice after 16 weeks treatment.
  • Title: Anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and AFOD in patient-derived tumor xenograft (PDX) models in nude mice.
  • Description: Patient-derived colorectal tumor xenograft (PDX) model was used to evaluate the anti-cancer efficacy of the high concentrated fibrinogen enriched a1at thrombin and AFOD at different 3 doses. The results showed that high concentrated fibrinogen enriched a1at thrombin and AFOD at all doses significantly inhibited the growth of PDX tumors implanted at 4 different locations of the peritoneum while having minor effects on mice body weights, which indicated high concentrated fibrinogen enriched a1at thrombin and AFOD is a potent anti-cancer agent on colorectal cancer with a limited side effect.
  • Subject: high concentrated fibrinogen enriched a1at thrombin and AFOD, fibrinogen, thrombin, patient-derived tumor xenograft model, colorectal cancer


Summary


Patient-derived colorectal tumor xenograft (PDX) models (CO-04-0001 or CO-04-0002) were used to evaluate the anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod at 3 doses. PDX tumors (CO-04-0001 or CO-04-0002) were implanted at 4 different locations in peritoneal cavity, and high concentrated fibrinogen enriched a1at thrombin and Afod, or a control agent was applied to peritoneum before and after tumor implantation. 30 days after implantation, the mice were sacrificed and tumors were dissected and weighed. The final tumor weights for all groups were statistically analyzed by one-way ANOVA with the significance level set at 0.05.


The data show that high concentrated fibrinogen enriched a1at thrombin and Afod at all 3 doses exhibits significant inhibitory effects on tumor growth in PDX colorectal cancer model while no significant toxicity was observed, which indicates high concentrated fibrinogen enriched a1at thrombin and Afod is a potential anti-tumor agent in colorectal cancer, warranting further development of the agent for clinical application.


Introduction


The aim of the study was to test anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod in patient-derived colorectal tumor xenograft (PDX) model in nude mice.


The model used in the study was derived from surgically resected, fresh patient tumor tissues. The first generation of the xenograft tumors in mice was termed passage 0 (P0), and so on during continual implantation in mice. The passage of xenograft tumors at P2 (CO-04-0002) or P3 (CO-04-0001) were used in this study.


All the experiments were conducted in the AAALAC-accrediated animal facility in compliance with the protocol approved by the Institutional Animal Care and Use Committee (IACUC).


Methods


Experimental Preparations


Animal Preparation


Female Balb/c nude mice, with a body weight of approximately 20 grams, were obtained from an approved vendor (Sino-British SIPPR/BK Lab. Animal Co. Ltd., Shanghai, China).


Acclimation/Quarantine:


Upon arrival, animals were assessed as to their general health by a member of a veterinary staff or authorized personnel. Animals were acclimated for at least 3 days (upon arrival at the experiment room) before being used for the study.


Animal Husbandry:


Animals were housed in groups during acclimation and individually housed during in-life. The animal room environment was adjusted to the following target conditions: temperature 20 to 25° C., relative humidity 40 to 70%, 12 hours artificial light and 12 hours dark. Temperature and relative humidity was monitored daily.


All animals had access to Certified Rodent Diet (Sino-British SIPPR/BK Lab. Animal Co. Ltd., Shanghai, China) ad libitum. Animals were not fasted prior to the study. Water was autoclaved before provided to the animals ad libitum. Periodic analyses of the water were performed and the results were archived at WuXi AppTec. There were no known contaminants in the diet or water which, at the levels detected expected to interfere with the purpose, conduct or outcome of the study.


Tumor Tissue Preparation


The colorectal xenograft tumor models were established from surgically resected clinical tumor samples. The first generation of the xenograft tumors in mice is termed passage 0 (P0), and so on during continual implantation in mice. The tumor tissues at passage 2 (CO-04-0002) or P3 (CO-04-0001) were used in this study.


Formulation


Test agent: high concentrated fibrinogen enriched a1at thrombin and Afod were provided by RAAS and prepared by RAAS scientist during experiment before use.


Control agent: Matrigel (BD Biosciences; cat. #356234).


Experimental Protocol


Establishment of Xenograft Model and Treatment


Grouping and Treatment


Nude mice were assigned to 6 different groups with 12-17 mice/group and each group received different treatment as shown in Table 9.1.


8 out 17 (9 left) mice in high dose high concentrated fibrinogen enriched a1at thrombin and Afod group died during the first experiment using PDX model CO-04-0002. To make up for the loss of mice in high dose group, 6 additional mice were implanted with tumor fragments collected from model CO-04-0001 and treated with high dose high concentrated fibrinogen enriched a1at thrombin and Afod. So the total mice number in high dose group was 15.









TABLE 9.1







Grouping and the treatment.










Group
Treatment
N
Remarks





1
Sham-operation
12
Open up the abdominal cavity and





close it with sutures. (No implants)


2
Vehicle control
12
Implant tumor fragments of 20 mm3 in





size into 4 corners of abdominal cavity. Close





body with sutures.


3
Matrigel
12
Embed tumor fragments of 20 min3 in





Matrigel. Implant the tumor fragments into 4





corners of abdominal cavity. Close body





with sutures.


4
3 ml of high concentrated
9 + 6
Spray high concentrated fibrinogen




fibrinogen enriched alat



enriched a1at thrombin and Afod to cover the





thrombin and Afod (high dose)


entire peritoneum and the internal organs.



on the peritoneum in

Implant the tumor fragments of 20 mm3 into



abdominal cavity of nude

4 corners of abdominal cavity. Close body with



mice

sutures.


5
2 ml of high concentrated
12
Spray high concentrated fibrinogen




fibrinogen enriched a1at



enriched a1at thrombin and Afod to cover the





thrombin and Afod (moderate


entire peritoneum and the internal organs.



dose) on the peritoneum in

Implant the tumor fragments of 20 mm3 into 4



abdominal cavity of nude mice

corners of abdominal cavity. Close body with





sutures.


6
1 ml of high concentrated
13
Spray high concentrated fibrinogen




fibrinogen enriched a1at



enriched a1at thrombin and Afod to cover the





thrombin and Afod (low dose)


entire peritoneum and the internal organs.



on the peritoneum in

Implant the tumor fragments of 20 mm3 into



abdominal cavity of nude

4 corners of abdominal cavity. Close body with



mice

sutures.


Total

76









Experiment Procedures

  • A. The animal was anesthetized by i.p. injection of sodium pentobarbital at 60-70 mg/kg. Disinfect the abdominal skin of nude mice with 70% ethanol solution. Open up the abdominal wall along the midline of the ventral surface to expose the peritoneal surface.
  • B. The surgeries for different groups were done according to table 9.1.
  • C. For groups using test agent, high concentrated fibrinogen enriched a1at thrombin and Afod was then applied on the peritoneal surface.
  • D. Tumor fragments were implanted at 4 different locations of the peritoneal cavity. The test agent acted as a glue to hold the fragments.
  • E. The test agent was applied again on the surface of tumor fragments and peritoneum.
  • F. After the fibrin membrane formed completely, the peritoneal cavity was closed. G. In Matrigel control groups, tumor fragments were embedded into matrigel before implantation.
  • H. Postoperative cares followed protocol SOP-BEO-0016-1.0.
  • I. Mice were palpated for tumors 2 weeks after implantation. The ratio of palpable tumors observed in each group was recorded.
  • J. 30 days after implantation, the mice were sacrificed and tumors were dissected and weighed.
  • K. The tissues surrounding tumor fragments were also checked to find out whether the tumors had spread to other organ sites within the peritoneal cavity.
  • L. Pictures of tumor-bearing mice and dissected tumors were taken.
  • M. If possible, tumor sizes were measured twice per week. Tumor volumes (mm3) are obtained by using the following formula: volume=(W2×L)/2 (W, width; L, length in mm of the tumor).
  • N. During the experiment, health conditions of mice were observed daily. Body weights of mice were monitored twice per week.


Evaluation of the Anti-Tumor Activity


Health conditions of mice were observed daily. Body weights were measured twice per week during the treatment. Mice were palpated for tumors 2 weeks after implantation. The ratio of palpable tumors observed in each group was recorded.


30 days after treatment, all mice were euthanized with CO2 and cervical dislocation was followed after respiratory arrest. Routine necropsy was performed to detect any abnormal signs of each internal organ with specific attention to metastases. Each tumor was removed and weighted.


Drugs and Materials


High concentrated fibrinogen enriched a1at thrombin and Afod were provided by RAAS; Matrigel was from BD Biosciences (San Jose, Calif., cat. #356234). Digital caliper was from Sylvac, Switzerland.


Data Analysis


Relative Change of Body Weight (RCBW)


Relative change of body weight (RCBW) was calculated based on the following formula: RCBW (%)=(BWi−BW0)/BW0×100%; BWi was the body weight on the day of weighing and BW0 was the body weight before surgery.


Tumor Weight


Tumors from each mouse were pooled and weighed after sacrificing mice.


Statistical Analysis


Data were expressed as mean±SEM; the difference between the groups was analyzed for significance using one-way ANOVA and Dunnett's test.


