MXPA06000615A - Therapy-enhancing glucan - Google Patents

Therapy-enhancing glucan

Info

Publication number
MXPA06000615A
MXPA06000615A MXPA/A/2006/000615A MXPA06000615A MXPA06000615A MX PA06000615 A MXPA06000615 A MX PA06000615A MX PA06000615 A MXPA06000615 A MX PA06000615A MX PA06000615 A MXPA06000615 A MX PA06000615A
Authority
MX
Mexico
Prior art keywords
glucan
beta
tumor
mice
cells
Prior art date
Application number
MXPA/A/2006/000615A
Other languages
Spanish (es)
Inventor
Nai Kong V Cheung
Original Assignee
Sloankettering Institute For Cancer Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sloankettering Institute For Cancer Research filed Critical Sloankettering Institute For Cancer Research
Publication of MXPA06000615A publication Critical patent/MXPA06000615A/en

Links

Abstract

This invention provides a method for introducing substances into cells comprising contacting a composition comprsing orally administered beta-glucan with said cells. This invention also provides a method for introducing substances into a subject comprising administering to the subject an effective amount of the above compositions. The substance which could be delivered orally includes but is not limited to peptides, proteins, RNAs, DNAs, chemotherapeutic agents, biologically active agents, plasmids, and other small molecules and compounds. Finally, this invention provides a composition comprising orally administered beta-glucan capable of enhancing efficacy of IgM and different uses of the said composition.

