USRE39788E1 - Gene therapy - Google Patents

Gene therapy Download PDF

Info

Publication number
USRE39788E1
USRE39788E1 US10/701,022 US70102203A USRE39788E US RE39788 E1 USRE39788 E1 US RE39788E1 US 70102203 A US70102203 A US 70102203A US RE39788 E USRE39788 E US RE39788E
Authority
US
United States
Prior art keywords
cells
til
human
gene
lymphocytes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US10/701,022
Inventor
W. French Anderson
R. Michael Blaese
Steven A. Rosenberg
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27502997&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=USRE39788(E1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by US Department of Health and Human Services filed Critical US Department of Health and Human Services
Priority to US10/701,022 priority Critical patent/USRE39788E1/en
Application granted granted Critical
Publication of USRE39788E1 publication Critical patent/USRE39788E1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464436Cytokines
    • A61K39/464438Tumor necrosis factors [TNF], CD70
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • This invention relates to the use of primary human cells as vehicles for human gene transfer. More particularly, this invention relates to the use of human cells (such as, for example, but not limited to, human blood cells) as vehicles for the transfer of human genes encoding therapeutic agents and/or genes encoding detectable markers.
  • human cells such as, for example, but not limited to, human blood cells
  • Retroviral-mediated gene transfer is a new therapeutic approach for the treatment of human disease (W. F. Anderson, Science 226:401 (1984)).
  • Initial attention has centered on candidate diseases affecting the bone marrow such as the hemoglobinopathies and severe combined immunodeficiency.
  • Early attempts at bone marrow gone transfer in large mammals and primates were only partially successful.
  • specific blood cells for example lymphocytes, have been used. Lymphocytes have several features which make them potentially attractive cellular vehicles for gene therapy (K. Culver, et al., J. Cellular Biochemistry Suppl. 12B:171 (1988); R. M. Blaese, et al, Clin. Research 37:599A (1989)).
  • Lymphocytes are readily available from peripheral blood as a single cell suspensions and they are easily manipulated in tissue culture where the availability of recombinant growth-factors such as rIL-2 permits their expansion by thousands of fold. This adaptability to tissue culture allows serial attempts at gene insertion, selection procedures and time to test for gene expression and other properties of the gene-transduced cells prior to their return to the patient. Long-lived antigen-specific memory lymphocytes proliferate when exposed to their appropriate antigen and thus the population of gene-treated lymphocytes can be selectively and specifically expanded in vivo by immunization of the host. Finally, some populations of antigen-specific lymphocytes “target” to sites in the body containing deposits of antigen.
  • gene-treated antigen-specific lymphocytes can be used to deliver specific gene products directly to the site of pathology, such as a tumor, in a treated patient.
  • pathology such as a tumor
  • gene-treated antigen-specific lymphocytes can be used to deliver specific gene products directly to the site of pathology, such as a tumor.
  • TIL transduced with genes promoting secretion of such a cytokine and using the TILs wn unique antigen-specific recept rs to target them to deposits f tumor will permit greater antitumor effect with less systemic toxicity.
  • RNA DNA
  • therapeutic agent RNA which encodes a therapeutic agent of interest
  • RNA DNA
  • therapeutic agent includes treatment and/or prophylaxis
  • a primary cell is one which will not have indefinite growth in culture, i.e., the cells has not been manipulated or transformed to provide for indefinite growth in culture.
  • the DNA (RNA) which is used for transducing the human cells may be one whose expression product is secreted from the cells.
  • the DNA (RNA) cures a genetic deficiency of the cells and the expression product is not secreted from the cells (for example, the expression product of DNA encoding ADA is not secreted from the cells).
  • the human cells may also be genetically engineering with DNA (RNA) which functions as a marker, as hereinafter described in more detail.
  • RNA DNA
  • primary human cells which are genetically engineered to include DNA which encodes a marker or therapeutic, with such cells expressing the encoded product in vivo.
  • the invention is directed to a method of enhancing the therapeutic effects of human primary cells.
  • a method of enhancing the therapeutic effects of human primary cells which specifically “target” to a tissue site in a patient, whereby DNA (RNA) which encodes an agent that enhances the therapeutic effects is inserted in the cells.
  • the tissue site can be, for example, a tumor.
  • the DNA (RNA) produces the agent in the patient's body, and in accordance with such embodiment the agent is expressed at the tissue site itself.
  • primary human cells which are genetically engineered need not be targeted to a specific site and in accordance with the invention, such engineered primary human cells function as a systemic therapeutic; e.g., a desired therapeutic agent can be expressed and secreted from the cells systemically.
  • the primary human cells may be primary human nucleated blood cells or primary human tumor cells.
  • the primarily human cells are primary human nucleated blood cells.
  • the primary human blood cell which can be used in the present invention include leukocytes, granulocytes, monocytes, macrophages, lymphocytes, immature forms of each of the previous cells (as well as immature erythroblasts) (CD 34 +cells) and totipotent stem cells.
  • human cells preferably blood cells, which are genetically engineering are cells which are targeted to a specific site; for example, the cells can be tumor infiltrating lymphocytes (TIL cells).
  • TIL cells tumor infiltrating lymphocytes
  • the engineered cells function as a therapeutic at such site.