Results


Tumor Growth Inhibition


Three weeks after implantation, all 12 mice in vehicle control group showed palpable tumors, while only less than 2 palpable tumors were found in each test agent-treated group. High concentrated fibrinogen enriched a1at thrombin and Afod treatment delayed the appearance of palpable tumors as shown in table 9.2, indicating high concentrated fibrinogen enriched a1at thrombin and Afod inhibited the growth of implanted colorectal tumors in vivo.


Thirty days after implantation, tumors in vehicle control group and matrigel group reached more than 1 g on average. Conversely, tumor weights in test agent high, moderate and low dose groups were 0.49 g (0.35 if when two models are combined), 0.28 g and 0.13 g, respectively. Compared with the vehicle control, high concentrated fibrinogen enriched a1at thrombin and Afod demonstrated significant anti-tumor activities in colorectal cancer PDX model at all 3 doses. The inhibition on tumor growth were shown in FIGS. 26.18 & 26.22 and table 9.2.


Effect on Body weight


Loss of body weight, a sign of toxicity, was not seen in test agent-treated groups, which only showed minor decrease in weight gain. Mortalities were observed within 3 days after surgery and treatment in high dose of test agent group, which may due to the large volume (3 ml) of test agent used in this group.


The effect on body weight was shown in FIG. 26.24 and table 9.3.


Discussion


Patient-derived colorectal tumor xenograft (PDX) model was used to evaluate the anti-cancer efficacy of the high concentrated fibrinogen enriched a1at thrombin and Afod at 3 doses. PDX tumors (CO-04-0001 and CO-04-0002) were implanted at 4 different locations in peritoneal cavity, and high concentrated fibrinogen enriched a1at thrombin and Afod, or a control agent was applied to peritoneum before and after tumor implantation.


Mice were palpated for tumors 2 weeks after implantation. The ratio of palpable tumors observed in each group was recorded. Test agent treatment inhibited the tumor growth as shown by the delayed appearance of palpable tumors. There weeks after implantation, all 12 mice in vehicle control group showed palpable tumors, while only less than 2 palpable tumors were found in each test agent-treated group (Table 9.2).


Thirty days after implantation, the mice were sacrificed and tumors were dissected and weighed. Tumors in vehicle control group and matrigel group reached more than 1 g on average. Conversely, tumor weights in test agent high, moderate and low dose groups were 0.49 g (0.35 when two models are combined), 0.28 g and 0.13 g, respectively. Compared with the vehicle control, high concentrated fibrinogen enriched a1at thrombin and Afod demonstrated significant anti-tumor activities in colorectal cancer PDX model at all 3 doses. Matrigel has been commonly used to facilitate the establishment of human tumor xenografts in rodents. In this study, matrigel group promoted an increase in tumor weight thought the increase was not statistically significant.


Loss of body weight, a sign of toxicity, was not seen in all test agent-treated groups, in which the animals only showed a minor decrease in weight gain compared to sham-operated group. Mortalities observed in test agent high dose group right after the surgery could be due to large volume of test agent (3 ml) used in this group. The mice of vehicle and matrigel groups started to loss body weights 2 weeks after surgery due to the continuously increased tumor volumes.


In summary, the results show that high concentrated fibrinogen enriched a1at thrombin and Afod at all doses significantly inhibits the growth of colorectal tumors in vivo while having minor effects on mice body weight. The results suggest that high concentrated fibrinogen enriched a1at thrombin and Afod is a potent anti-tumor agent in colorectal cancer.


Figures



FIG. 165. Anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod in PDX model CO-04-0002.


Colorectal cancer: CO-04-0002 P3


Tumor weights from model CO-04-0002 were used. Data are expressed as mean±SEM. *<0.05, ***<0.001 vs vehicle group (one-way ANOVA and Dunnett's test).



FIG. 166. Anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod in PDX model CO-04-0002 and CO-04-0001.


Colorectal cancer: CO-04-0002 P3+CO-04-0001 P4


Tumor weights of 6 mice from model CO-04-0001 were combined with the data from model CO-04-0002. There were 15 mice in total in high dose of test agent group. Data are expressed as mean±SEM. *<0.05, ***<0.001 vs vehicle group (one-way ANOVA and Dunnett's test).



FIG. 167. Photographs of tumors dissected from abdominal cavity of each group.


Tumors from each mouse were pooled and weighed. The tumors in frame were from model CO-04-0002 (upper panels) and the rest were form model CO-04-0001 (bottom panel). Scale bar, 1 cm.



FIG. 168. Relative change of body weight (%) of different groups.


Data are expressed as mean±SEM. Relative change of body weight (RCBW) was calculated based on the following formula: RCBW (%)=(BWi−BW0)/BW0×100%; BWi was the body weight on the day of weighing and BW0 was the body weight before surgery.


Confidential


Tables









TABLE 9.2







Ratios of palpable tumors observed in each group.















Days after










surgery
15
16
17
18
20
21
24
28


















Sham-operated
0/12
0/12
0/12
0/12
0/12
 0/12
 0/12
 0/12


group










vehicle control
0/12
1/12
4/12
4/12
8/12
12/12
12/12
12/12


group










Matrigel
1/12
3/12
5/12
5/12
5/12
 8/12
11/12
12/12


high dose of
0/9
0/9
0/9
0/9
0/9
 0/9
 0/9
 5/9


test agent










moderate dose of
0/13
0/13
1/13
1/13
1/13
 2/13
 2/13
 5/13


test agent










low dose of
0/12
0/12
1/12
1/12
1/12
 1/12
 2/12
 7/12


test agent





Mice were palpated for tumors at 15, 16, 17, 18, 20, 21, 24, 28 days after implantation.


The ratios of palpable tumors observed in each group were recorded.













TABLE 9.3







Relative change of body weight (%) of different groups.






















Days




































after
















surgery
0
1
2
3
4
5
6
7
8
9
14
21
24
28





Group
RCB
RCB
RCB
RCB
RCB
RCB
RCB
RCB
RCB
RCB
RCB
RCB
RCB
RCB



W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)
W (%)






















Sham-
Mean
0.00
−7.41
−3.46
−0.73
−1.24
3.08
3.18
2.84
2.45
8.67
11.20
16.61
16.46
15.70


operated
SD
0.00
2.98
3.03
3.03
4.19
2.94
3.18
3.18
4.21
4.21
5.70
5.31
4.87
5.07



SEM
0.00
0.86
0.87
0.88
1.21
0.85
0.92
0.92
1.21
1.21
1.65
1.53
1.41
1.46


Vehicle
Mean
−2.14
−7.06
−4.16
−2.12
−0.99
2.37
2.24
3.38
2.55
3.92
−1.48
1.02
−2.70
−6.01


control
SD
0.87
2.62
2.93
3.82
3.99
4.06
4.10
3.85
4.34
5.74
8.75
9.95
9.36
8.72



SEM
0.25
0.76
0.85
1.10
1.15
1.17
1.18
1.11
1.25
1.66
2.53
2.87
2.70
2.52


Matrigel
Mean
−1.97
−9.20
−7.41
−4.43
−3.47
0.62
0.09

1.50
−0.29

−7.17
−6.25
−8.92



SD
1.14
2.37
3.60

2.17
2.74
2.46
3.07
3.35
4.95
7.26
8.33
7.24
6.50



SEM
0.33
0.68
1.04
0.73
0.63
0.79
0.71
0.89
0.97
1.43
2.10
2.40
2.09
1.88


High
Mean
2.66
−8.41
−7.80
−7.78
−3.48
−2.09
−0.26
0.50
0.73
7.71
6.72
9.28
5.90
2.48


dose
SD
6.60
2.76
4.29
5.40
3.19
5.68
6.05
6.52
5.77
7.22
7.93
8.90
10.53
10.39


Of test
SEM
2.20
0.92
1.43
1.80
1.06
1.89
2.02
2.17
1.92
2.41
2.64
2.97
3.51
3.46


agent

















Mod-
Mean
5.95
−6.73
−5.23
−3.70
−1.70
0.30
2.37
2.55
2.66
7.00
8.46
11.16
10.55
7.68


erate
SD
4.04
2.13
2.43
4.38
4.61
5.08
4.15
5.29
5.85
5.58
7.03
7.98
10.25
9.57


dose of
SEM
1.12
0.59
0.67
1.22
1.28
1.41
1.15
1.47
1.62
1.55
1.95
2.21
2.84
2.66


test

















agent

















Low
Mean
1.82
−5.27
−3.04
−2.75
1.93
1.00
2.86
2.11
3.85
7.08
7.08
12.03
12.27
9.18


Dose
SD
2.74
2.24
2.55
2.53
2.90
2.15
3.00
2.89
3.17
3.78
3.78
3.65
3.77
4.16


of test
SEM
0.79
0.65
0.74
0.73
0.84
0.62
0.87
0.84
0.92
1.09
1.09
1.05
1.09
1.20


agent









Relative change of body weight (RCBW) was calculated based on the following formula: RCBW (%)=(BWi−BW0)/BW0×100%;


BWi was the body weight on the day of weighing and BW0 was the body weight before surgery.