Description

GLUCANO THERAPY IMPROVER RELATED REQUEST This application is a continuation in part of E.U.A. Series No. /621, 027, filed July 16, 2003, the contents of which are incorporated herein by reference in their entirety in this application. Throughout this application, several references are cited. The descriptions of these publications in their entirety are incorporated by reference in this application to more fully describe the state of the art to which this invention pertains.
BACKGROUND OF THE INVENTION This description refers to a method for introducing substances, into cells comprising contacting a composition comprising beta-glucan orally administered with said cells. A feature of this invention provides a method for introducing substances into a subject comprising administering to the subject an effective amount of the above compositions. The substance that could be orally delivered includes but is not limited to peptides, proteins, RNA, DNA, chemotherapeutic agents, biologically active agents and plasmids. Other compounds and small molecules can also be used. Another feature of The present invention is a composition comprising orally administered beta-glucan capable of improving the effectiveness of IgM antibodies. Glucans derived from yeast cell walls, such as Saccharomyces cervisiae or mutant yeast strains described in the U.S. Pat. No. 5,250,436, the disclosure of which is incorporated herein in its entirety by reference, may be used in the above compositions. Glucans having β (1-3) and β (1-6) linkages can be prepared by the process described in the U.S. Patents. No. 5,233,491 and 4,310,646, the description of which is hereby incorporated in its entirety. Soluble or aqueous glycans that are suitable for oral administration can be produced by the process described in the U.S. Patents. No. 4,810,646 and 5,519,008, the disclosure of which is incorporated herein by reference in its entirety. Beta-glucans have been tested for tumor therapy in mice for almost 40 years1,2. Several forms of beta-glucan derived from mushrooms are used clinically to treat cancer in Japan, including PSK (from Coriolus versicolor), lentinan and schizophyllan. In randomized trials in Japan, PSK has moderately but significantly improved survival rates in some cancer trials: after gastrectomy3,4, colorectal surgery5'6, and esophagectomy7 to remove primary tumors. The results have been less encouraging in breast cancer8,9 and leukemia10. Schizophyllene has improved the survival of patients with operable gastric cancer11, gastric cancer inoperable 'and cervical cancer. Again, although the differences in survival between the groups were statistically significant, these improvements were moderate. While beta-glucans are not widely used by Western oncologists, botanical medicines containing beta-glucan such as Reishi and Maitake15 are widely used by cancer patients in E.U.A. as alternative / complementary therapies for cancer. These previous studies looking for a therapeutic effect of beta-glucan, did not incorporate the co-administration of therapeutic monoclonal antibodies (MoAb) as part of the protocol. There is increasing evidence that the antibody is necessary to deposit iC3b that acts as a potent opsonine of human tumors. When beta-glucan is administered without co-administration of MoAb, its cytotoxic tumor effect requires the presence of naturally occurring antitumor antibodies that can be highly variable among patients and even in experimental mice. The antitumor effect of beta-glucan when combined with cancer-specific antibodies was described above. Previous studies have shown that oral beta-glucans derived from barley or oats can greatly improve the antitumor activity of anti-tumor monoclonal antibodies in xenographic models. See Therapy-Enchancing Glucan, International Application No. PCT / US02 / 01276, filed January 15, 2002. Cheung et al., Oral (1-3), (1-4) -beta-glucan syngergizes with anti-ganglioside GD2 monoclonal antibody 3F8 in the therapy of neuroblastoma. Clin Cancer Res. 2002; 8: 1217-1223. Cheung NK et al., Orally administered beta-glucans enhance anti-tumor effects of monoclonal antibodies. Cancer Immunol Immunother. 2002; 51: 557-564. The Phase I clinical trial supports the prediction that barley beta-glucan may improve the effect of antibody on metastatic cancer. As indicated above, lentinan and laminarin, both (1-> 3), (1- »6) - ß-D-glucans, were not as effective as barley glucans16. In addition, among the (1-> 3), (1-4) - ß-D-glucans, the small molecular weight preparations and Lichenans were not as effective. The molecular size and fine structure of beta-glucan can have a substantial influence on its synergistic effect on antibodies to tumors. In Europe and the USA, beta-glucans, especially baking yeast, have long been used as an additive for animal feed, as human dietary supplements17, in the treatment of wounds18, and as an active ingredient in skin cream formulations. . The basic structural unit in beta-glucans is the β (1- 3) -linked glucosyl units. Beta-glucans have several degrees of branching and linkage in the side chains depending on the source and method of isolation. The frequency and hinge structure of the side chains determines its immunomodulatory effect. The beta-glucans of fungal and yeast origin are normally insoluble in water, but can be made soluble either by acid hydrolysis or by derivation by introducing charged groups such as -phosphate, -sulfate, -amine, -carboxymethyl, etc., to the molecule19,20. The soluble glucan with the molecular structure in which the units of (1? 3) -β-D-glucan form the base structure with branches constituted by units of (1? 3) -β-D-glucan located in hinges of ( 1- »6) -β-D-glucan were isolated from baking yeast, Saccharomyces cervisiae. High molecular weight fractions are obtained and tested for synergy with monoclonal antibodies in tumor models. The antitumor effect of soluble yeast beta-glucan was found to be comparable to the antitumor effect of soluble barley beta-glucan, when combined with monoclonal antibodies specific for human cancer as detailed below.
BRIEF DESCRIPTION OF THE INVENTION This invention provides a method for introducing substances into cells comprising contacting a composition comprising beta-glucan orally administered with said cells.
Another aspect of the present invention is a method for introducing substances into a subject which comprises administering to the subject an effective amount of the above compositions. The substance that could be delivered orally includes but is not limited to peptides, proteins, RNA, DNA chemotherapeutic agents, biologically active agents, and plasmids. Other compounds and small molecules can also be used. A further aspect of the present invention is a composition comprising orally administered beta-glucan capable of improving the efficacy of IgM antibodies.
DETAILED DESCRIPTION OF THE FIGURES Figures 1A and 1B (1-> 3), (1? 4) -β-D-qlucan plus antibody in the treatment of metastatic neuroblastoma in patients. The scrutiny of MIBG before and after treatment in a patient with metastatic neuroblastoma refractory to multiple chemotherapy regimens. Patients received intravenous anti-GD2 3F8 antibody (10 mg / m2 / day) for a total of 10 days, plus oral barley beta-glucan during the same period. Figure 1A shows MIBG patient baseline scan. Extensive bone metastasis can be seen in the femoral, fibula, pelvis, ribs, left scapula, right clavicle, humerus, skull, and spine. The heart, liver, stomach and colon are physiological. Figure 1B shows MIBG scrutiny of the same patient 2 months later, following an individual cycle of therapy with 3F8 plus glucan. The areas of metastasis have improved significantly. Figures 2A to 2C (1? 3), (1- >; 4) -β-D-giucan from barley plus antibody in the treatment of subcutaneous human lymphoma xenografted in SCID mice. SCID mice with Daudi xenografts (n = 9) (Figure 2A), Hs445 (n = 5) (Figure 2B), LCL derived from EBV LCL (n = 9) (Figure 2C) and RPMI 6666 (n = 10; no established subcutaneous data are used) were treated with either 200 ug intravenous rituximab twice a week for 8 doses (__.), 400ug (1- »3), (1-> 4) -β-D- Glucan administered orally by intragastric forced administration daily for 29 days (?) or a combination of rituximab and (1-> 3), (1-4) -β-D-glucan (x), or left untreated (). The percentage of tumor growth is plotted on the axis and the days after treatment start on the x axis. The error bars represent SEM and has been shown only for rituximab alone and combination groups. For all xenografts, only the combination treatment was associated with reduction in tumor growth. The reduction in tumor growth per day in the group that received beta-glucan in addition to rituximab compared to rituximab alone was 2.0% (95% CI 1.3-2.7%, p <= 0.0005) for Daudi, 0.8% for LCL derived of EBV (95% CI 0.4-1.2%, p <001), 2.2% for Hs445 (95% CI1.2% -3.2%, p = 0.0009), and 1.8% for RPMI6666 (95% Cl 1.0-2.7%; p <0.0002, no data shown) xenografts. Figures 3A to 3B. (1? 3), (1- »4) -β-D-glucan plus antibody in the treatment of disseminated human lymphoma xenografted in SCID mice. 5 × 10 6 Daudi (Fig. 3A) or Hs445 (Fig. 3B) cells in 100 μl of normal saline were injected intravenously (IV) into SCID mice. Mice were treated with either 200 ug of intravenous rituximab twice a week for 8 doses (---), 400ug (1-3), (1-4) -β-D-glucan administered orally by intragastric forced administration daily for 29 days (...) or a combination of rituximab and (1? 