  • the cells are n t cells which are targeted to a specific site and in such aspect such cells function as a systemic therapeutic.
  • the DNA carried by the cells can be any DNA having clinical usefulness, for example, any DNA that directly or indirectly enhances the therapeutic effects of the cells.
  • the DNA carried by the cells can be any DNA that allows the cells t exert a therapeutic effect that the cells would not exert.
  • suitable DNA which can be used for genetically engineering, f r example, bl od cells, include those that encode cytokines such as tumor necrosis factor (TNF), interleukins (for example, interleukins 1-12), interferons ( ⁇ , ⁇ , ⁇ -interferons). T-cell receptor proteins and the Fc receptors for antigen-binding domains of antibodies, such as immunoglobulins.
  • the inserted gases are marker genes which permit determination of the traffic and survival of the transformed cells in vivo.
  • marker genes include the neomycin resistance (neoR) gene, multi-drug resistant gene, thymidine kinase genes, ⁇ -galactosidase, dehydrofolate reductase (DHFR) and chloroamphenicol acetyl transferase.
  • RNA DNA
  • the cells may be genetically engineered in vitro or in vivo.
  • cells may be removed from a patient; genetically engineered in vitro with DNA (RNA) encoding the therapeutic agent, with such genetically engineered cells being readministered to the patient.
  • RNA DNA
  • Such a treating procedure is sometimes referred to as an ex vivo treatment.
  • DNA (RNA) encoding the therapeutic agent may be administered to the patient for delivery of the DNA in vivo to the targeted cells.
  • RNA e.g., a retroviral or other viral vector; liposomes etc.
  • a patient is provided with human primary cells which are genetically engineered with DNA (RNA) encoding a therapeutic agent whereby such therapeutic agent may be expressed in vivo.
  • RNA DNA
  • such genetically engineered cells may be provided by administering to the patient cells which have been genetically engineered ex vivo or by administering the DNA (RNA) as part of a delivery system for genetically engineering targeted cells in vivo.
  • RNA DNA
  • a composition comprising (i) primary human cells genetically engineered with DNA (RNA) encoding a therapeutic agent and (ii) a pharmaceutically acceptable carrier suitable for administration to a patient.
  • the primary human cells are primary human nucleated blood cells.
  • the carrier may be a liquid carrier (for example, a saline solution) or a solid carrier, e.g., an implant.
  • the engineered cells may be introduced, e.g., intravenously, sub-cutaneously, intramuscularly, intraperitoneally, intralesionaly, etc.
  • FIG. 1A dem nstrates by assay for function nal enzyme that murine 14.1-T cells containing an inserted SAX vector express high levels of the neomycin resistant (neoR) gene product, aeomycin phosphotranferase (NPT).
  • FIG. 1B demonstrates the expression f human adenosine deaminase (hADA), also encoded by the SAX vector inserted in the murine 14.1-T cells.
  • hADA human adenosine deaminase
  • FIG. 2 shows the proliferative response of murine splenic lymphocytes and 14.1-T cells to stimulation with phytohemagglutinin and IL-2and the effect of G418 on this proliferati n.
  • FIG. 3A demonstrates that human N2-transduced TIL expresses sufficient NPT to become resistant to the toxic effects of G418 on cellular proliferation.
  • FIG. 3B shows growth curves of human N2-transduced and untreated TIL for the 30 day period following gene insertions;
  • FIG. 4 demonstrates absolute Blood T Cell levels of patient # 1 receiving ADA gene therapy in accordance with the procedure of Example 4.
  • the bar portion of the figure is the transduced T-cells infused at the day indicated;
  • FIG. 5 demonstrates the ADA enzyme activity of patient # 1 during treatment.
  • the present invention relates to the use of primary human cells as cellular vehicles for gene transfer.
  • the genes can be any gene having clinical usefulness, for example, therapeutic or marker genes.
  • the primary human cells are blood cells.
  • blood cells as used herein is meant to include all forms of nucleated blood cells as well as progenitors and precursors thereof, as hereinabove described.
  • the invention is directed to a method of enhancing the therapeutic effects of human primary cells, preferably blood cells, that are infused in a patient, comprising: (i) inserting into the human primary cells of a patient a DNA (RNA) segment encoding a product that enhances the therapeutic effects of the human primary cells; and (ii) introducing cells resulting from step (1) into the patient.
  • the gene can be inserted into the human primary cells using any gene transfer procedures, for example, retroviral-mediated gene transfer, electroporation, calcium phosphate mediated transfection, microinjection or proteoliposomes.
  • Other vectors can be used besides retroviral vectors, including those derived from DNA viruses and other RNA viruses.
  • an RNA virus such as virus includes RNA which encodes the desired agent, with the human primary cells which are genetically engineered with such RNA virus thus being provided with DNA encoding the agent.
  • a method of enhancing the therapeutic effects of blood cells, that are infused in a patient comprising: (i) inserting into the blood cells of a patient a DNA (RNA) segment encoding a product that enhances the therapeutic effects of the blood cells; and (ii) introducing cells resulting from step (i) into the patient under conditions such that the cells resulting from step (i) “target” to a tissue site.
  • the cells are not “targeted” and function as a systemic therapeutic.
  • the genes are inserted in such a manner that the patient's transformed blood cell will produce the agent in the patient's body.