  • Title: Anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod in a patient-derived tumor xenograft (PDX) model of lung cancer in nude mice.
  • Description: Patient-derived tumor xenograft (PDX) model of lung cancer was used to evaluate the anti-cancer efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod at different 3 doses. The results showed that high concentrated fibrinogen enriched a1at thrombin and afod at all doses significantly inhibited the growth of PDX tumors implanted at 4 different locations of the peritoneum while having minor effects on mice body weights, which indicates high concentrated fibrinogen enriched a1at thrombin and Afod is a potent anti-cancer agent on lung cancer with a limited side effect.
  • Subject: high concentrated fibrinogen enriched a1at thrombin and Afod, patient-derived tumor xenograft model, lung cancer


Summary


Patient-derived tumor xenograft (PDX) model of lung cancer (LU-01-0032) was used to evaluate the anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod at 3 doses. PDX tumors (LU-01-0032) were implanted at 4 different locations in peritoneal cavity, and high concentrated fibrinogen enriched a1at thrombin and Afod or a control agent was applied to peritoneum before and after tumor implantation. Forty five days after implantation, the mice were sacrificed and tumors were removed and weighed. The final tumor weights for all groups were statistically analyzed by one-way ANOVA with the significance level set at 0.05.


The data show that high concentrated fibrinogen enriched a1at thrombin and Afod at all 3 doses exhibits significant inhibitory effects on tumor growth in the lung cancer model while no significant toxicity was observed, which indicates high concentrated fibrinogen enriched a1at thrombin and Afod was a potential anti-tumor agent in lung cancer, warranting further development of high concentrated fibrinogen enriched a1at thrombin and Afod for clinical application.


1. Introduction


The aim of the study was to test anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod in patient-derived lung tumor xenograft (PDX) model in nude mice.


The model used in the study was derived from surgically resected, fresh patient tumor tissues. The first generation of the xenograft tumors in mice was termed passage 0 (P0), and so on during continual implantation in mice. The passage of xenograft tumors at P5 (LU-01-0032) were used in this study.


All the experiments were conducted in the AAALAC-accrediated animal facility in compliance with the protocol approved by the Institutional Animal Care and Use Committee (IACUC).


2. Methods


Mental Preparations


2.1.1. Animal Preparation


Female Balb/c nude mice, with a body weight of approximately 20 grams, were obtained from an approved vendor (Sino-British SIPPR/BK Lab. Animal Co. Ltd., Shanghai, China).


Acclimation/Quarantine:


Upon arrival, animals were assessed as to their general health by a member of a veterinary staff or authorized personnel. Animals were acclimated for at least 3 days (upon arrival at the experiment room) before being used for the study.


Animal Husbandry:


Animals were housed in groups during acclimation and individually housed during in-life. The animal room environment was adjusted to the following target conditions: temperature 20 to 25° C., relative humidity 40 to 70%, 12 hours artificial light and 12 hours dark. Temperature and relative humidity was monitored daily.


All animals had access to Certified Rodent Diet (Sino-British SIPPR/BK Lab. Animal Co. Ltd., Shanghai, China) ad libitum. Animals were not fasted prior to the study. Water was autoclaved before provided to the animals ad libitum. Periodic analyses of the water were performed and the results were archived at WuXi AppTec. There were no known contaminants in the diet or water which, at the levels detected expected to interfere with the purpose, conduct or outcome of the study.


2.1.2. Tumor Tissue Preparation


The lung xenograft tumor models were established from surgically resected clinical tumor samples. The first generation of the xenograft tumors in mice is termed passage 0 (P0), and so on during continual implantation in mice. The tumor tissues at passage 5 (LU-01-0032) were used in this study.


2.1.3. Formulation


High concentrated fibrinogen enriched a1at thrombin and Afod were provide by RAAS and prepared by RAAS scientist during experiment before use.


Matrigel (BD Biosciences; cat. #356234).


2.2. Experimental Protocol


2.2.1. Establishment of Xenograft Model and Treatment


Grouping and Treatment


Nude mice were assigned to 6 different groups with 11-19 mice/group and each group received different treatments as shown in Table 1.









TABLE 1







Grouping and the treatment.










Group
Treatment
N
Remarks





1
Sham-operation
12
Open up the abdominal cavity and close it with





sutures. (No implants)


2
Vehicle control
13
Implant tumor fragments of 20 mm3 in size into





4 corners of abdominal cavity. Close body with





sutures.


3
Matrigel
13
Embed tumor fragments of 20 mm3 in Matrigel.





Implant the tumor fragments into 4 corners of





abdominal cavity. Close body with sutures.


4
3 ml high concentrated
19
Spray high concentrated fibrinogen enriched a1at



fibrinogen enriched a1at

thrombin and Afod to cover the entire



thrombin and Afod (high dose)

peritoneum and the internal organs. Implant the



on the peritoneum in abdominal

tumor fragments of 20 mm3 into 4 corners of



cavity of nude mice

abdominal cavity. Close body with sutures.


5
2 ml high concentrated
14
Spray high concentrated fibrinogen enriched a1at



fibrinogen enriched a1at

thrombin and Afod to cover the entire



thrombin and Afod (moderate

peritoneum and the internal organs. Implant the



dose) on the peritoneum in

tumor fragments of 20 mm3 into 4 corners of



abdominal cavity of nude mice

abdominal cavity. Close body with sutures.


6
1 ml high concentrated
11
Spray high concentrated fibrinogen enriched a1at



fibrinogen enriched a1at

thrombin and Afod to cover the entire



thrombin and Afod (low dose)

peritoneum and the internal organs. Implant the



on the peritoneum in abdominal

tumor fragments of 20 mm3 into 4 corners of



cavity of nude mice

abdominal cavity. Close body with sutures.


Total

82









Experiment Procedures

  • A. Measured the body weight of each mouse before surgery.
  • B. The animal was anesthetized by i.p. injection of sodium pentobarbital at 60-70 mg/kg. Disinfect the abdominal skin of nude mice with 70% ethanol solution. Open up the abdominal wall along the midline of the ventral surface to expose the peritoneal surface.
  • C. The surgeries for different groups were done according to table 1.
  • D. For groups using test agent high concentrated fibrinogen enriched a1at thrombin and Afod, the test agent was then applied on the peritoneal surface.
  • E. Tumor fragments were implanted at 4 different locations of the peritoneal cavity. The test agent acted as a glue to hold the fragments.
  • F. The test agent high concentrated fibrinogen enriched a1at thrombin and Afod was applied again on the surface of tumor fragments and peritoneum.
  • G. After the fibrin membrane formed completely, the peritoneal cavity was closed.
  • H. In Matrigel control groups, tumor fragments were embedded into matrigel before implantation.
  • I. Postoperative cares followed protocol SOP-BEO-0016-1.0.
  • J. Mice were palpated for tumors 2 weeks after implantation. The ratio of palpable tumors observed in each group was recorded.
  • K. Forty five days after implantation, the mice were sacrificed and tumors were dissected and weighed.
  • L. The tissues surrounding tumor fragments were also checked to find out whether the tumors had spread to other organ sites within the peritoneal cavity.
  • M. Pictures of tumor-bearing mice and dissected tumors were taken.
  • N. If possible, tumor sizes were measured twice per week. Tumor volumes (mm3) are obtained by using the following formula: volume=(W2×L)/2 (W, width; L, length in mm of the tumor).
  • O. During the experiment, health conditions of mice were observed daily. Body weights of mice were monitored twice per week.


2.2.2. Evaluation of the Anti-Tumor Activity


Health conditions of mice were observed daily. Body weights were measured twice per week during the treatment. Mice were palpated for tumors 2 weeks after implantation. The ratio of palpable tumors observed in each group was recorded. 45 days after treatment, all mice were euthanized with CO2 and cervical dislocation was followed after respiratory arrest. Routine necropsy was performed to detect any abnormal signs of each internal organ with specific attention to metastases. Each tumor was removed and weighted.


2.3. Drugs and Materials


High concentrated fibrinogen enriched a1at thrombin and Afod were provided by RAAS; Matrigel was from BD Biosciences (San Jose, Calif., cat. #356234).


Digital caliper was from Sylvac, Switzerland.


2.4. Data Analysis


2.4.1. Relative Change of Body Weight (RCBW)


Relative change of body weight (RCBW) was calculated based on the following formula: RCBW (%)=(BWi−BW0)/BW0×100%; BWi was the body weight on the day of weighing and BW0 was the body weight before surgery.


2.4.2. Tumor Weight


Tumors from each mouse were pooled and weighed after sacrificing mice.


2.4.3. Statistical Analysis


Data were expressed as mean±SEM; the difference between the groups was analyzed for significance using one-way ANOVA and Dunnett's test.


3. Results


3.1. Tumor Growth Inhibition


Four weeks after implantation, 9 out of 13 mice in vehicle control group showed palpable tumors, while only less than 5 palpable tumors were found in each high concentrated fibrinogen enriched a1at thrombin and Afod-treated group. High concentrated fibrinogen enriched a1at thrombin and Afod treatment delayed the appearance of palpable tumors as shown in table 2, indicating high concentrated fibrinogen enriched a1at thrombin and Afod inhibited the growth of implanted lung tumors in vivo. After sacrificing the mice, tumors were found in all the mice in vehicle control group, while some tumors completely regressed in several high concentrated fibrinogen enriched a1at thrombin and Afod-treated mice (FIG. 3).