3), (1- »4) -β-D-glucan (-), or left untreated (____) beginning 10 days after tumor implant. The tumors grew systematically and the mice were paralyzed when the tumor cells infiltrated the spinal canal, resulting in paralysis in the hind legs. The mice were sacrificed at the onset of paralysis or when the animals lost 10% of their body weight. The Kaplan-Maier survival curves for the various groups are shown in Figures 2A (Daudi) and 2B (Hs445). Mice treated with a combination of (1? 3), (1-4) -β-D-glucan and rituximab had a significantly increased survival when compared to all other treatment groups (p <0.0005 for Daudi and p = 0.001 for Hs445) or when compared with rituximab alone (p <0.0005 for Daudi and p = 0.01 for Hs445). The median survival for untreated mice, rituximab alone, BG, and rituximab + BG groups was 27, 71, 43 and 124 days respectively for Daudi xenografts, and 12, 16, 31 and 243 days respectively for xenografts of Hs445. Figure 4. Response to the dose of 3G6 (anti-GD2 IgM antibody) in the presence of barley beta-glucan in the treatment of human neuroblastoma. Two million LAN1 neuroblastoma cells were xenografted subcutaneously in athymic Balb / c mice. Treatment started in groups of 5 mice each, 2 weeks after tumor implantation when visible tumors reached 0.7-0.8 cm in diameter. The 3G6 group (solid frames) was treated with 200 ug of intravenous 3G6 grafted through the retro-orbital plexus twice a week (M and Th). The 3G6 + BG group was treated with 200 ug i.v. 3G6 twice a week plus oral beta-glucan (BG) 400 ug daily by forced feeding for a total of 14-18 days. 3G6 was administered in 3 different doses (open triangle d ug per dose, open chart 40 ug, open circle 200 ug). The BG group (solid circle) received 400 ug of oral beta-glucan alone. The size of the tumor was measured from the first day of treatment, and the product of the largest diameters was expressed as a percent of the size on day 0 of treatment. The vertical bars represented standard errors, illustrated only in 4 groups for clarity. Although BG alone and 3G6 only showed no antitumor effect, the BG + 200 ug 3G6 group showed shrinkage and its highly significant tumor growth dependent on the 3G6 dose (p < 0.05).
Figure 5. Treatment of human neuroblastoma using 3G6 (anti-GD2 IgM antibody) in the presence of yeast (1- »3), (1-» 6) -β-D-glucan. Two million LAN1 neuroblastoma cells were xenografted subcutaneously in athymic Balb / c mice. The treatment started in groups of 5 mice each, 2 weeks after tumor implantation when visible tumors reached 0.7-0.3 cm in diameter. The 3G6 group (solid frames) was treated with 200 ug of intravenous 3G6 grafted through the retro-orbital plexus twice a week (M and Th) for a total of 5 doses. The particle yeast glucan group (solid triangles) received 400 ug of oral particulate glucan alone. 3G6 + whole yeast particles (open diamonds) was treated with 200 ug i.v. 3G6 twice a week plus yeast particles of 400 ug daily by forced feeding for a total of 14-1 d days. The 3G6 + soluble yeast glucan group was treated with 200 ug i.v. 3G6 twice a week plus soluble yeast glucan 400 ug daily by forced feeding for a total of 14-16 days. The group of 3G6 + particulate yeast glucan was treated with 200 ug i.v. 3G6 twice a week plus glucan in particles 400 ug daily by forced feeding for a total of 14-13 days. The tumor size was measured from the first day of treatment, and the product of the largest diameters was expressed as a percent of size on day 0 of treatment. The vertical bars represent standard errors, illustrated only in 4 groups for clarity. Although glucan alone and 3G6 alone showed no effect antitumor, the soluble and particulate yeast glucan when combined with the 3G6 group showed shrinkage and its highly significant tumor pressure (p <0.05). Figure 6. Treatment of human neuroblastoma using 3Fd (anti-GD2 IgG antibody) in the presence of barley and yeast beta-glucan. Two million LAN1 neuroblastoma cells were xenografted subcutaneously in athymic Balb / c mice. Treatment started in groups of 5 mice each, 2 weeks after tumor implantation when visible tumors reached 0.7-0.d cm in diameter. The 3F3 group (solid diamonds) was treated with 200 ug of intravenous 3F8 grafted through the retroorbital plexus twice a week (M and Th) for a total of 5 doses. The barley glucan group (solid cadres) received 400 ug of barley glucan alone. The 3F3 + barley glucan group (open diamond) was treated with 200 ug i.v. 3Fd twice a week plus barley glucan 400 ug daily by forced feeding for a total of 14-1 d days. The 3F3 + soluble yeast glucan group (open squares) was treated with 200 ug i.v. 3F3 twice a week plus soluble yeast glucan 400 ug daily by forced feeding for a total of 14-18 days. The size of the tumor was measured from the first day of treatment, and the product of the largest diameters was expressed as a percent of the size of day 0 of treatment. Vertical bars represent standard errors. Although glucan alone and 3F3 alone did not show antitumor effect, glucan from barley and soluble yeast when combined with the 3F3 group showed highly significant shrinkage and tumor suppression (p <0.05). Figure 7. Treatment of dissemination of human lymphoma in SCID mice using Rituxan and barley or yeast beta-glucan. 5x10e6 Daudi cells in 100 μl of normal saline were injected intravenously (IV) into SCID mice. The tumors grew systematically and the mice were paralyzed when the tumor cells infiltrated the spinal canal, resulting in paralysis in the hind legs. The mice were sacrificed at the onset of paralysis or when the animals lost 10% of their body weight. The therapy was started ten days after the injection of the tumor cells. 40 μg of rituximab (Genentech, San Francisco, CA) was injected intravenously twice a week for a total of eight injections and 400 μg of glucan was administered orally by intragastric forced feeding for 29 days. The mice were weighed weekly and clinically observed at least once a day. Mice that received rituxan plus barley glucan or rituxan plus soluble glucan from yeast had a highly significant prolonged survival (p < 0.05). Figure d illustrates the pEGP-C1 vector purchased from BD Biosciences (Palo Alto, CA). Figure 9 shows that glucan facilitates the transfer of genes into monocytes. Figure 10 illustrates beta-glucan of high molecular weight and gene transfer. Figure 11 illustrates the presence of GFP mRNA in circulating monocytes.
DETAILED DESCRIPTION OF THE INVENTION This invention provides a composition for oral substance absorption comprising an appropriate amount of carbohydrates. In one embodiment, the carbohydrate is glucan. When administered orally, glucan is absorbed by macrophages and monocytes that carry these carbohydrates to the bone marrow and the reticuloendothelial system from where they are released, in an appropriately processed form, on myeloid cells including neutrophils, and on lymphoid cells including natural killer cells (NK) This processed glucan binds CR3 on these neutrophils and NK cells, activating them in tumor cytotoxicity in the presence of tumor-specific antibodies. Since macrophages and monocytes ingest glucan (either soluble, in gel or particles) from the intestine, glucan is a potential conduit for gene therapy. Unlike proteins, DNA or plasmids are relatively stable to heat and can be easily incorporated into warm soluble barley glucan that gels when cooled to room or body temperature. When the mice are fed with These DNA-glucan complexes, reporter genes can be detected in monocytes and peripheral blood macrophages within a few days. More importantly, these reporter genes are expressed in these cells, a few days after the ingestion of these DNA complexes. These findings have potential biological implications. Glucan and similar carbohydrates can be conduits for DNA or plasmids to reach the human body. Oral glucan can be a convenient vehicle for correcting genetic defects of macrophages / monocytes, or administering genetic vaccines. As can be readily appreciated by one skilled in the art, other carbohydrates capable of functioning as a glucan could be identified and used in a similar manner. An easy screening for such carbohydrates can be established using glucan as the positive control. The glucan includes but is not limited to glucan-bond of β (1-3) and β (1-4) mixed, and the glucan is of high molecular weight. The glucan can also have ß (1-3) and ß (1-6) bonds. This invention also provides a method for introducing a substance into cells comprising contacting the above compositions with said cells. Reporter genes or other markers can be used to evaluate the efficiency of the introduction. Reporter genes or markers are well known in the field of molecular biology. In addition, this invention provides a method for introducing substance into a subject comprising administering to the subject an effective amount of the above compositions. This invention provides a composition for the oral delivery of one or more substances comprising an effective amount of an orally administered beta-glucan and one or more chemotherapeutic agents. In one embodiment, the glucan contains mixed 1, 3-1, 6 or 1, 3-1, 4 bonds, or a mixture of mixed 1, 3-1, 6 and 1, 3-1, 4 bonds. In another embodiment, the glucan increases the efficacy of chemotherapeutic agents or anti-cancer antibodies. In a further embodiment, the glucan is derived from herbs, plants, mushrooms, yeast, barley, fungi, wheat or algae. The glucan can be of high molecular weight. The molecular weight of the glucan can be at least 10,000 Daltons. In a further embodiment, the substance is a peptide, protein, RNA, DNA, plasmid or chemotherapeutic agent. As used herein, chemotherapeutic agents include chemicals that fight disease in the body of an animal or drugs used to treat various forms of cancer. This invention provides a method for introducing substance into cells which comprises contacting the composition described above with said cells. The substance that could be orally delivered includes but is not limited to peptides, proteins, RNA, DNA and plasmids. I also know they can use other compounds and small molecules. This invention provides a method for treating a subject comprising administering to the subject an effective amount of the above composition. In one embodiment, the method further comprises the substance. This invention provides a method for treating a subject with a genetic disorder comprising administering to the subject an effective amount of the composition described above and a substance capable of correcting said genetic disorder. The substance includes but is not limited to one peptide, protein, RNA, DNA, plasmid and another small molecule and compound. This invention provides a composition comprising an effective amount of orally administered (1? 