  • the specificity of the blood cells f r the antigen is n t lost when the cell produces the product.
  • DNA (RNA) may be inserted into the blood cells of a patient, in vivo, by administering such DNA (RNA) in a vehicle which targets such blood cells.
  • the method of the invention can-be used, for example, in the treatment of cancer in a human by inserting into human primary cells, such as, f r example, blood cells, which specifically “target” to a tumor and which have been removed from a cancer patient and expanded in culture, genes that enhance the anti-tumor effects of the blood cells.
  • human primary cells such as, f r example, blood cells, which specifically “target” to a tumor and which have been removed from a cancer patient and expanded in culture, genes that enhance the anti-tumor effects of the blood cells.
  • the blood cells can be expanded in number before or after insertion of the genes.
  • the method of gene transfer in the blood cells is as described above.
  • the procedure is performed in such a manner that upon injection into the patient, the transformed blood cells will produce the agent in the patient's body, preferably at the site of the tumor itself.
  • the gene carried by the blood cells can be any gene which directly or indirectly enhances the therapeutic effects, of the blood cells.
  • the gene carried by the blood cells can be any gene which allows the blood cells to exert a therapeutic effect that it would not ordinarily have, such as a gene encoding a clotting factor useful in the treatment of hemophilia.
  • the gene can encode one or more products having therapeutic effects. Examples of suitable genes include those that encode cytokines such as TNF, interleukins (interleukins 1-12), interferons ( ⁇ , ⁇ , ⁇ -interferons), T-cell receptor proteins and Fc receptors for antigen-binding domains of antibodies, such as immunoglobulins.
  • suitable genes include genes that modify blood cells to “target” to a site in the body to which the blood cells would not ordinarily “target,” thereby making possible the use of the blood cell's therapeutic properties at that site.
  • blood cells such as TIL can be modified, for example, by introducing a Fab portion of a monoclonal antibody into the cells, thereby enabling the cells to recognize a chosen antigen.
  • blood cells having therapeutic properties can be used to target, for example, a tumor, that the blood cells would not normally target to.
  • Other genes useful in cancer therapy can be used to encode chemotactic factors which cause an inflammatory response at a specific site, thereby having a therapeutic effect.
  • genes encoding soluble CD4 which is used in the treatment of AIDS and genes encoding ⁇ -antitrypsin, which is useful in the treatment of emphysema caused by ⁇ -antitrypsin deficiency.
  • the gene therapy of the present invention is useful in the treatment of a variety of diseases including but not limited to adenosine deaminase deficiency, sickle cell anemia, thalassemia, hemophilia, diabetes, ⁇ -antitrypsin deficiency, brain disorders such as Alzheimer's disease, and other illnesses such as growth disorders and heart diseases, for example, those caused by alterations in the way cholesterol is metabolized and defects of the immune system.
  • diseases including but not limited to adenosine deaminase deficiency, sickle cell anemia, thalassemia, hemophilia, diabetes, ⁇ -antitrypsin deficiency, brain disorders such as Alzheimer's disease, and other illnesses such as growth disorders and heart diseases, for example, those caused by alterations in the way cholesterol is metabolized and defects of the immune system.
  • a method of detecting the presence of human primary cells comprising (i) inserting into human primary cells removed from the patient a DNA (RNA) segment encoding the marker under conditions such that the marker is present in the blood cells; (ii) introducing cells resulting from step (i) into the patient; (iii) removing from the patient an aliquote f tissue (which can be, for example, normal tissue, cancerous tissue, blood, lymph nodes, etc.) including cells resulting from step (ii) and their progeny; and (iv) determining the quantity of the cells resulting from step (ii) and their progeny, in said aliquot.
  • human primary cells such as, for example, blood cells containing a marker present in a patient
  • the present invention comprises a method f detecting the presence f blood cells containing a marker, present at a site in a patient, to which site the blood cells “targets”, c mprising (i) inserting into blood cells removed from the patient that are specific for an antigen (either known or unknown) present at the site and that target to site, a DNA segment encoding the marker under conditions such that the marker is present in the blood cells, (ii) introducing cells resulting from step (i) into the patient under conditions such that cells resulting from step (ii) can be found at the site; (iii) removing from the patient an aliquot of tissue from the site, which aliquote includes cells resulting from step (ii) and their progeny; and (iv) determining the quantity of the cells resulting from step (ii) and their progeny, in said aliquot.
  • the marker gene can be, for example, inserted into blood cells which specifically “target” to a tumor in order to determine the traffic and survival of the transformed blood cells in vivo.
  • the transformed blood cells circulate through the body, rather than targeting to a particular tissue site.
  • the marker gene can be any gene which is different from the genes in the blood cell into which the marker gene is inserted.
  • marker genes include neoR, multi-drug resistant gene, thymidine kinase gene, ⁇ -galactosidase, dehydrofolate reductase (DHFR) and chloroamphenicol acetyl transferase.
  • the marker gene can be inserted into a human primary cell, such as a blood cell, together with a therapeutic gene or separately.
  • the marker gene and the therapeutic gene may also be one and the same.