Forty-five days after implantation, tumors in vehicle control group reached more than 0.7 g on average. Conversely, tumor weights in high concentrated fibrinogen enriched a1at thrombin and Afod high, moderate and low dose groups were 0.19 g, 0.16 g and 0.16 g, respectively. Compared with the vehicle control, high concentrated fibrinogen enriched a1at thrombin and Afod demonstrated significant anti-tumor activities in lung cancer PDX model at all 3 doses (FIG. 1˜2).


The inhibition on tumor growth were shown in FIG. 1˜3 and table 2.


3.2. Effect on Body Weight


Loss of body weight, a sign of toxicity, was not seen in high concentrated fibrinogen enriched a1at thrombin and Afod-treated groups, indicating the test agent has no/little side effects.


The effect on body weight was shown in FIG. 4 and table 3.


4. Discussion


Patient-derived tumor xenograft (PDX) model of lung cancer was used to evaluate the anti-cancer efficacy of the high concentrated fibrinogen enriched a1at thrombin and Afod at 3 doses. PDX tumors (LU-01-0032) were implanted at 4 different locations in peritoneal cavity, and high concentrated fibrinogen enriched a1at thrombin and Afod or a control agent was applied to peritoneum before and after tumor implantation.


Mice were palpated for tumors 2 weeks after implantation. The ratio of palpable tumors observed in each group was recorded. High concentrated fibrinogen enriched a1at thrombin and Afod treatment inhibited the tumor growth as shown by the delayed appearance of palpable tumors and decreased tumor incidence. Four weeks after implantation, 9 out of 13 mice in vehicle control group showed palpable tumors, while only less than 5 palpable tumors were found in each high concentrated fibrinogen enriched a1at thrombin and Afod-treated group (Table 2).


Forty-five days after implantation, the mice were sacrificed and tumors were dissected and weighed. After sacrificing the mice, tumors were found in all the mice in vehicle control group, while some tumors completely regressed in several high concentrated fibrinogen enriched a1at thrombin and Afod-treated mice. Tumors in vehicle control group reached more than 0.7 g on average. Conversely, tumor weights in high concentrated fibrinogen enriched a1at thrombin and Afod high, moderate and low dose groups were 0.19 g, 0.16 g and 0.16 g, respectively. Compared with the vehicle control, high concentrated fibrinogen enriched a1at thrombin and Afod demonstrated significant anti-tumor activities in lung cancer PDX model at all 3 doses. Matrigel has been commonly used to facilitate the establishment of human tumor xenografts in rodents. In this study, matrigel group also showed a significant inhibitory effect on tumor weight.


Loss of body weight, a sign of toxicity, was not seen in all high concentrated fibrinogen enriched a1at thrombin and Afod-treated groups, indicating the test agent has no/little side effects.


In summary, the results show that high concentrated fibrinogen enriched a1at thrombin and Afod at all doses significantly inhibits the growth of lung tumors in vivo while having minor effects on mice body weight. The results suggest that high concentrated fibrinogen enriched a1at thrombin and Afod is a potent anti-tumor agent in lung cancer.


5. Figures



FIG. 169. Anti-tumor efficacy of high concentrated fibrinogen enriched a1at thrombin and Afod in PDX model LU-01-0032.


Tumor weights from model LU-01-0032 were used. Data are expressed as mean±SEM. *<0.05, **<0.01, ***<0.001 vs vehicle group (one-way ANOVA and Dunnett's test).



FIG. 170. Photographs of tumors dissected from abdominal cavity of each group.


Tumors from each mouse of model LU-01-0032 were pooled and weighed. Scale bar, 1 cm. A, sham-operated; B, vehicle control; C, matrigel; D, test agent high dose; E, test agent moderate dose; F, test agent low dose.



FIG. 171. Ratios of mice with palpable tumors observed in each group.


After sacrificing the mice, the tumors from each mouse of model LU-01-0032 were pooled and the ratios of mice bearing tumors in each group were recorded.



FIG. 172. Relative change of body weight (%) of different groups.


Data are expressed as mean±SEM. Relative change of body weight (RCBW) was calculated based on the following formula: RCBW (%)=(BWi−BW0)/BW0×100%; BWi was the body weight on the day of weighing and BW0 was the body weight before surgery.


6. Tables


Relative change of body weight (RCBW) was calculated based on the following formula: RCBW (%)=(BWi−BW0)/BW0×100%; BWi was the body weight on the day of weighing and BW0 was the body weight before surgery.


















Title:
In Vivo Test of Efficacy of FS in the Treatment of




BEL-7404 Peritoneal Implantation Model



Description:
BEL-7404 peritoneal Implantation hepatic cancer




model was used to evaluate the anti-cancer efficacy




of the FS at 2 ml/mouse. The results showed that FS




had inhibition on tumor growth.



Subject:
FS, BEL-7404, hepatic cancer



Project ID:
RAAS-20130425










Summary


BEL-7404 peritoneal Implantation hepatic cancer model was used to evaluate the anti-cancer efficacy of FS at 2 ml/mouse. On day 21 after implantation, all mice in vehicle group and positive group exhibited palpable tumors, while no mice in FS group exhibited any palpable tumors. On day 28 after implantation, the tumor in vehicle group reached 0.88 g, while FS group was only 0.06 g. On day 52 after implantation, Five mice treated with FS did not exhibit any palpable tumor. The results showed that FS had inhibition on tumor growth.


No body weight loss and toxicity were found in FS-treated groups, which showed FS had no side effect.


Introduction


The objective of the research is to evaluate the in vivo efficacy of FS in the treatment of a hepatic cancer model.


All the experiments were conducted in the AAALAC-accrediated animal facility in compliance with the protocol approved by the Institutional Animal Care and Use Committee (IACUC).


Methods


Experimental Preparations


Animal Preparation


Female Balb/c nude mice, with a body weight of approximately 20 grams, were obtained from an approved vendor (Shanghai BK Laboratory Animal Co., LTD., Shanghai, China).


Acclimation/Quarantine:


Upon arrival, animals were assessed as to their general health by a member of a veterinary staff or authorized personnel. Animals were acclimated for at least 3 days (upon arrival at the experiment room) before being used for the study.


Animal Husbandry:


Animals were housed in groups during acclimation and individually housed during in-life. The animal room environment was adjusted to the following target conditions: temperature 20 to 25° C., relative humidity 40 to 70%, 12 hours artificial light and 12 hours dark. Temperature and relative humidity was monitored daily.


All animals had access to Certified Rodent Diet (Shanghai BK Laboratory Animal Co., LTD., Shanghai, China) ad libitum. Animals were not fasted prior to the study. Water was autoclaved before provided to the animals ad libitum. Periodic analyses of the water were performed and the results were archived at WuXi AppTec. There were no known contaminants in the diet or water which, at the levels detected expected to interfere with the purpose, conduct or outcome of the study.


Cell Culture:


The BEL-7404 tumor cells were maintained in vitro as a monolayer culture in RPMI 1640 medium supplemented with 10% heat inactivated fetal bovine serum, 100 U/ml penicillin and 100 μg/ml streptomycin, and L-glutamine (2 mM) at 37° C. in an atmosphere of 5% CO2 in air. The tumor cells were routinely subcultured twice weekly by trypsin-EDTA treatment. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation. When the average tumor volume reached 1000 mm3, sacrificed the tumor bearing mice and removed the tumor for orthotopic implantation.


Formulation


FS were provide by RAAS and prepared by RAAS scientist during experiment before use.


Matrigel (BD Biosciences; cat. #356234)


Experimental Protocol


Establishment of Xenograft Model and Treatment


Grouping and Treatment


Nude mice were assigned to 6 different groups with 3 mice/group and each group received different treatment as shown in Table 1.









TABLE 1







Grouping and the treatment










Group
Treatment
N
Remarks













1
Vehicle control
3
Implant a tumor fragment of 40 mm3 into the abdominal





cavity. Close body with sutures


2
Positive control
3
Embed a tumor fragment of 40 mm3 in Matrigel. Implant the





tumor fragment into abdominal cavity. Close body with





sutures


3
3-FS (2 ml) +
3
Spray FS to cover the entire peritoneum and the internal



sorafenib

organs. Implant a tumor fragment of 40 mm3 into abdominal





cavity, spread some sorafenib on the tumor. Spray FS to cover





the tumor. Close body with sutures


4
FS alone (2 ml)
3
Spray FS to cover the entire peritoneum and the internal





organs. Implant a tumor fragment of 40 mm3 into abdominal





cavity. Spray FS to cover the tumor. Close body with sutures


5
FS alone (2 ml)
3
Spray FS to cover the entire peritoneum and the internal





organs. Implant a tumor fragment of 40 mm3 into abdominal





cavity. Spray FS to cover the tumors. Close body with sutures


6
FS + oral
3
Spray FS to cover the entire peritoneum and the internal



application (2 ml)

organs. Implant a tumor fragment of 40 mm3 into abdominal





cavity. Spray FS to cover the tumors. Close body with sutures.