3), (1? 6) beta-glucan capable of improving the efficacy of IgM antibodies. This invention provides a composition comprising an effective amount of (1? 3), (1-> 6) orally administered beta-glucan capable of improving the effectiveness of antibodies. Glucans derived from yeast cell walls, such as Saccharomyces cervisiae or mutant yeast strains described in U.S. Pat. No. 5,250,436, the disclosure of which is incorporated herein by reference in its entirety, may be used in the above compositions. Glucans having β (1-3) and β (1-6) linkages can be prepared by the process described in the U.S. Patents. No. 5,233,491 and 4,610,646, the disclosure of which is hereby incorporated by reference in its entirety. Soluble or aqueous glycans that are suitable for oral administration can be produced by the procedure described in the patents of E.U.A. No. 4,810,646 and 5,519,009, the disclosure of which is incorporated herein by reference in its entirety. In one embodiment, the antibody is a monoclonal antibody, or an antibody against cancer or tumor cells, including but not limited to anti-CEA antibody, anti-CD20 antibodies, anti-CD25 antibodies, anti-CD22 antibodies, anti-CD26 antibodies, HER2, anti-tenascin antibodies, MoAB M195, Dacluzimab, anti-TAG-72, R24, Herceptin, Rituximab, 528, IgG, IgM, IgA, C225, Epratuzumab, and MoAb 3F3 antibodies. In another embodiment, the antibody is a tumor binding antibody. Moreover, the antibody is capable of activating complement and / or activating antibody-mediated cell-mediated cytotoxicity. In another embodiment, the antibody modulates the function of T cells or B cells. In a further embodiment, the antibody is targeted at the epidermal growth factor receptor, a ganglioside, such as GD3 or GD2. In a further embodiment, the antibodies are effective against cancers including neuroblastoma, melanoma, non-Hodgkin's lymphoma, Epstein-Barr-related lymphoma, Hodgkin's lymphoma, retinoblastoma, small cell lung cancer, brain tumors, leukemia, squamous cell carcinoma, cancer of prostate, renal cell carcinoma, transitional cell carcinoma, breast cancer, ovarian cancer, lung cancer, colon cancer, liver cancer, stomach cancer, or other gastrointestinal cancers.
In a further embodiment, the composition described above is in a pharmaceutically acceptable carrier. This invention provides a method for treating a subject comprising administering the above-described composition to a subject. This invention provides a composition comprising an effective amount of orally administered (1-3), (1- * 6) beta-glucan capable of improving vaccine efficacy. In one embodiment, the vaccine is against cancer or infectious agents, such as bacteria, viruses, fungi or parasites. This invention provides a composition comprising an effective amount of (1 ~ »3), (1? 6) beta-glucan capable of improving the efficacy of natural antibodies or infectious agents. This invention provides a composition comprising an effective amount of orally administered (1-> 3), (1-6) beta-glucan capable of improving host immunity. This invention provides a composition comprising an effective amount of (1- »3), (1-> 6) beta-glucan capable of improving the action of an agent in the prevention of tissue rejection. In one embodiment, the tissue is transplanted tissue or transplanted organ or the host as in graft-versus-host disease. In one embodiment, the glucan of the composition described above has a high molecular weight. The molecular weight of glucan is at least 10,000 Daltons. In another embodiment, the glucan is derived from barley, oats, mushrooms, seaweed, mushrooms, yeast, wheat or moss. In a further embodiment, the glucan is stable to heat treatment. In a further embodiment, the composition described above is stable after boiling for 3 hours. The effective dose of the composition described above is about > = 25 mg / kg / day, five days a week for a total of 2-4 weeks. The invention will be better understood by reference to the experimental details that follow, but those skilled in the art will readily appreciate that the detailed specific experiments are illustrative only, and are not intended to limit the invention as described herein, which is defined by claims that are given below.
EXAMPLE I Phase I study of barley ß-qlucan in combination with anti-GD2 antibody in stage 4 neuroblastoma A total of 24 patients were studied. These patients are all children or adolescents with recurrent or metastatic refractory stage 4 neuroblastoma to bone, bone marrow or distant lymph nodes, some with large soft tissue masses. Beta-glucan was well tolerated without dose-limiting toxicities. Antitumor responses were recorded for bone marrow disease (histology, MIBG scrutiny), soft tissue tumors (CT), as well as biochemical markers (markers of VMA tumor and HVA in the urine). An example of tumor response is shown in Figures 1A and 1B: the scrutiny of 131I-metaiodobenzylguanidine (MIBG) showing almost complete resolution of extensive metastasis after a 3F3 plus beta-glucan treatment cycle. These responses are not common in patients with recurrent or refractory metastatic stage 4 B treated with 3F8 alone or 3F3 in combination with cytokines. The best response regimen for 3F3 to date was the phase II trial of the combination of 3Fd plus GMCSF where 7 of 33 (21%) children achieved MIBG improvement. On the contrary, 62% (13 of 21) of the evaluable patients on 3F8 + beta-gíucano had improvement of MIBG, an almost triplication of the response regimen (p = 0.008 by X2). In addition, among 15 patients with bone marrow disease, 5 achieved remission of complete bone marrow (30%) and d with stable disease in the bone marrow (see figures 1A and 1B).
EXAMPLE II Rituximab activates complement-mediated and antibody-mediated cell-mediated cytotoxicities and is effective against B-cell lymphomas. Beta-glucans are naturally occurring glucose polymers that bind to the lectin domain of CR3, a widely expressed receptor among leukocytes, initiating it to bind to antibody-activated iC3b. (1? 3), (1- »4) - ß-D-glucan derived from barley (BG), when administered orally (400 μg per day x 29 days), strongly synergizes with subtherapeutic doses of intravenous rituximab (200 μg twice / week x 8 doses) in the therapy of CD20-positive human lymphomas. The growth of established subcutaneous non-Hodgkin lymphoma (NHL) (Daudi and B-NHL derived from EBV) or Hodgkin's disease (Hs445 or RPM16666) xenografted in SCID mice was significantly suppressed, when compared with mice treated with rituximab or BG alone. The survival of mice with disseminated lymphoma (Daudi and Hs445) increased significantly. There was no weight loss or clinical toxicity in treated animals. This therapeutic efficacy and lack of toxicity of BG plus rituximab supports further investigation into its clinical utility.
Introduction The chimeric anti-CD20 antibody rituximab is being evaluated in an increasing number of disorders. After clinical efficacy was initially demonstrated against recurrent and refractory follicular / low grade non-Hodgkin lymphoma1, responses to rituximab have been reported in other malignant and non-malignant B cell disorders2. Several mechanisms of action have been proposed, including the activation of apoptotic pathways3, the elaboration of cytokines4, and the extraction of host-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). Although many patients with B cell disorders respond to rituximab, remissions often They are transient6. More than 50% of recurrent lymphomas after treatment with rituximab did not respond the second time7. The mechanisms of resistance to rituximab are not yet clear, and may include a small number of target antigen8, pharmacokinetic variations among individual patients, polymorphism of FcR9, resistance to complement activity10, or inherent gene expression of lymphoma11. Beta-glucans are complex glucose polymers with affinity for the lectin site of the CR3 receptor on leukocytes12. With bound beta-glucan, CR3 (CD11b) is initiated to attach to the iC3b fragments deposited on cells by complement activating antibodies. This receptor mediates leukocyte diapedesis through the endothelium and stimulates phagocytosis, degranulation and tumor cytotoxicity. Many fungi present beta-glucan or CR3-binding ligand similar to beta-glucan on its cell surface. Therefore, when deposition of ¡C3b occurs, both the CD11b and lectin sites' '*' -. and respiratory burst is triggered13. By e! contrary, tumor cells lack such molecules, and even when they are coated with iC3b they do not generally activate CR3 and do not activate leukocytes. Soluble forms of beta-glucan bind to lectin sites and initiate both phagocytic and NK cells to kill tumor targets coated with iC3b14. (1-3), (1? 4) -D-ß-glucan (BG), a soluble beta-glucan derivative has advantages over (1? 3), (1- »6) -β-glucans previously studied, particularly effective when administered orally and a good security profile. The in vivo synergism between BG and the fixed antibody of complement 3F8 against human neuroblastoma xenografts14,16 was recently demonstrated. The synergism between BG and rituximab against lymphoma is not reported now.