  • the retroviral gene transfer vector SAX (P. W. Kantoff, et al, PNAS 83:6563 (1986)) was used to insert the genes for human adenosine deaminase (hADA) and neomycin resistance (neoR) into the murine T cell line 14.1
  • SAX is a moloney virus based vector with the neoR gene promoted from the retroviral LTR and the hADA gene promoted from an internal SV40 promoter.
  • the 14.1 T cell line was derived from the draining lymph nodes of a BIO.D2 mouse previously immunized with sperm whale myoglobin (SWM) by repeated cycles of antigen stimulation of these lymphocytes in tissue culture (I. Berkower, et al., J. Immunol. 135:2628 (1985)). These 14.1 T-cells proliferate when challenged with SWM in the presence of histocompatible antigen-presenting cells (without exogenous IL-2) or when stimulated with PHA and rIL-2 in vitro. An average of 20% of the 14.1-T cells achieved stable insertion of the SAX vector after a supernatant infection protocol (P. W. Kantoff, et al, PNAS 83:6563 (1986)).
  • FIG. 1A demonstrates by assay for functional enzyme that the cells containing the inserted SAX vector are expressing high levels of the neoR gene product, neomycin phosph transferase (NPT).
  • FIG. 1B demonstrates expression of human adenosine deaminase (hADA), also encoded by the SAX vector inserted in these T cells.
  • hADA human adenosine deaminase
  • a thymic nude mice were then injected intraperitoneally with 20 ⁇ 10 6 14.1SAX T-cells and immunized with 100 ⁇ g soluble SWM. Athymic recipients were chosen for these experiments because they have very little endogenous Te cell activity and therefore any T cells recovered from these mice at a later date should represent the original injected 14.1 SAX cells or their progeny.
  • FIG. 2 shows the proliferative response of splenic lymphocytes and 14.1 cells to stimulation with phytohemagglutinin and IL-2 and the effect of G418 on this proliferation. While control nude mouse splenocytes did proliferate modesty to PHA+IL-2, this response was abolished by low concentrations of G418.
  • TIL tumor infiltrating lymphocytes
  • TIL therapy many issues remain to be clarified including a more complete understanding of the features of individual TIL preparations which correlate with clinical antitum r effects, the in vivo distribution and time of persistence f TTL after infusion, and whether distinct TIL functional phenotypes localize in the body in ways which will permit prediction of their clinical efficacy.
  • a gene label will also equally mark progeny cells derived by continued proliferation of the originally labelled cell population as occurs when TIL proliferation is driven by IL-2 infusions given to the patient.
  • the target cell acquires resistance to the neomycin analog G418 which permits, as we have shown for mouse 14.1SAXT-cells, the selective recovery of the gene-marked cells by regrowth of recovered cells in the presence of G418. With recovery, the cells from different sites could then be analyzed for their functional phenotypes and potential correlations with clinical efficacy.
  • N2 contains the gene neoR promoted by the retroviral LTR and has been used extensively by the inventors in studies of bone marrow gene-transfer in mide and primates during the past 5 years (M. E. Egitis, et al, Science 230:1396 (1985); D. Kohn, et al, Blood Cells 13:285-298 (1987)); P. W. Kantoff, et al, J. Exp. Med.
  • 3B shows growth curves of N2-transduced and untreated TIL for the 30 day period following gene insertion. Growth of the N2-transduced TIL population as a whole slows initially when G418 is added to the culture (not shown, but resumes exponential expansion as growth of the NeoR-expressing G418-resistant subpopulation becomes established. After completion of selection, the N2-transduced cells as a rule grew at a rate comparable to that of TIL not transduced with the neoR gene.
  • TIL transduced with N2 and selected with 0.3 mg/ml G418 for 10-14 days were compared with non-transduced TIL from 6 patients with methastatic malignant melanoma. These populations were analyzed for their surface membrane phenotype as revealed by FACS analysis of 1 ⁇ 10 6 TIL stained monoclonal antibodies. Antibodies to CD4(Leu 3) and CD8 (Leu 2) were used on populations and the percentage of the cells positive for each determinant are shown.
  • TIL constitutive cytotoxic function of each TIL population was determined for autologous tumor targets (auto) and the NK sensitive target K562 (NK) as well as for allogeneic tumor and sensitive target cells (not shown) by a standard 4 hour 56 Cr release assay at E:T ratios of 40:1, 15:1, and 4.5:1. Data shows target lysis at the 40:1 E:T ratio.
  • TIL isolated from different individual donors have different phenotypic profiles as determined by various cell membrane determinants (CD4, CD8, etc.) and different levels of cytotoxic activity toward autologous tumor and other target cell types.
  • Human TIL of both major phenotypic subgroups (CD4 and CD8) were readily transduced by the N2 vector and expressed G418 resistance.
  • TIL transduced and selected TIL populations
  • Some transduced and selected TIL populations showed more drift in their phenotypic composition than others, and as expected with more prolonged culture, all of the TIL populations became progressively oligoclonal whether or not they had been transduced with N2 (not shown).
  • TIL are often cytolytic for autologous tumor cells but not other target cell types in vitro.
  • TIL isolated from two were cytolytic to autologous tumor cells, but not alleogeneic melanoma tumor cells, NK (K562) or LAK (Daudi) sensitive targets.