After implantation, the mice were treated according to table2


Total

18
















TABLE 2







treatment schedule in group 6











Medicine
Concentration
Route
Dosage
Schedule





kh afcc
NA
Drink
NA
22 h on 2 h off (D0-D8)


kh afcc
NA
PO
0.4 ml
BID (D9-D31)


KH-R1
NA
Drink
NA
24 h (D32-D42)


5bp121
NA
IP
0.5 ml
QD (D43-D54)









The doses of group6 was changed during the experiment as requested by the sponsor


Experiment Procedures

  • A. Establishment of cell line xenograft tumor model in female Balb/c nude mice: cells were injected at 3×106 cells per animal subcutaneously.
  • B. When the tumor reached about 1000 mm3, sacrifice these animals, removed tumor for peritoneal implantation.
  • C. Fifteen mice were anesthetized by i.p. injection of sodium pentobarbital at 60-70 mg/kg. The animal skin was sterilized with ethanol solution. Then the body wall was opened and the peritoneal surface was exposed.
  • D. The test agent was then applied on the peritoneal surface, including both sides. The amount of the test agent was listed on the above table.
  • E. Tumor fragment was implanted into the peritoneum. The test agent acted as a glue to hold the fragment.
  • F. After the fibrin membrane formed completely, the peritoneal cavity was closed and sutured.
  • G. In the positive control group, tumor fragment was embedded in Matrigel.
  • H. Body weight was measured upon completion of surgery.
  • I. Postoperative cares followed protocol SOP-BEO-0016-1.0.
  • J. Group 1, 2 and 4 were sacrificed 4 weeks after implantation with tumor growing in all vehicle and positive control. Group 3, 5 and 6 were for long term study for 54 days.
  • K. During the period of the experiment, health conditions of mice were observed daily. Body weight of mice was monitored twice per week.
  • L. Tumor sizes were measured twice per week when tumor could be measured. Tumor volumes (mm3) were obtained by using the following formula: volume=(W2×L)/2 (W, width; L, length in mm of the tumor)
  • M. Mice, which showed a significant loss of body weight (>20%), or which were unable to eat or drink, or exhibit ulceration on the skin/tumor, or the tumor size reaches 2,000 mm3, were euthanized immediately to minimize the pain and distress. Such actions need to notify the sponsor within 24 hrs (48 hrs during the weekends).


Evaluation of the Anti-Tumor Activity


Health conditions of mice were observed daily. Body weights were measured twice per week during the treatment. The ratio of palpable tumors observed in each group was recorded. Group 1, 2 and 4 were sacrificed 4 weeks after implantation with tumor growing in all vehicle or positive control. Group 3, 5 and 6 were for long term study for 54 days. Each tumor and FS was removed and weighed.


Drugs and Materials


FS was provided by RAAS; Matrigel was from BD Biosciences (San Jose, Calif., cat. #356234).


Digital caliper was from Sylvac, Switzerland.


Data Analysis


Ratios of Palpable Tumors Observed in Different Groups


Record the palpable tumors of each mouse observed as an indicator of efficacy


Tumor Weight


Group 1, 2 and 4 were sacrificed 4 weeks after implantation with tumor growing in all vehicle or positive control. Group 3, 5 and 6 were for long term study for 54 days. Each tumor was removed and weighted.


Relative Change of Body Weight (RCBW)


Relative change of body weight (RCBW) was calculated based on the following formula: RCBW (%)=(BWi−BW0)/BW0×100%; BWi was the body weight on the day of weighing and BW0 was the body weight before surgery.


Statistical Analysis


Data was expressed as mean±S.E.;


Results


Ratios of Palpable Tumors Observed in Different Groups


On day 21 after implantation, all mice in vehicle group and all mice in positive group exhibited palpable tumor, while no mice in FS groups exhibited any palpable tumors. On 52 days after implantation, five mice treated with FS did not exhibit any palpable tumor. The summary of ratios of palpable tumors observed in different groups was shown in table 3.


Tumor Weight


On day 28 after implantation, the tumor weight of vehicle group, positive group and FS group were 0.88 g, 1.02 g and 0.06 respectively, the tumor weight was shown in table 4.


Body Weight


Loss of body weight or a sign of toxicity was not found in FS-treated groups.


The effect on body weight was shown in table 5.


Discussion


EL-7404 peritoneal Implantation hepatic cancer model was used to evaluate the anti-cancer efficacy of FS at 2 ml/mouse. On day 21 after implantation, all mice in vehicle group and positive group exhibited palpable tumors, while no mice in FS group exhibited any palpable tumors. On day 28 after implantation, the tumor in vehicle group reached 0.88 g, while FS group was only 0.06 g. On day 52 after implantation, Five mice treated with FS did not exhibit any palpable tumor. The results showed that FS had inhibition on tumor growth.


No body weight loss and toxicity were found in FS-treated groups, which showed FS had no side effect.


Tables









TABLE 3







Ratios of palpable tumors observed in different groups









Days after surgery

















Group
21
24
28
31
35
38
42
45
49
52





Vehicle
3/3
3/3
3/3
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed


Positive
3/3
3/3
3/3
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed


FS (2 ml) +
0/3
0/3
0/3
0/3
0/3
0/3
0/3
0/3
0/3
0/3


sorafenib












FS (2 ml)
0/3
0/3
1/3
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed
sacrificed


FS (2 ml)
0/3
0/3
0/3
0/3
0/3
1/3
1/3
1/3
1/3
1/3


FS (2 ml) +
0/3
0/3
0/3
1/3
2/3
2/3
2/3
2/3
2/3
2/3


oral












application
















TABLE 4







The summary of FS weight and tumor weight














Pre-


Days





test


after
FS Weight
Tumor Weight


Group
Mice #
Mice #
status
implantation
(g)
(g)
















Vehicle
1
1-1
sacrificed
28
N/A
0.87



2
1-2
sacrificed
28
N/A
0.68



3
1-3
sacrificed
28
N/A
1.09


Positive
4
2-1
sacrificed
28
N/A
3.56



5
2-2
sacrificed
28
N/A
1.15



6
2-3
sacrificed
28
N/A
1.35


FS (2 ml) +
7
3-1
died
22
1.25
0.00


sorafenib
8
3-2
sacrificed
54
1.22
0.00



9
3-3
sacrificed
54
0.92
0.00


FS (2 ml)
10
4-1
sacrificed
28
1.06
0.10



11
4-2
sacrificed
28
0.72
0.00



12
4-3
sacrificed
28
0.78
0.09


FS (2 ml)
13
5-1
sacrificed
54
0.81
0.75



14
5-2
sacrificed
54
0.80
0.00



15
5-3
sacrificed
54
0.81
0.00


FS (2 ml) +
16
6-1
sacrificed
54
1.08
2.16


oral application
18
6-3
sacrificed
54
0.95
0.00



19
6-2
died
51
0.91
3.03













1) Ovarian Cancer Survival Rate in Mice


Ovarian cancer study has been conducted at Wuxi AppTec under Dr. Yong Cang and the study is still ongoing as of Aug. 4, 2013. However in order to show the comparison mean days of the death of the mice in each group to compare with the positive drug group which has 76.8 days. The vehicle with 128.5 days and two AFOD RAAS 108 from A1AT from fraction IV has 165.2 days. And AFCC RAAS 2 from fraction IV, has 162.3 days.


Mean Survival Time (Days)
















Mean (days)
std error




















AFOD RAAS 1
115.8
12.7



AFOD RAAS 104
114.5
22.0



AFOD RAAS 108
165.2
8.5



AFOD RAAS 109/121
125.3
10.5



AFOD RAAS 110
131.8
5.9



AFOD RAAS 113
110.7
9.2



AFOD RAAS 114
97.8
10.0



AFOD RAAS 120
84.8
5.6



AFCC RAAS 1
97.0
26.9



AFCC RAAS 2
162.3
11.6



positive
76.8
14.2



vehicle
128.5
14.7










Survival curve for AFOD RAAS 1, AFOD RAAS 104, AFOD RAAS 108, AFOD RAAS 109/121. AFOD RAAS 110, AFOD RAAS 113, AFOD RAAS 114, AFOD RAAS 120 AND AFCC RAAS 1, AFCC RAAS 2, POSITIVE AND VEHICLE.



FIGS. 173, 174, 175, 176, 177, 178, 179, 180, 181 and 182


Bioluminescent images of RAAS-20120628-SK-OV-3-luc for all groups.



FIGS. 183—Vehicle group



FIGS. 184—AFOD KH1 (Kieu Hoang AFCC TM Wine). At day 63 after ovarian cancer has started an experiment of Kieu Hoang AFCC TM by oral application of four mice left from the entire group. That means that by day 63 ovarian cancer has already developed. In the beginning we forced the mice to drink without water, therefore the mice died in early stage. Among the four mice, one lasted to 192 days like those mice in AFOD RAAS 8 and AFCC RAAS 2 totaling 6 mice.