Study design Cell lines Line of human Burkitt lymphoma cells, Daudi, and Hodgkin's disease (HD) cell line Hs445 and RPMI 6666 were purchased from the American Type Culture Collection (Rockville, MD). Human EBV-BLCL were established using the previously described methods17.
Mice Fox Chase ICR mice (Taconic, White Plains, NY) were kept under the guidelines and institutionally approved protocols.
Tumor models Simultaneous tumors were established by injecting 5x106 cells suspended in 0.1 ml of Matrigel (Becton-Dickinson, Franklin Lakes, NJ) on mouse flanks. The tumor dimensions were measured two to three times a week and the tumor size was calculated as the product of two large diameters. The mice were sacrificed when the maximum size of the tumor exceeded 20 mm. A disseminated tumor model was established in SCID mice as described previously18. Briefly, 5x106 Daudi or Hs445 cells in 100 μl in saline were injected intravenously into SCID mice. The tumors grew systematically and the mice were paralyzed when the tumor cells infiltrated the spinal cord resulting in paralysis in the hind legs. The mice were sacrificed at the onset of paralysis or when the animals lost 10% of their body weight.
Treatment regimens For mice with subcutaneous tumors, therapy was started after the tumors were established (7-d mm in diameter). For the disseminated tumor model, therapy was initiated ten days after the injection of tumor cells. Groups of at least five mice per treatment regimen received either rituximab, BG, none or both. 200 μg of rituximab (Genentech, San Francisco, CA) was injected intravenously twice a week for a total of eight injections and 400 μg was administered orally by intragastric forced feeding daily for 39 days. Animals were weighed weekly and clinically observed at least once a day.
Statistical analysis Tumor growth was calculated by fitting a regression slope to each individual mouse to values transformed by logarithms of tumor size. The slopes were compared between groups using tests of t by a method described above for census observations19. Survival in mice with disseminated diseases was compared using Kaplan-Meier analysis and the death rate was compared by the Fisher exact X2 test. The analyzes were conducted using STATA 7 (Stata Corporation, College Station, TX).
Results and Discussion In all subcutaneous xenograft models, significant reduction in tumor growth was observed in mice treated with a combination of rituximab and BG. Mice treated with rituximab alone showed a moderate reduction in tumor growth, while those treated with BG alone or that were not treated had non-despondent tumor growth (Figures 1A, 1 B, 1C). All tumors except those treated with combination therapy grew beyond 20 mm in size and the mice had to be sacrificed. Mice in combination treatment had persistent tumor suppression even after the treatment was stopped. In a multivariable linear model of tumor growth rate, using simulation variables for treatment, the interaction between BG and rituximab was positive and significant, demonstrating synergism For disseminated xenografts, there was a significant difference in survival between the combination and control groups for both NHL and HD model (p <0.005, by logarithmic range) (Figures 2A to 2C). 5 / 2d mice and 2/6 mice with disseminated Daudi and Hs445 tumors respectively treated with combination of BG and rituximab survived >12 months after the therapy was discontinued suggesting the complete eradication of the disease. In contrast, 0/29 and 0 / d mice that received rituximab only in respective groups survived (15% vs. 0% survival, X2 = 0.01). There was no significant weight loss or other clinically evident adverse effects. The fact that BG is absorbed can be inferred from the fact that it could be detected intracellularly within fixed peripheral blood leukocytes permeabilized by immunofluorescence (data not shown). In these studies, the synergism between BG and rituximab was highly significant regardless of the type of CD20 positive lymphoma. The improved responses of Daudi xenografts compared to Hs445 can be attributed to higher CD20 expression in the first one (mean geometric fluorescence channel for Daudi 241 compared to 1d4 for Hs445). When progressing tumors were examined for CD20 expression by immunofluorescence studies of individual cell suspensions or indirect immunohistochemistry of frozen sections, no significant difference was observed between the groups treated with rituximab, BG alone or rituximag + BG (no data shown), indicating that treatment with rituximag + BG was not associated with CD20 loss. The synergism between other complement activating monoclonal antibodies and BG15'16 was demonstrated above. Current data extend this observation to rituximab. CDC is considered an important mechanism for cytotoxicity of rituximab. The complement of rodents is not efficiently inhibited by human complement regulatory proteins (mCRP). Therefore, CDC can be an effective antitumor mechanism in xenograft models. However, in one study, subtherapeutic doses of antibody, ADCC and CDC mediated by rituximab were not sufficient to affect the death of tumor cells. Since BG has no direct effect on ADCC20, this synergy is most likely a result of tumor cytotoxicity mediated by iC3b. Lymphoma cells express CRP including CD46, CD55, and CD5910'21. However, the cytotoxicity mediated by iC3b is not affected by the presence of CD59 that only affects MAC22 mediated cytotoxicity. Furthermore, in tumors of human breast carcinoma, the deposition of ¡C3b has been demonstrated despite the presence of mCRP23, which indicates that unlike its inhibitory effect on MAC, the effect on tumor cytotoxicity mediated by IC3b is not absolute. If this synergistic effect can be reproduced safely in humans, cytotoxicity mediated by iC3b may be a potential strategy to overcome resistance of rituximab in patients with B cell malignancies. Since neither T nor B cells are required for this synergistic effect, BG may have a potential role even in patients with immunocompromised lymphoma. In addition, in patients with autoimmune diseases, B-cell depletion can be increased with this non-toxic oral therapy. In contrast, beta-glucans can increase the release of cytokines such as TNF-a and IL-624, and because acute toxicities of rituximab are also related to cytokine release secondary to complement activation25, there is a potential for toxicity increased when BG and rituximab are used in combination. The carefully designed phase I studies are necessary to define the safety and efficacy in the development of BG as an adjunct to rituximab therapy in the treatment of B-cell diseases and in antibody-based therapies of other cancers.
References for Example II 1. Maloney DG, Liles TM, Czerwinski DK, Waldichuk C, Rosenberg J, Grillo-Lopez A, Levy R. Phase I clinical trial using escalating single-dose infusion of chimeric anti-CD20 monoclonal antibody (IDEC- C2B8) in patients with recurrent B-cell lymphoma. Biood. 1994; 84: 2457-2466. 2. Cheson BD. Rituximab: clinical development and future directions. Expert Opin Biol Ther. 2002; 2: 97-110. 3. S wings, Emmanouilides C, Bonavida B. Inhibition of interleukin 10 by Rituximab results in Down-regulation of Bcl-2 and sensitization of B-cell Non-Hodgkin's lymphoma to apoptosis. Clin Cancer Res. 2001; 7: 709-723. 4. Chow KU, Sommerlad WD, Boehrer S, Schneider B, Seipeit G, Rummel MJ, Hoelzer D, Mitrou PS, Weidmann E. Anti-CD20 antibody (IDEC-C2B8, rituximab) enhances the efficacy of cytotoxic drugs on neoplastic lymphocytes in vitro : role of cytokines, complement, and caspases. Haematology 2002; 87: 33-43. 5. Reff ME, Carner K, Chambers KS, Chinn PC, Leonard JE, Raab R, Newman RA, Hanna N, Anderson DR. Defection of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood. 1994; 83: 435-445. 6. McLaughlin P, Grillo-Lopez AJ, Kink BK, Levy R, Czuczman MS, Williams ME, Heyman MR, Bence-Bruckler I, White CA, Cabanillas F, Jain V, Ho AD, Lister J, Wey K, Shen D, Dallaire BK. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients responded to four-dose treatment program. J Clin Oncol. 1998; 16: 2825-2833. 7. Davis TA, Grillo-Lopez AJ, White CA, McLaughlin P, Czuczman MS, Link BK, Maloney DG, Weaver RL, Rosenberg J, Levy R. Rituximab anti-CD20 monoclonal antibody therapy in non- Hodgkin's - lymphoma: safety and efficacy of re-treatment. J Clin Oncol. 2000; 18: 3135-3143. D. Davis TA, Czerwinski DK, Levy R. Therapy of B-cell lymphoma with anti-CD20 antibodies can result in the loss of CD20 antigen expression.