  • the functional cytotoxic activity profile of the TIL was unaffected by N2 transduction and G418 selection, even in those TIL populations showing some drift in T cell subset composition measured by surface membrane antigen phenotype.
  • murine 14.1-T cells containing an inserted SAX vector express high levels of the neoR gene product, NPT, as well as expressing hADA.
  • the SAX-transdermal 14.1 cells were placed under selection with 0.3 mg/ml G418 for 14 days.
  • the SAX-transduced G418 selected cells were then expanded by repeated 14 day cycles of antigen stimulation and rest on syngeneic feeder cells. Lysates from the G418-selected 14.1 population were prepared and assayed for the presence of NPT by measuring the phosphorylation of kanamycin with P32 labelled ATP after separation of NPT from endogenous phosphotransferases on a non-denaturing polyacrylamide gel (B. Reiss, et al, Gene 30:211 (1984)).
  • the 14.1 cells containing the inserted SAX vector expresses high levels of NPT.
  • Lysates of the 14.1 cells were separated by electrophoresis on cellulose acetate (B. Lira, et al, Molecular Cellular Biol. 7:3459 (1987)) which was then stained for the presence of the murine and human isozymes of ADA (FIG. 1 B).
  • the human T cell line CEM was included as a control for the migration of hADA.
  • FIG. 1B demonstrates the expression of hADA.
  • the following experiment was performed in order to demonstrate the continued expression of a transferred gene in gene modified T-cells. Specifically, the effect of G418 on the proliferative response of murine splenic lymphocytes and 14.1-T cells to stimulation with phytohemagglutinin and IL-2 was studied.
  • FIG. 2 shows, as the mean of triplicate cultures, the proliferative response of the murine splenic lymphocytes and 14.1-T cells to stimulation with phytohemaglutimin and IL-2 and the effect on G418 on this proliferation.
  • a review of the figure shows that while control nude in use splenic lymphocytes proliferated modestly to PHA+IL-2, this response was abolished by low concentrati ns of G418.
  • the untreated TIL (no vector) was compared with TIL which had been transduced with N2 and selected with G418 (N2+G418) and with an N2-transduced TIL population which had not been further selected in G418 (N2). Proliferative was measured by 3H-thymidine incorporation as described in Example 2.
  • FIG. 3B shows the growth curves of the N2-transduced and the untreated TIL for the 30 day period following gene insertion. These results were obtained using the following procedure.
  • TIL from patent 1 were grown from a tumor biopsy as described (Tobalian et al, J. Immune Method 138:4006-4011 (1987)) and then an aliquot was transduced with the N2 vector by two 4-hour exposures to viral supernatant (note: LNL 6 is identical to N2 except that LNL6 has several additional safety features). Forty-eight hours later the transduced population was placed under selection for 10 days with 0.3 mg/ml G418. Non-transduced TIL exposed to this selection protocol were all dead by day 10. Growth of the N2-transduced TIL population allowed when placed in G418, but resumed at an exponential rate while still under selection as the cells expressing the neoR gene became dominant by day 10. TIL cultures were fed and split as required (1-2 times per week) and the cumulative cell total calculated when the running total reached >10 6 cells to that larger numbers of cells would not have to be maintained in culture.
  • the N2 - treated TIL population expressed the inserted gene sufficiently to permit growth in 0.3 mg/ml G418.
  • the N2-transduced cells at a rule grew at a rate comparable to that of TIL not transduced with the neoR gene.
  • the TNF-NeoR vector is constructed by modifying the Monkey murine leukemia vector.
  • the TNF gene containing water, LT125N was constructed from the vector LXSN (Miller AD and Rossman G., 1989, Biotechniques 7:980-990.), usign the entire 233 amino acid sequence encoding cDNA f the natural TNF genes (Pennies, et al, 1984, Nature 312, 724-729, Wang, et al, Science 228, 149-154).
  • the ribosome binding site used upstread of the TNF gene was a syntehtic one, a consensus sequence f r translation initiation based on Kozak's rules (Kozak, Nucle. Acids Res. 12, 857-872 (1984) and having the nucleotide sequence 5′ TTCCGCAGCAGCCCGCCACC3′.
  • the vectors construct was packaged using the PA317 packaging cell line (Miller et al., Mol. Cell Biol. 6, 2895-2902 (1986)).
  • Retroviral vector supernatant is produced by harvesting the cell culture medium from the PA317 packaging line developed by Dr. A. Dusty Miller. This line has been extensively characterized and was used by us in our previous studies of the infusion of TIL modified by the N2 vector.
  • the TNF-Neo vector preparations from PA317 are extensively tested to assure that no detectable replication competent virus is present. Tests for replication-competent virus are conducted on both the vector supernatant and on the TIL after transduction. The follwoing tests are run on the producer line and/or the viral supernatant:
  • the viral titer is determined on 3T3 cells. Viral preparations with titers greater than 5 ⁇ 10 4 colony forming units/ml are used.
  • TNF production by the product line is measured and should be significantly above baseline control values.
  • TNF is assayed using standard biologic assays on the L929 amino cell line (Asher et al. J. Immunol 138:963-974, 1987) or by ELISA assay (R&D Systems, Minneapolis, Minn.).
  • Sterility of the producer line and the supernatant is assured by testing for aerobic and anaerobic bacteria, fungus and for mycoplasma.