FIG. 185—Positive control



FIG. 186—Tested group 1: AFOD RAAS 1 group



FIG. 187—Tested group 2: AFOD RAAS 104



FIG. 188—Tested group 3: AFOD RAAS 108



FIG. 189—Tested group 4: AFOD RAAS 109/121



FIG. 190—Tested group 5: AFOD RAAS 110



FIG. 191—Tested group 6: AFOD RAAS 113



FIG. 192—Tested group 7: AFOD RAAS 114



FIG. 193—Tested group8: AFCC RAAS 1



FIG. 194—Tested group 9: AFCC RAAS 2



FIG. 195—Tested group 10: AFOD RAAS 120


Characterization of Lymphoid Tissues and Peripheral Blood in Nude Mouse Treated with and without AFCC


Executive Summary


The purpose of this study was to investigate the effect of AFCC on curing tumor through characterizing distinct cell lineage in lymphoid tissues and peripheral blood in nude mouse treated with and without AFCC. Distinct cell lineage was differentiated by cell surface marker proteins. T cells, B cells, activated B cells, myeloid dendritic cell (mDC), plasmacytoid dendritic cell (pDC), granulocytes, and monocytes/macrophages were characterized.


In spleen and lymph nodes except in peripheral blood, AFCC treatment resulted in increased CD3+T cell population compared with that in nude mouse with tumor. In spleen, lymph nodes, and peripheral blood, with AFCC treatment, B cell population together with activated B cells also increased compared with those in nude mouse with tumor. In spite of the increased cell number of B cells and T cells after AFCC treatment, granulocytes decreased. Macrophages were found to decrease after AFCC treatment in peripheral blood and spleen but not in draining lymph nodes. mDC and pDC percentages were not greatly affected in nude mouse in the presence of AFCC.


List of Abbreviations


















FACS
Flow Cytometry



mDC
Myeloid dendritic cell



pDC
Plasmacytoid dendritic cell










Materials and Methods


Materials


Reagents


FITC, Rat Anti-Mouse CD4, BD, Cat: 557307


FITC, Rat Anti-MouseCD3 molecular complex, BD, Cat: 561798


PerCP-Cy5.5, Rat Anti-Mouse CD4, BD, Cat: 550954


PE, Rat Anti-Mouse B220/CD45R, BD, Cat: 553089


APC, Rat Anti-MouseCD11b, BD, Cat: 553312


APC, Ar Ham Anti-MouseCD11c, BD, Cat: 550261


PE, Rat Anti-MouseGR-1(Ly-6G and Ly-6C), BD, Cat: 553128


Purified, Rat Anti-MouseFc blocker CD16/32, BD, Cat: 553141


APC, Ar Ham Rat Anti-MouseCD69, BD, Cat: 560689


7-AAD, BD. Cat: 559925


ACK Lysing buffer, Invitrogen, Cat: A10492-01


PBS, Dycent Biotech (Shanghai) CO., Ltd. Cat: BJ141.


FBS, Invitrogen Gibco, Cat: 10099141


Materials


Cell strainer (70 μm), BD, Cat: 352350


BD Falcon tubes (12×75 mm, 5 ml), BD, Cat: 352054


Equipments


Vi-CELL Cell Viability Analyzer, Beckman Coulter, Cat: 731050


FACSCalibur flow cytometer, BD, Cat: TY1218


Methods


Cell Isolation and Staining


Peripheral blood was collected through cardiac puncture. After removing red blood cells with lysis buffer followed by two rounds of washing using 1×PBS, mononuclear cells (monocytes, macrophages, dendritic cells, and lymphocytes) and granulocytes were obtained. Spleen and lymph nodes cell suspension were also obtained after filtering through 70 μm cell strainer. Cell viability and number were analyzed by Vi-CELL Cell Viability Analyzer. Cell surface labeling was performed after that. Blocked with Fc blocker CD16/CD32 at 4° C. for 15 min, cells were centrifuged and resuspended in staining buffer (0.08% NaN3/PBS+1% FBS). Fluorescent-conjugated antibodies were then added into the suspension at the indicated dilution according to the antibody usage protocol from the company. After 30 min incubation at 4° C. for 30 min in the dark, cells were washed twice with 0.08% NaN3/PBS (200 μl per sample), and resuspended with 400 μl 0.08% NaN3/PBS in BD Falcon tubes (12×75 mm, 5 ml) followed by FACS analysis.


Data Analysis


FACS data were analyzed by flowjo software.


Study Summary


Study Initiation Date and Completion Date


The study was initiated and finished on Apr. 13, 2012.


Study Purpose


The purpose of this study was to investigate the effect of AFCC on curing tumor through characterizing distinct cell lineage in lymphoid tissues and peripheral blood in nude mouse treated with and without AFCC.


Study Results


Mice Information


All the mice were transferred from oncology team from Wuxi Apptec. FIG. 1 and FIG. 2 contained the treatment and age information of the mice.


1: Nude mice with tumor: nude mice grafted with MDA-MB-231-Luc tumor cells as vehicle for the study.



FIG. 196


10 nude mice from group 2-5 which have been implanted with tumor cells from the 2-5 mice positive control group using Docetaxel in another study done at another CRO lab.



FIG. 197


3: One of the 10 nude mice with MDA-MB-231-Luc tumor cells transferred from 2-5 positive control group using Docetaxel and it is used as positive control for the re-implantation study,



FIG. 198


Graph showing the tumor volume of Mice #6-10 from the study done from Jul. until Nov. 11, 2011 when the dead body of mouse #6-10 was removed from one CRO lab to another one for further study.



FIG. 199


Mouse #6-10 taken from Aug. 23, 2011 to Nov. 3, 2011 showing the growth of the tumor which had been detached from the body was under recovery from breast cancer using AFCC proteins for treatment.



FIG. 200


The tissue from the area of mouse #6-10 where the tumor had been detached was used to implant in the 10 nude mice 66 days after re-implantations show no tumor growth.



FIG. 201


After 66 days with no growth, then we implanted the cancer tumor for a second time. The growth of the tumor in mice 6-10 which had been treated prior with AFCC at another CRO lab after re-implantation on Nov. 11, 2011.



FIG. 202


Graph showing 5 groups of nude mice after tumor volume change after the second re-implantation with the breast tumor cancer, including mice #6-10 and mice #2-10 treated with Docetaxel.



FIG. 203


The picture of the 10 mice in group #6-10 showing mice #5-1 and mice #5-3 growing the tumor after second re-implantation both had been treated with AFCC on Feb. 29, 2012.



FIG. 204


2: Nude Mice with AFCC Treatment:

    • Grafted with tumor cells numbered #6-10 starting at Nov. 11, 2011; received with AFCC provided by RAAS though I.V. or I.P. injection from Feb. 29, 2012. In April mice #6-10 with the second re-implantation has been completely recovered due to the AFCC proteins which contain good healthy cells which sent signal to the DNA of the infected mice with breast cancer tumor, to transform the RNA to synthesize good proteins against the breast cancer cell.



FIG. 205.


Among the groups in the study for breast cancer from mid-Jul. to Nov. 11, 2011 nude mouse #4-6 has shown the quickest recovery period within 24 days. From day 15 when the tumor started to grow to day 39 when the tumor detached from the body.



FIG. 206


Mouse #4-6 grew the tumor on August 23rd and self-detached from the body Sep. 1, 2011.



FIG. 207


Mouse #4-6 on October 18th completely recovered from breast cancer due to the AFCC KH protein which contains good healthy cells which sent signal to the DNA of the infected mice with breast cancer tumor, to transform the RNA to synthesize good proteins against the breast cancer cell.



FIG. 208


The 9 mice from the #4-6 group first re-implantation of the tumor which had never grown and one of these mice #4 was used in this study for analysis of the cells.



FIG. 209


4: Nude mouse with no tumor: grafted with tumor cells numbered #4-6 starting at Nov. 18, 2011, no further treatment needed due to failure of the tumor growth because good healthy cells from the AFCC treated, which contains good healthy cells which sent signal to the DNA of the infected mice with breast cancer tumor, to transform the RNA to synthesize good proteins against the breast cancer cell.



FIG. 210


5: Nude naïve mouse at 8 weeks old was used as a negative normal control to determine the normal nude mice cells.



FIG. 211


6: C57BL/6 mouse at 8 weeks old was used as a negative normal control to determine the normal nude mice cells.



FIG. 212


Cell Population in Peripheral Blood


After whole blood withdrawal, distinct cell lineage was differentiated by cell surface marker proteins. T cells, B cells, activated B cells, mDC, pDC, granulocytes, and monocytes/macrophages were characterized (FIG. 3 to FIG. 8).


As shown by FIG. 3, AFCC treatment didn't affect CD3+T cell population compared with that in nude mouse with tumor and without tumor. After AFCC treatment, B cell population, on the other hand, increased to the similar percentage as seen in nude mouse no tumor and nude naïve mouse, suggesting the potential effect of AFCC on B cell lineage (FIG. 4). Activated B cells also increased with AFCC treatment, which was illustrated in FIG. 5. Macrophages and granulocytes decreased after AFCC treatment compared with those in nude mouse with tumor (FIG. 6 and FIG. 7). Nude mouse no tumor and nude mouse with AFCC treatment had similar mDC and pDC percentage shown in FIG. 8.