Clin Cancer Res. 1999; 5: 611-615. 9. Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bards P, Colombat P, Watier H. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fe receptor FcgammaRllla gene. Blood. 2002; 99: 754-758. 10. Golay J, Zaffaroni L, Vaccari T, Lazzari M, Borleri GM, Bernasconi S, Tedesco F, Rambaldi A, Introna M. Biologic response of B lymphoma cells to anti-CD20 monoclonal antibody rituximab in vitro: CD55 and CD59 regulate complement -mediated cell lysis. Blood. 2000; 95: 3900-3908. 11. Bohen SP, Troyanskaya OG, Alter O, Warnke R, Botstein D, Brown PO, Levy R. Variation in gene expression patterns in follicular lymphoma and the response to rituximab. Proc Nati Acad Sci U S A. 2003; 100: 1926-1930. 12. Bohn JA, BeMiller JN. (1-3) -B-D-Glucans as biological! response modifiers: a review of structure-functional activity relationships. Carbohydr Polymers. 1995; 28: 3-14. 13. Ross GD, Cain JA, Myones BL, Newman SL, Lachmann PJ. Specificity of membrane complement receptor type three (CR3) for beta-glucans. Complement Inflamm. 1987; 4: 61-74. 14. Xia Y, Vetvicka V, Yan J, Hanikyrova M, Mayadas T, Ross GD. The beta-glucan-binding lectin site of mouse CR3 (CD11b / CD18) and its function in generating a primed state of the receptor that mediates cytotoxic activation in response to iC3b-opsonized target cells. J Immunol. 1999; 162: 2261-2290. 15. Cheung NK, Modak S. Oral (1-3), (1-4) -beta-glucan syngergizes with anti-ganglioside GD2 monoclonal antibody 3F3 in the neuroblastoma therapy. Clin Cancer Res. 2002; d: 1217-1223. 16. Cheung NK, Modak S, Vickers A, Knuckles B. Orally administered beta-glucans enhance anti-tumor effects of monoclonal antibodies. Cancer Immunol Immunother. 2002; 51: 557-564. 17. Koehne G, Gallardo HF, Sadelain M, O'Reilly RJ. Rapid selection of antigen-specific T lymphocytes by retroviral transduction. Blood. 2000; 96: 109-117. 18. Wei BR, Ghetie MA, Vitetta ES. The combined use of an immunotoxin and radioimmunoconjugate to treat disseminated human B-cell lymphoma in immunodeficient mice. Clin Cancer Res. 2000; 6: 631-642. 19. Vardi Y, Ying Z, Zhang C-H. Two-sample tests for growth curves under dependent right censoring. Biometrika 2001; 88: 949-960. 20. Yan J, Vetvicka V, Xia Y, Coxon A, Carroll MC, Mayadas TN, Ross GD. B-glucan a "Specific" biologic response modifier that uses antibodies to target tumors for cytotoxic recognition by leukocyte complement receptor type 3 (CD11 b / CD18). J Immunol. 1999; 163: 3045-3052. 21. Treon SP, Mitsiades C, Mitsiades N, Young G, Doss D, Schlossman R, Anderson KC. Tumor cell expression of CD59 is associated with resistance to CD20 serotherapy in patients with B-cell malignancies. J Immunother. 2001; 24: 263-271. 22. Jurianz K, Ziegler S, Garcia-Schuler H, Kraus S, Bohana-Kashtan O, Fishelson Z, Kirschfink M. Complement resistance of tumor cells: basal and induced mechanisms. Mol immunol. 1999; 36: 929-939. 23. Vetvicka V, Thornton BP, Wieman TJ, Ross GD. Targeting of natural killer cells to mammary carcinoma via naturally occurring tumor cell-bound iC3b and beta-glucan-primed CR3 (CD11 b / CD18). J Immunol. 1997; 159: 599-605. 24. Adachi Y, Okazaki M, Ohno N, Yadomae T. Enhancement of cytokine production by macrophages stimulated with (1- > 3) -beta-D-glucan, grifoian (GRN), isolated from Grifola leafy. Biol Pharm Bull. 1994; 17: 1554-1560. 25. Van der Kolk LE, Grillo-Lopez AJ, Baars JW, CE Hack, van Oers MH. Complement activation plays a key role in the side effects of rituximab treatment. Br J Haematol. 2001; 115: 807-811.
EXAMPLE III Extract of barley ß-glucan synthesized with IgM antibodies Natural IgM antibody from human serum when administered intravenously was cytotoxic to human neuroblastoma (NB) cells that had the effect of stopping the growth of subcutaneous solid human NB xenografts in hairless rats. (1, 2) IgM was absorbed by tumors with activation of perivascular complement and accumulation of granulocytes after 24 hours. (3) The metastatic NB model, the IgM antibody was effective in the elimination of tumors in 90% of the mice. (4) The absence of this anti-NB IgM antibody during infancy and between patients with NB (of any age), and its predominance after 12 months of age has led to the hypothesis that natural IgM antibodies could play a role. role as an immunological control mechanism against NB. (5) 3G6 is a mouse IgM monoclonal antibody (MoAb) anti-GD2. Within 48 hours after the intravenous injection of biotinylated 3G6, the subcutaneous NB xenografts showed tumor cell membrane staining. Although 3G6 had lower mean fluorescence (53 ± 19 fluorescent channel units, n = 7 mice) when compared to 3F3, a MoAb IgG (149 ± 44, n = 7), 3G6 plus beta-glucan was effective against human NB (p <0.05), with a dose response curve (Figure 4) comparable to that of 3F8. (6) These findings were consistent with those who used human natural anti-NB IgM. (1, 2) These data support the idea that beta-glucan can increase not only IgG-inducing vaccines, but also IgM-inducing vaccines.
References for example III 1. David K, Ollert MW, Juhl H, et al: Growth arrest of solid human neuroblastoma xenografts in nude rats by natural IgM from healthy humans. Nat Med 2: 636-9, 1996. 2. Ollert MW, David K, Schmitt C, et al: Normal human serum contains a natural IgM antibody cytotoxic for human neuroblastoma cells. Proc Nati Acad Sci U S A 93: 4496-503, 1996. 3. Ollert MW, David K, Vollmert C, et al: Mechanisms of in vivo anti-neuroblastoma activity of human natural IgM. Eur J Cancer 33: 1942-6, 1997. 4. Engler S, Thiel C, Forster K, et al: A novel metastatic animal model reflecting the clinical appearance of human neuroblastoma: growth arrest of orthotopic tumors by natural, cytotoxic human immunoglobulin M antibodies. Cancer Res 61: 2963-73, 2001. 5. Erttmann R, Schmitt C, Ollert MW, et al: Naturally occurring humoral cytotoxicity against neuroblastoma (NB) cells and healthy persons and NB patients. Pediatr Hematol Oncol 13: 545-d, 1996. 6. Cheung N, Modak S: Oral (1-3), (1-4) -beta-glucan syngergizes with anti-ganglioside GD2 monoclonal antibody 3F3 n the therapy of neuroblastoma . Clin Cancer Res d: 1217-1223, 2002.
EXAMPLE IV (1? 3), (1? 6) ß-glucan derived from yeast for baking (derived from Saccharomvces cerevisiae) is also effective in improving cancer therapy with antibodies LAN-1 tumor cells were seeded (2x106 cells) in 100 μl of Matrigel (Sigma) subcutaneously. The dimensions of the tumor were measured two to three times a week with vernier caliper, and the size of the tumor was calculated as the product of the two largest perpendicular diameters. All treatment studies started in groups of 4-5 mice when the tumor diameters reached 0.7 to 0.8 cm. Mice were treated with antibody (3F3 or 3G6) (200 ug per day) intravenously (by tail vein injection) twice a week x 5 dose and oral beta-glucan (400 ug per day) by intragastric injection every day for a total of 14-13 days (see figures 5 and 6). Glucans derived from yeast cell walls, such as Saccharomyces cervisiae or mutant yeast strains described in U.S. Pat. No. 5,250,436, the disclosure of which is incorporated herein by reference in its entirety, can be used in the above compositions. Glucans having ß (1-3) and β (1-6) linkages can be prepared by the procedure described in the U.S. Patents. Nos. 5,233,491 and 4,610,646, the descriptions of which are hereby incorporated by reference in their entirety. Soluble or aqueous glucans that are suitable for oral administration are can produce by the process described in the patents of E.U.A. Nos. 4,310,646 and 5,519,009, the descriptions of which are hereby incorporated by reference in their entirety. Beta-glucans such as beta-1, 3/1, 6 soluble glucan or SBG manufactured by Biotec Pharmacon (Norway) can also be used. In similar experiments, a model of subcutaneous lymphoma was studied. Here, 5x10 6 cells suspended in 0.1 ml of Matrigel (Becton-Dickinson, Franklin Lakes, NJ) were seeded on mouse flanks. The dimensions of the tumor were measured two to three times a week and the size of the tumor was recorded as the product of the two largest diameters. The mice were sacrificed when the maximum tumor dimension exceeded 20mm. 200 μg of rituximab (Genentech, San Francisco, CA) was injected intravenously twice a week for a total of eight injections and 400 μg of glucan was administered orally by intragastric forced feeding daily for 29 days. The mice were weighed weekly and clinically observed at least once a day. The tumor response rate and the percent of mice that achieved complete remissions were comparable between barley glucan and yeast glucan. These subcutaneous tumor model series showed that (1? 3), (1? 6) soluble yeast beta-glucan of large molecular weight (> 10,000 Daltons) is equally as potent as (1-3), (1? 4) barley beta-glucan. In addition, the source and physical form of yeast glucan can make substantial differences. The model of metastatic lymphoma was also studied. A The disseminated tumor model was established in SCID mice as described above. (1) In brief, 5x106 Daudi cells in 100 μl of normal saline were injected intravenously (i.v.) in SCID mice. The tumors grew systematically and the mice were paralyzed when the tumor cells infiltrated the spinal canal, resulting in paralysis of the hind legs. The mice were sacrificed at the onset of paralysis or when the animals lost 10% of their body weight. The therapy was started 10 days after the injection of tumor cells. 40 μg of rituximab (Genentech, San Francisco, CA) was injected intravenously twice a week for a tota! of eight injections and 400 μg of glucan administered orally by intratragastric forced feeding daily for 29 days. The mice were weighed weekly and clinically observed at least once a day (see figure 7). Again both barley glucan and yeast glucan showed comparable effect when combined with Rituxan. Neither barley glucan nor yeast glucan had any effect on survival when used alone (data not shown).