  • Viral testing is performed including:
  • Electron microscopy is performed to assure the absence of adventitions agents.
  • Sterility is assured by testing for aerobic anaerobic bacteria, fungus and mycoplasma.
  • Reverse transcriptase assay must be negative.
  • TNF protein assay to assure the production of TNF.
  • Cells must be producing at least 100 pg TNF/10 6 cells/24 hours.
  • IL-2 will be withdrawn from the culture medium of an aliqu t f r at least one week to assure that cells do n t exhibit auton mous growth in the absence f IL-2.
  • peripheral blood lymphocytes are collected by leukapheresis for four hours. These are Ficoll-Hypaque separated and the mononuclear cells from the interface, washed in saline, and placed in culture in roller bottles at 10 6 cells/ml. Half are placed into AIM-V (a serum free medium, Gibco Laboratories) with 6000 IU/ml IL-2 (Cetus), and Half are placed into RPMI supplemented with 2% type-compatible human serum, penicillin (unless the patient is allergic), gentamicin, and 600 IU/ml IL-2. After 3 to 4days cells are centrifuged and the supernatants are collected and filtered. These are referred to as LAK supernatants.
  • AIM-V a serum free medium, Gibco Laboratories
  • 6000 IU/ml IL-2 Cetus
  • RPMI RPMI supplemented with 2% type-compatible human serum, penicillin (unless the patient is allergic), gentamicin
  • the specimen(s) is transported to the laboratory in a sterile container and placed on a sterile dissection board in a laminar flow hood.
  • a small reprensentative portion is taken for pathologic analysis, and the rest is minced into pieces roughly 4 mm in diameter.
  • These are placed into an enzyme solution of collagenase, DNAse type I, and hyaluronidase type V for overnight digestion at room temperature.
  • the resulting suspension is filtered through a wire mesh to remove any large debris, washed in saline, and placed on Ficoll-Hypaque gradients.
  • the interface containing viable lymphocytes and tumor cells is collected and washed in saline, and a portion is frozen for subsequent use as targets.
  • TIL cultures are initiated at 5 ⁇ 10 5 ml viable cells (tumor plus lymphocytes) in 80% fresh medium/20% LAK supernatants.
  • the fresh medium is AIM-V supplemented with penicillin, fungizone, and 6000 IU/ml IL-2; for the other half, the fresh medium is RPMI supplemented with 10% human serum, penicillin, gentamicin, fungizone, and 6000 IU/ml IL-2.
  • the cultures are placed into 6-well tissue culture dishes and incubated at 37° in humidified incubators with 5% CO 2 .
  • lymphocyte density is not much increased at the end of seven days in culture, and the cultures are collected, centrifugal, and resuspended at 5 ⁇ 10 5 total viable cells/ml in newly prepared 80%/20% medium mixtures of the same type. Occasionally a culture will have increased lymphocyte density and need medium replenishment prior to seven days.
  • TIL are subcultured by dilution when the density is between 1.5 ⁇ 10 6 and 2.5 ⁇ 10 6 cells per ml; densities of subcultures are established between 3 ⁇ 10 5 and 6 ⁇ 10 5 ml. Cultures are kept in 6-well dishes when the volume is less than 1 liter, and transferred to 3 liter polyolefins bags (Fenwall) when the volume reaches one liter.
  • the subculture from bags are accomplished with Fluid Fill/Weight Units (Fenwall), which are programmed to pump prescribed weights of TIL culture and fresh medium into a new bag.
  • Fluid Fill/Weight Units Frawall
  • the fresh medium used in AIM-5 When subculture volumes exceed 3 liters, the fresh medium used in AIM-5. Cultures growing in serum-containing medium are thus diluted into AIMW, and no further LAK supernatant is added to cultures growing in serum-containing or serum-free medium.
  • Tumor-infiltrating lymphocytes are transduced when the total number of lymphocytes is about 1-5 ⁇ 10 8 or higher. Up t one-half f the TIL culture is centrifuged, the medium is saved, and the cells are resuspended in Viral Supernatant with 5 ug/ml protamine. Multiplicities of infection are about 1.5 to 10. The cells in Vital Supernatant are placed int 800 ml tissue culture flasks at 200 ml/flask and incubated at 37° f r 2 hours. During incubati n, the flasks are agitated every 15 minutes to resuspend the cells. The original medium is centrifuged to remove any remaining cells and decanted into new containers.
  • the cells are centrifuged and resuspended in the original cleared medium. If the density is such that subculturing is necessary, the cells are diluted slightly to a density of about 10 6 /ml and placed into fresh 6-well tissue dishes for continued incubation. The following day, the above transduction procedure is repeated. If the cell density at the conclusion of this second transduction is such that subculturing is necessary, the cells are diluted to 5 ⁇ 10 5 /ml for continued incubation.
  • TIL When TIL are to be selected to G418, the TIL are cultured for 3 to 5 days after the second transduction and then G418 is added directly to the culture bags to a final concentration of 300 ug/ml G418. After 10 to 20 days the cells are washed and resuspended at 3 to 6 ⁇ 10 5 cells/ml in fresh medium not containing G418 and then cultured as described above.