FIG. 213—The percents of B cells in peripheral blood.



FIG. 214—The percents of activated B lymphocytes in peripheral blood.



FIG. 215—The percents of monocytes/macrophages in peripheral blood. CD11b macrophages were analyzed.



FIG. 216—The percents of mDC and pDC in peripheral blood.


Cell Population in Spleen


Distinct cell lineage in spleen cell suspension was further characterized by cell surface marker proteins. T cells, B cells, activated B cells, mDC, pDC, granulocytes, and monocytes/macrophages were included (FIG. 9 to FIG. 14).


As shown by FIG. 9, AFCC treatment slightly increased CD3+T cell population compared with that in nude mouse with tumor and nude mouse without tumor. After AFCC treatment, B cell population, on the other hand, increased to the similar percentage as seen in nude mouse no tumor, suggesting the potential effect of AFCC on B cell lineage (FIG. 10). Activated B cells also increased with AFCC treatment, which was illustrated in FIG. 12. Macrophages and granulocytes dramatically decreased after AFCC treatment compared with those in nude mouse with tumor (FIG. 13 and FIG. 14). Nude mouse no tumor and nude mouse with AFCC treatment had similar mDC and pDC percentage shown in FIG. 11.



FIG. 217



FIG. 218



FIG. 219



FIG. 220



FIG. 221



FIG. 222


Cell Population in Draining Lymph Nodes


Distinct cell lineage in draining lymph nodes suspension was further characterized by cell surface marker proteins. T cells, B cells, activated B cells, mDC, pDC, granulocytes, and monocytes/macrophages were included.


As shown by FIG. 15, AFCC treatment dramatically increased CD3+T cell population compared with that in nude mouse with tumor. T cells in nude mouse with AFCC treatment and mouse no tumor had the similar percentage (FIG. 15). After AFCC treatment, B cell population, on the other hand, increased to the similar percentage as seen in nude mouse no tumor, suggesting the potential effect of AFCC on B cell lineage (FIG. 16). Activated B cells also increased with AFCC treatment, which was illustrated in FIG. 20. Granulocytes dramatically decreased after AFCC treatment compared with those in nude mouse with tumor and naïve nude mouse (FIG. 18). mDC and pDC also decreased in the presence of AFCC compared to those in nude mouse with or without tumor (FIG. 17). Macrophages still maintained the similar percentage with and without AFCC treatment (FIG. 19).



FIG. 223



FIG. 224



FIG. 225



FIG. 226



FIG. 227



FIG. 228


Conclusions


The effect of AFCC on curing tumor through characterizing different cell lineage in lymphoid tissues and peripheral blood in nude mouse was investigated using staining with different marker proteins for distinct cell lineages followed by FACS. T cells, B cells, activated B cells, mDC, pDC, granulocytes, and monocytes/macrophages were characterized in 6 mice illustrated in FIG. 1 and FIG. 2.


FACS analysis showed that AFCC treatment had the effect on the population of major cell lineages in immune system. Increased CD3+T cell population was found in nude mouse treated with AFCC compared with that in nude mouse with tumor in spleen and lymph nodes (FIG. 9, 15). B cells including activated B cells also increased compared with that in nude mice with tumor in spleen, lymph nodes, and peripheral blood (FIG. 4, 10, 16, 5, 10, 20). Granulocytes and macrophages, however, were found to decrease after AFCC treatment in peripheral blood and spleen (FIGS. 7, 14, 18, 6, 13, and 19). The decrease as one of the lymphocytes, white blood cells, which are present in the peripheral blood of the nude mice with the breast cancer cell proves that the vehicle and positive control mice when the breast tumor grew the cancer cell have affected the peripheral blood. Even though the mice has not been metastasized. This make the inventor to believe that any cancer tumor grow the cancer cells are already in the peripheral blood.


Cells expressing KH proteins 1—Send signals to the cells contributing to disease, which triggers the synthesis of good proteins transforming these cells into healthy cells; 2—Send signals to the other currently undamaged cells to synthesize healthy proteins, which protect them from being damaged, infected and prone to DNA and other cellular alterations; 3—Send signals to the body to synthesize new healthy cells and inhibit them from being affected by intra- and extracellular damaging signals, thus treating and preventing disease, viral and bacterial infection, auto immune disease, neurological disorders, solid and blood cancers, and various other afflictions.


Macrophage populations have been found to decrease after AFCC treatment in peripheral blood and spleen; however it their incidence has not decreased in the vehicle and positive control mice.


Macrophages function in both non specific defense as well as help initiate specific defense mechanisms. Their role is to phagocytose, or engulf and then digest cellular debris and pathogens, either as stationary or as mobile cells. They also stimulate lymphocytes and other immune cells to respond to pathogens. They are present in all living tissues, and have a function in regeneration. The level of macrophages in the vehicle or positive control increases as the RNA of the damaged cells synthesize unhealthy proteins responsible in their contribution to cancer. Cells expressing healthy KH proteins help reduce the incidence and proliferation of breast cancer.


Taken together, this study suggests that AFCC plays a role in reducing tumors by changing the population of major cell lineages in the immune system, including the spleen, lymph nodes and peripheral blood.


Efficacy Study of AFOD RAAS 1 (APOA1) on Atherosclerosis:

    • 1) APOE KO mice
    • 2) LDLR KO mice
    • 3) Rabbit


This study performed on APOE KO and LDLR KO mice and finally rabbits has shown, in APOE KO and LDLR KO mice, APOA1 is effective in the reduction and prevention of atherosclerosis.



FIG. 229—APOE KO mice the area of atherosclerosis


The inhibition of inflammation factors RNA transcription.



FIG. 230


















Genebank


Gene
Forward
Reverse
ID







Lp-
GAGCGTCTTCGTGCGTTTG
GCGGGTATTTTTCTCCAGTC
NM_013737


PLA2








MCP-1
CCTGCTGTTCACAGTTGCC
TGTCTGGACCCATTCCTTCT
NM_011333





ICAM-
GCTGTATGGTCCTCGGCTG
GCCCACAATGACCAGCAGTA
NM_010494


1








VCAM-
TGAACCCAAACAGAGGCAGA
CGGAATCGTCCCTTTTTGTAG
NM_011693


1








MMP-2
CAAAGAAAGGTGCTGACTGT
GAAGGAAACGAGCGAAGG
NM_008610





MMP-9
CAGCCAACTATGACCAGGAT
TGCCGTCTATGTCGTCTTTA
NM_013599





TNF-α
GGGCAGGTCTACTTTGGAG
AGCCCATTTGAGTCCTTGAT
NM_13693





β-Actin
GGGAAATCGTGCGTGACA
CAAGAAGGAAGGCTGGAAAA
NM_007393










FIG. 231—The increase of pre-B HDL after injection of APOA1 Protein—Western blot of pre-b HDL



FIG. 232, 233—There is no antibody of APOA1 raised in mice



FIG. 234, 235—Lipid change in LDLR knock out mice



FIG. 236—LDLR KO mice—the area of atherosclerosis


AFOD RAAS 1 (APOAI) Efficacy Pilot Study in 6-OHDA Rat PD Model


Animal Model


I. Animal Description

    • Species: Rat
    • Strain: SD rat, SPF grade
    • Age or weight: 6 to 8 weeks
    • Sex: Male


II. Experiment Outline

    • Objectives: To test the effects of AFOD RAAS 1 (APOA1) product on 6-OHDA rat model of Parkinson's disease (PD).
    • Experiment design:
    • 6-OHDA Lesion:
    • Each rat will receive an injection of 50 μg 6-OHDA into the both sides of medial forebrain bundle to induce severe lesion n of the dopamine system. This high dose will cause the death of the rats at around 15 days after lesion. RAAS product will be injected daily right after lesion.
    • Group: vehicle, n=6; AFOD, n=6


III. Preliminary Result

    • One rat survived in AFOD group and all vehicle rats died. The second batch of animal experienced the same mortality. The body weight of AFOD group rat increased significantly.
    • Then rats were sent to Ruijin Hospital for PET/CT scanning to determine the function recovery of damaged brain.
    • Equipment: Siemens Inveon
    • PET/CT scanning results: There has been signal improvement in the brain of the two rats left, in the first batch and in the second batch to compare with the two normal control rats.