References for example IV 1. Wei BR, Ghetie MA, Vitetta ES: The combined use of an immunotoxin and radioimmunoconjugate to treat disseminated human B-cell lymphoma in immunodeficient mice. Clin Cancer Res 6: 631-642, 2000.
EXAMPLE V Mechanism by which orally administered ß-glucans work with anti-tumor monoclonal antibodies to mediate tumor regression (1) Using syngeneic tumor (GD2 + RMA-S) in wild type C57B.I / 6 (WT) mice versus either C57B1 / 6 mice deficient in CR3 (CD11b - / -) or deficient in C3 (C3 - / -), MoAb only did not induce tumor regression, whereas the combination of anti-GD2 MoAb intravenously with barley beta-glucan or oral yeast induced significant regression in WT but it is not mice deficient in CR3. In addition, the combined treatment with intravenous MoAb and oral beta-glucans produced 60-100% of tumor-free survivors in WT mice, but only 0-20% survival in mice deficient in CR3. These experiments demonstrated an almost absolute requirement for CR3 of leukocytes for the antitumor effect, especially when oral barley beta-glucan was given with MoAb antitumor. A therapy protocol comparing WT with C3 deficient mice showed similarly that oral beta-glucan therapy required serum C3. When barley beta-glucan and yeast beta-glucan were labeled with fluorescein (BG-F and YG-F) and given to mice by intragastric injection, trafficking of beta-glucan was followed. Within three days of daily oral administration of BG-F or YG-F, the macrophages in the baso and lymph nodes contained fluorescein-labeled beta-glucan.
After 4 days, YG-F and BG-F were also observed in bone marrow macrophages. When the uptake of YG-F and BG-F by WT versus mice deficient in CR3 were compared, no differences were evident in either the percentage of macrophages containing ingested beta-glucan-F or the amount of beta-glucan-F by cell. Therefore, the uptake of beta-glucan from barley and yeast by gastrointestinal macrophages does not require CR3 and is probably mediated rather by Dectin-1. (2) In vitro macrophages and bone marrow were able to degrade large molecules of beta-glucan from barley or yeast into smaller biologically active fragments of beta-glucan that are then released. To determine whether the soluble beta-glucan-F released by macrophages had indeed been absorbed by bone marrow granulocytes, the WT or CR3-deficient mice given YG-F or BG-F for 10 days were He injected them intraperitoneally with the thioglycollate medium to induce the marginalized pool of granulocytes from bone marrow in the peritoneal cavity. Only WT granulocytes were able to collect the YG-F and BG-F released from macrophages. These data suggest a sequential ingestion of beta-glucan by gastrointestinal macrophages that release beta-glucan to the bone marrow where the soluble degradation fragments are released and absorbed by granulocytes by means of membrane CR3. When the peripheral granulocytes were isolated from WT and mice deficient in CR3 given oral beta-glucan, only WT granulocytes were able to kill tumor cells coated with iC3b in vitro. These experiments show that bone marrow granulocytes and tissue macrophages acquire soluble beta-glucan bound to CR3 membrane of gastrointestinal macrophages, and that this bound beta-glucan initiates CR3 or both granulocytes and macrophages so that when recruited to a site of inflammation can kill tumor cells coated with ¡C3b.
References for Example V 1. Hong F, Yan J, Baran JT, et al: Mechanism by which orally administered beta (1, 3) -glucans function with anti-tumor monoclonal antibodies to mediate tumor regression and tumor-free survival. J Exp Med, 2004. 2. Herre J, Gordon S, Brown GD: Dectin-1 and its role in the recognition of beta-glucans by macrophages. Mol Immunol 40: 369-76, 2004.
EXAMPLE VI β-Soluble Glucan can be used as a conduit for plasmids The main obstacles to the supply of DNA, RNA and proteins orally are the acid and proteolytic environment of the stomach, and the limited absorption of proteins by GALT. It is believed that M cells within Peyer patches and phagocytes are the predominant vehicles for microparticle absorption. However, nanoparticles can also access GALT through a paracellular mechanism1, 2 and by transcytosis3. In any case, the observed absorption of particles can be improved by using particles with mucoadhesive properties or affinity for receptors on the cells. Many polymers that have been used to make nanoparticles are mucoadhesives. Among them are alginate, carrageenans and pectin. Although these materials are often used as the core polymers in nanoparticles, no specific receptor has been identified for these polymers and the absorption efficiency remains suboptimal. Dectin-1 is now known to be a universal receptor for beta-glucan, and is found in many human tissues including monocytes and phagocytes. The gelling properties of high molecular weight beta-glucan allows the RNA, DNA and proteins to be embedded. Since sugars are highly resistant to acidic conditions and enzymes, proteins, RNA and DNA remain protected as they pass through! gastrointestinal tract. Through the high affinity Dectin-1 receptor for beta-glucan, these substances can be introduced into phagocytes as potential vehicles to the rest of! body. The vector pEGP-C1 (see figure d) was purchased from BD Biosciences (Palo Alto, CA) and prepared in accordance with the manufacturers' instructions. pEGFP-C1 encodes the red-shifted variant of wild-type GFP (1-3) that has been optimized for brighter fluorescence and higher expression in mammalian cells. (Maximum excitation = 48d nm, maximum emission = 507 nm.) The base structure of the vector also contains an SV40 origin for replication in mammalian cells only if they express the SV40 T antigen. A bacterial promoter towards the 5 'end of this cassette expresses resistance to kanamycin in E. coli. The base structure of pEGFP-C1 also provides a replication pUC origin for propagation in E. coli and an origin of f1 for production of single-stranded DNA. Mice were fed with 50 μg of plasmid pEGFP-d mixed in 400 μg of beta-glucan (-200,000 Daltons) in 100 μl of saline for forced feeding now! whereas the control mice were given plasmid alone. The oral feeding was done during 3 consecutive days (days 1, 2 and 3). 50 μl of blood obtained from the tail vein were analyzed by FCAS analysis after lysis of red blood cells and the percent of cells expressing GFP in the monocyte population was recorded. The average ratio of percent green cells to glucan versus groups without glucan (n = 4-9 mice per group) is presented in Figure 9. Throughout the 14 days of the experiment, the percent of green monocytes in the Group without glucan remained stable at background levels. On the other hand, after day 1 of oral forced feeding, there was a greater percentage consisting of circulating green monocytes, which reached a peak around day d. Since GFP is not normally found in mouse monocytes, the presence of green cells is consistent with expression of GFP protein after the entry of the plasmid into the monocytes circulating in the blood. The experiment was repeated using barley ß-glucan higher molecular weight (-350,000 Daltons) with better gelling properties. In figure 10, a similar kinetics was seen, with a higher percentage of green cells that persisted from day d to day 11 (n = 4 mice per group). The presence of GFP mRNA was tested using analysis of Quantitative reverse transcription PCR. Mice were fed with 50 μg of plasmid pEGFP-d mixed in 400 μg of high molecular weight (-350,000 Daltons) of beta-glucan in 100 μl of saline by oral forced feeding while the control mice were given plasmid alone. 50 μl of peripheral blood was used to extract total RNA, reverse transcribed and quantitative real-time PCR was performed using a modification of the method described above4. The mouse GAPDH maintenance gene is used as an internal control. The level of transcription is calculated using known GFP and standard GAPDH. The transcription units were calculated separately for GFP and GAPDH and the results are obtained as a ratio of GFP over GAPDH. In Figure 11, the level of average RNA (GFP / GAPDH) is expressed as a ratio of glucan to no glucan groups (n = 4 mice per group). GFP mRNA was detected until day 10.
References for example VI 1. Damge C, Aprahamian M, Marcháis H, et al: Intestinal absorption of PLAGA microspheres in the rat. J Anat 1 d9 (Pt 3): 491-501, nineteen ninety six. 2. Jani P, Halbert GW, Langridge J, et al: Nanoparticle uptake by the gastrointestinal mucosal rat: quantitation and particle size dependency. J Pharm Pharmacol 42: 321-6, 1990. 3. Florence AT: The oral absorption of micro- and nanoparticulates: neither exceptional noror unusual. Pharm Res 14: 259-66, 1997. 4. Cheung IY, Piccolo MS, Collins N, et al: Quantitation of GD2 synthase mRNA by real-time reverse transcription-polymerase chain reaction: utility in bone marrow purging of neuroblastoma by anti -GD2 antibody 3F3. Cancer 94: 3042-3, 2002.