  • TILs When the total TILs for a patient are ready for harvest, 5 ⁇ 10 6 cells are taken for cytological examination. Cytospins are examined for the presence of remaining tumor. At least 200 cells are studied and therapy proceeds only when no tumor cells are found. Other TIL samples are taken for characterization of cells surface markers and for assessment of cytotoxicity. Briefly, TILs are stained with fluorescent-labeled antibodies (Leu2, Leu3, Leu4, Lue7, Leu11, Leu15, Leu19, LeuM3, HLADR, and Tac). Chromium release assays are performed with K562, Daudi, autologous tumor, and allogeneic tumor targets.
  • the TILs are collected in two or more batches by continuous flow centrifugation. Some of the TILs are then cryopreserved for future use in 10 10 cell aliquots.
  • TIL for infusion TIL are reharvested.
  • one liter of saline for injection is pumped through the collection chamber and the centrifuge is stopped.
  • TILs are resuspended in the collection bag, the centrifuge is started again, and another liter of saline is pumped through to fully wash the TILs free of tissue culture medium components.
  • the cells are then filtered through a platelet administration set into 600 ml transfer packs (Fenwall) and 50 ml of 25% albumin and 450,000 IU of IL-2 are added to the 200 to 300 ml volume of cells in saline.
  • the TIL are infused over 30 to 60 minutes through a central venous catheter.
  • TNF-TIL TNF-modified TIL
  • Fresh peripheral blood mononuclear cells are separated from the red cells and neutrophils by Ficoll-Hypaque density gradient centrifugation. The MNCs are then washed, counted and cultured at approximately 1 ⁇ 10 6 cells/well in 24-well tissue culture plates in AIM-V which consists of AIM-V (GIBCO) with 2 mM glutamine, 50 U/ml penicillin, 50 ⁇ g/ml streptomycin, 2.5 ⁇ g/ml Fungizone and 25-1,000 U/ml f IL-2 (Cetus). At the initial plating, 10 ng/ml OKT3 (Ortho) monoclonal antibody is added to each well. The cells are cultured at 37° C. in a humidified incubator with 5% CO.
  • AIM-V which consists of AIM-V (GIBCO) with 2 mM glutamine, 50 U/ml penicillin, 50 ⁇ g/ml streptomycin, 2.5 ⁇ g/ml Fungizone and
  • LASN vector Bood, V L 72(2) pages 876-811 2 ml.
  • LASN vector-containing supernatant contianing protamine 5-10 ⁇ g/ml and up to 1,000 U/ml IL-2 are added to the wells after aspirating off the top half of the medium. This is repeated 1-2 times daily for a period of up to 7 days. After the final exposure to retroviral vector the cells are fed with fresh AIM-V and cultured another 2-7 days to permit the cultures to return to exponential growth.
  • T cells Approximately 80% of the culture 0.1-2.5 ⁇ 10 10 T cells are infused into the patient and the remaining cells cryopreserved for future use or returned to culture for studying growth and selection procedures, phenotype analysis, T cell repertoire analysis and percentage of cells demonstrating vector integration.
  • the transduced cells are harvested, washed, and resuspended in normal saline.
  • the final cell preparation is filtered through a platelet filter and transferred into a syrinige or transfusion pack for infusion.
  • a test dose of 5% of the total volume is infused by peripheral vein followed by an observation period of 5-10 min.
  • Transduced cells were administered to patient # 1 in the amounts and at the times shown in FIG. 4 .
  • the ADA enzyme levels of patient # 1 are shown in FIG. 5 .
  • the TNF-NeoR vector was constructed as in Example 4, and retroviral vector supernatant is prepared and tested as disclosed therein.
  • Tumor cell lines are established in tissue culture from tumor fragments or single cell suspensions using standard tissue culture techniques. (Topalian, et al., J. Immunol, Vol. 144, pgs. 4487-4495 (1990)). Tumor and normal tissue are obtained immediately after surgery.
  • the tumors were minced into 1 mm 3 fragments and dissociated with agitation in serum free DMEM (Dulbecco Modified Eagle Medium)(Bio-fluids) containing 2 mM glutamine, 0.1 mg/ml hyaluronidase, 0.01 mg/ml DNase I an d0.1 mg/ml collagenase for 3 hours at room temperature.
  • DMEM Denbecco Modified Eagle Medium
  • the cell suspension was then centrifuged at 800 g for 5 minutes and the pellet resuspended in a culture medium consisting of 5 ml of DMEM high glucose (4.5 g/l) with penicillin and glutamine supplemented with 10% fetal calf serum.
  • the cells were either centrifuged prior to being frozen in 90% FCS, 10% DMSO at ⁇ 80° C., or plated in appropriate dishes or culture flasks in culture medium. Plated cells were incubated at 37° C. in a humidified atmosphere of 5% CO 2 and 95% air. Within 48 hrs, the culture medium was changed in order to remove all non-attached material. Subsequently, cultures were incubated for a period of 6 to 8 days without medium change. The tum rs grow as adherent monolayers in tissue culture flasks (Falcon #3028; 175 cm ⁇ 2 ; 750 ml) containing about 50 ml of medium.