FIG. 237—PET/CT scans



FIG. 238—PET/CT scans



FIG. 239—PET/CT scans



FIG. 240—PET/CT scans



FIG. 241—PET/CT scans



FIG. 242—PET/CT scans

Claims
  • 1. A method of treating or preventing disease and infection in a mammal comprising, administering to the mammal a composition, compound, or solution containing an effective amount of at least one isolated purified plasma product selected from the group consisting of: cryoprecipitate;fraction III;fraction III-II;fraction IV;prothrombin;human factor VIII;human fibrinogen;human immunoglobulin;human thrombin;human albumin; andtransferrin.
  • 2. The method according to claim 1 wherein the at least one isolated purified plasma product further comprises at least one protein defined by an amino acid sequence selected from the group consisting of: SEQ ID NOs 1-55.
  • 3. A method of treating human immunodeficiency virus comprising, administering to an individual infected with HIV a composition, compound, or solution containing an effective amount of a blood plasma product comprising purified factor II, purified factor VII, and purified factor X.
  • 4. The method according to claim 3, wherein the composition, compound, or solution has a concentration of at least 3.5% of at least one protein defined by an amino acid sequence selected from the group consisting of SEQ ID NOs 1-55.
  • 5. The method according to claim 3, wherein composition, compound, or solution has a concentration of at least 400 ug/ml.
  • 6. A method of treating human immunodeficiency virus comprising, administering to an individual infected with HIV a composition, compound, or solution containing an effective amount of a blood plasma product comprising purified antithrombin III and at least one protein defined by an amino acid sequence selected from the group consisting of: SEQ ID NO: 21;SEQ ID NO: 22;SEQ ID NO: 23;SEQ ID NO: 24;SEQ ID NO: 25;SEQ ID NO: 26;SEQ ID NO: 27;SEQ ID NO: 48;SEQ ID NO: 49; andSEQ ID NO: 50.
  • 7. The method of claim 6, wherein the composition, compound, or solution has a concentration of at least 15% of at least one protein selected from the group consisting of: CP 98 kDa;CP Ceruloplasmin;KRT2 Keratin, type II cytoskeletal 2 epidermal;APOA1;human albumin;transferrin;vimentin; andHaptoglobin.
  • 8. A method of treating hepatitis C virus in a mammal comprising, administering to said mammal a composition, compound, or solution containing an effective amount of a blood plasma concentrate containing: CP 98 kDa;CP Ceruloplasmin;KRT2 Keratin, type II cytoskeletal 2 epidermal;APOA1;human albumin;transferrin; andhaptoglobin.
  • 9. The method of claim 8, wherein the blood plasma concentrate comprises at least 10% of the composition, compound, or solution.
  • 10. The method of claim 8, wherein the blood plasma concentrate has a concentration of at least 400 ug/ml.
  • 11. A method of treating hepatitis C virus in a mammal comprising, administering to said mammal a composition, compound, or solution containing an effective amount of a blood plasma concentrate comprising factor II, factor VII, factor IX, and factor X.
  • 12. The method of claim 11, wherein the blood plasma concentrate comprises at least 4% of the composition, compound, or solution.
  • 13. The method of claim 11, wherein the blood plasma concentrate has a concentration of at least 400 ug/ml.
  • 14. A method of treating hepatitis B virus in a mammal comprising, administering to said mammal a composition, compound, or solution containing an effective amount of a blood plasma concentrate comprising: CP 98 kDa;CP Reuloplasmin;KRT2 Keratin, type II cytoskeletal epidermal;a protein defined by amino acid sequence SEQ ID NO: 22;a protein defined by amino acid sequence SEQ ID NO: 23;a protein defined by amino acid sequence SEQ ID NO: 24;a protein defined by amino acid sequence SEQ ID NO: 25;APOA1;human albumin;transferrin;vimentin; andhaptoglobin.
  • 15. The method of claim 14, wherein the blood plasma concentrate has a concentration of at least 1.25 ug/ml.
  • 16. The method of claim 14, wherein the blood plasma concentrate has a concentration of at least 10 ug/ml.
  • 17. A method of treating hepatitis B virus in a mammal comprising, administering to said mammal a composition, compound, or solution containing an effective amount of a blood plasma concentrate derived from fraction III IVIG, the blood plasma concentrate derived from fraction III IVIG comprising TF serotransferrin.
  • 18. The method of claim 17, wherein the blood plasma concentrate comprises at least 25% of the composition, compound, or solution.
  • 19. A method of treating influenza in a mammal comprising, administering to said mammal a composition, compound, or solution containing an effective amount of a purified plasma product, wherein said purified plasma product is selected from the group consisting of: a first protein concentrate comprising proteins CP 98 kDa, CP Ceruloplasmin, KRT2 Keratin-type II cytoskeletal 2 epidermal, APOA1, human albumin, transferrin, vimentin, and haptoglobin; anda prothrombin complex protein concentrate comprising proteins factor II, factor VII, factor IX, and factor X.
  • 20. The method of claim 19, wherein an effective dose of the purified plasma product is administered to the mammal for at least two weeks.
  • 21. The method of claim 19, wherein the first protein concentrate comprises at least 10% of the composition, compound, or solution.
  • 22. The method of claim 19, wherein the prothrombin complex protein concentrate comprises at least 0.0020% of the composition, compound, or solution.
  • 23. The method of claim 19, wherein the first protein concentrate has a concentration of at least 69.06 ug/ml.
  • 24. A method of treating diabetes mellitus in a mammal comprising, administering to said mammal a composition, compound, or solution containing an effective amount of a purified plasma product, wherein the purified plasma product is selected from the group consisting of: a first protein concentrate comprising protein 1CP 98 kDa, wherein protein 1CP98 kDa containing Nup98 and Nup96;a second protein concentrate comprising transferrin; anda third protein concentrate comprising CP 98 kDa, CP Ceruloplasmin, KRT2 Keratin type II cytoskeletal 2 epidermal, APOA1, human albumin, transferrin, vimentin, and haptoglobin.
  • 25. The method according to claim 24, wherein the second protein concentrate further comprises at least one protein having an amino acid sequence selected from the group consisting of SEQ ID NOs 21-27 and 48-50.
  • 26. The method of claim 24, wherein the first protein concentrate comprises at least 0.05% of the composition, compound, or solution.
  • 27. The method of claim 24, wherein the second protein concentrate comprises at least 0.1% of the composition, compound, or solution.
  • 28. The method of claim 24, wherein third protein concentrate comprises at least 0.1% of the composition, compound, or solution.
  • 29. A method of treating and preventing atherosclerosis and related cardiovascular diseases comprising, administering to an individual a daily dose of a composition, compound, or solution containing an effective amount of purified Apolipoprotein A-1 for at least 16 weeks.
  • 30. The method of claim 29, wherein Apolipoprotein A-1 comprises at least 5% protein CPD by concentration.
  • 31. A method of treating cancer in a mammal comprising, administering to said mammal a composition, compound, or solution containing an effective amount of at least one plasma product selected from the group consisting of: high concentrated fibrinogen, enriched a1at, thrombin, and AFOD.
  • 32. The method according to claim 31 further comprising: a) surgically exposing a tumor; andb) coating the tumor and a peritoneal surface surrounding the tumor with the composition, compound, or solution containing the at least one plasma product.
  • 33. A method of treating cancer in a mammal comprising, administering to a mammal a composition, compound, or solution containing an effective amount of high concentrated fibrinogen enriched a1at thrombin and AFOD.
  • 34. The method according to claim 33 further comprising: a) surgically exposing the tumor; andb) coating the tumor and a peritoneal surface surrounding the tumor with the composition, compound, or solution containing the at least one plasma product.
  • 35. The method according to claim 33, wherein the cancer being treated is diagnostically associated as colorectal, lung, hepatic, ovarian, or breast in origin.
  • 36. The method according to claim 35, wherein the cancer being treated is diagnostically associated as colorectal, lung, hepatic, ovarian, or breast in origin.
  • 37. A method of treating Parkinson's disease in a mammal comprising: a) performing a PET/CT scan on said mammal to determine baseline brain function;b) intravenously administering an effective daily dose of a ApoAI to said mammal;c) performing at least one additional PET/CT scan;d) determining whether brain signal function has improved in a time period between performing the at least one additional PET/CT scan and the previous PET/CT scan; ande) discontinuing intravenous administration of ApoAI to said mammal once it is determined that brain signal function has not improved in the time period between performing the at least one additional PET/CT scan and the previous PET/CT scan.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority under 35 USC 119(e) to provisional applications Nos. 61/926,286 and 61/926,287, both filed on Jan. 11, 2014, which are incorporated herein by reference in their entireties. This application is also a continuation in part of non-provisional application Ser. No. 13/756,478, filed on Jan. 31, 2013, which claims priority to provisional application No. 61/593,164, filed on Jan. 31, 2012, provisional application No. 61/593,183, filed on Jan. 31, 2012, provisional application No. 61/593,196, filed Jan. 31, 2012, provisional application No. 61/648,281, filed on May 17, 2012, provisional application No. 61/692,273, filed on Aug. 23, 2012 and provisional application No. 61/710,930, filed on Oct. 8, 2012, all of which are hereby incorporated herein by reference in their entireties.

Provisional Applications (7)
Number Date Country
61926286 Jan 2014 US
61926287 Jan 2014 US
61593164 Jan 2012 US
61593196 Jan 2012 US
61648281 May 2012 US
61692273 Aug 2012 US
61710930 Oct 2012 US
Continuation in Parts (1)
Number Date Country
Parent 13756478 Jan 2013 US
Child 14471466 US