Claims (13)

NOVELTY OF THE INVENTION CLAIMS
1. - A composition for oral intake of substance comprising an appropriate amount of carbohydrates.
2. A composition for the oral delivery of one or more substances comprising an effective amount of an orally administered beta-glucan and one or more chemotherapeutic agents.
3. The composition according to any of claims 1 or 2, further characterized in that the carbohydrate is glucan.
4. The composition according to claim 3, further characterized in that the glucan contains mixed 1, 3-1, 6 or 1, 3-1, 4 bonds or a mixture of both bonds 1, 3-1, 6 and 1 , 3-1, 4 mixed.
5. The composition according to claim 3, further characterized in that the glucan improves the efficacy of chemotherapeutic agents or anti-cancer antibodies.
6. The composition according to claim 3, further characterized in that the glucan is derived from grass, plants, mushrooms, yeast, barley, fungi, wheat or algae.
7. The composition according to claim 3, further characterized in that e! Glucan is of high molecular weight.
8. - The composition according to claim 3, further characterized in that the substance is a peptide, protein, RNA, DNA or plasmid.
9. The composition according to claim 3, further characterized in that the substance is a chemotherapeutic agent.
10. A composition comprising an effective amount of (1-3), (1? 6) or (1? 3), (1? 4) orally administered beta-glucan capable of improving the efficacy of IgM antibodies.
11. The composition according to claim 10, further characterized in that the antibody is an antibody against cancer.
12. The composition according to claim 11, further characterized in that the antibody is an antibody that binds to tumor.
13. The composition according to claim 12, further characterized in that the antibody is capable of activating the complement.
MXPA/A/2006/000615A 2003-07-16 2006-01-16 Therapy-enhancing glucan MXPA06000615A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10621027 2003-07-16

Publications (1)

Publication Number Publication Date
MXPA06000615A true MXPA06000615A (en) 2006-10-17

Family

ID=

Similar Documents

Publication Publication Date Title
CA2536632C (en) Therapy-enhancing glucan
US7906492B2 (en) Therapy-enhancing glucan
EP1536820B2 (en) Cancer therapy using whole glucan particles and antibodies
Liu et al. Combined yeast-derived β-glucan with anti-tumor monoclonal antibody for cancer immunotherapy
US20100166751A1 (en) Cancer therapy using whole glucan particles and antibodies
US8323644B2 (en) Therapy-enhancing glucan
JP2009528267A (en) Glucan to enhance treatment
US20090053221A1 (en) Immune response enhancing glucan
Liu et al. Yeast-derived β-glucan in combination with anti-tumor monoclonal antibody therapy in cancer
MXPA06000615A (en) Therapy-enhancing glucan
CN101426510A (en) Therapy-enhancing glucan
CA2637205A1 (en) Therapy-enhancing glucan