  • the medium When the cells are actively growing and not yet confluent the medium will be poured off and 30-50 ml of medium containing the retroviral supernatant with 5 ⁇ g/ml protamine will be added to the flask. Cornetta, et al., J. Virol. Meth., Vol. 23, Pgs. 186-194 (1989). The flasks will be incubated at 37° C. for six hours at which time the medium will be changed. This procedures will be repeated up to three times. After 24 to 48 hours medium containing 300 ⁇ g/ml G418 is added directly to the flask and the ells will be grown and subcultured for 7 to 14 days in G418 containing medium. The G418 concentrations may be raised to 1 mg/ml depending on the health of the culture.
  • Sterility will be assured by testing for aerobic and anaerobic bacteria, fungus and mycoplasma.
  • TNF protein assay to assure the production of TNF.
  • Cells must be producing at least 100 pg TNF/10 6 cells/24 hours.
  • Gene-modified tumor cells are harvested from the culture flasks by exposure to 0.25% versene (EDTA) for 10 minutes. The cells are washed three times by suspension in 50 mls normal saline and centrifugation. The final cell pellet will be suspended in normal saline and counted. 2 ⁇ 10 8 viable cells in 1 ml normal saline are injected subcutaneously just beneath the skin in the anterior mid thigh and the overlying skin marked with a tatoo dot. If 2 ⁇ 10 8 cells are not available, fewer may be given but not less than 2 ⁇ 10 7 cells will be injected.
  • EDTA 0.25% versene
  • the patient will receive two intradermal injections (separated by 1 cm) of 2 ⁇ 10 7 gene modified tumor cells in 0.1 ml normal saline and these sites also marked by a tatoo dot. These sites are monitored weekly by a physician.
  • the patient undergoes excisional biopsy of superficial inguinal lymph nodes (without formal dissection) in the area draining the inoculation site for growth of lymphocytes. If tumor grows at any of these sites they will be excised when they reach 1 to 2 cm for growth of TIL. If no tumor growth is evident that the sites of tumor injection will be excisionally biopsied at 8 weeks after injection of pathologic analysis.
  • peripheral blood lymphocytes are collected by leukapheresis for four hours, are processed, and then cultured as described in Example 4.
  • TIL tumor necrosis virus
  • a patient's condition has deteriorated to an unacceptable level, or if the patient has developed significant cardiac, renal, pulmonary, or hematologic dysfunction, then the patient will not receive the infusion of TIL or of IL-2 as described hereinbelow.
  • lymphocytes f r a patient are ready f r harvest
  • 5 ⁇ 10 6 cells are taken from cyt 1 gical examination. Cytospins are examined f r the presence f remaining tum r. At least 200 cells are studied and therapy proceeds only when no tumor cells are found.
  • Other lymphocyte samples are taken f r characterization of cell surface markers and for assessment of cytotoxicity. (Berd, et al., Cancer Res., Vol. 46, pgs. 2572-2577 (1986)). Briefly, lymphocytes are stained with fluorescent-labeled antibodies (Leu2, Leu3, Lue4, Lye7, Lue11, Leu5, Leu9, LeuM3, HLADDR and Tac). Chromium release assays are performed with K562, Daudi autologous tumor, and allogeneic tumor targets.
  • TIL infusion TIL
  • saline for injection is pumped through the collection chamber and the centrifuge is stopped. Lymphocytes are resuspended in the collection bag, the centrifuge is started again, and another liter of saline is pumped through to wash fully the TIL's free of tissue culture medium components.
  • the cells are then filtered through a platelet administration set into 600 ml transfer packs (Fenwal), and 50 ml of 25% albumin and 450,000 IU of IL-2 are added to the 200 to 300 ml volume of cells in saline.
  • the TIL are infused over 30 to 60 minutes through a central venous catheter.
  • the recombinant IL-2 used in this trial is provided by the Division of Cancer Treatment, National Cancer Institute (supplied by Cetus Corporation, Emeryville, Calif.) and will be administered exactly as specified in Rosenberg, et al., New Engl. J. Med., Vol. 323, pgs. 570-578 (1990).
  • the IL-2 is provided as a lyophilized powder and will be reconstituted with 1.2 ml/vial. Each vial contains approximately 1.2 mg of IL-2 (specific activity 18 ⁇ 10 6 IV/mg). Less than 0.04 ng of endotoxin are present per vial as measured by the limulus amebocyte assay.
  • Each vial also contains 5% mannitol and approximately 130-230 ⁇ g of sodium dodecyl sulfate/mg of IL-2.
  • the IL-2 is diluted in 50 ml of normal saline containing 5% human serum albumin, and is infused intravenously at a dose of 720,000 IU/kg over a 15 minute period every 8 hr, beginning from two to 24 hr after the TIL infusion.
  • IL-2 will be given for up to five consecutive days as tolerated. Under no circumstances is more than 15 doses of IL-2 be administered. Doses may be skipped depending on patient tolerance. Doses will be skipped if patients reach grade III or grade IV toxicity. If this toxicity is easily reversed by supportive measures then additional doses may be given.
  • Patients may receive concomitant medications to control side effects.
  • the patients may be given acetaminophen (650 mg every 4 hours), indomethacin (50-75 mg every 6 hours), and ranitidine (150 mg every 12 hours) throughout the course of the treatment.
  • Patients may also receive intranveous meperidine (25-50 mg) to control chills if they occur.
  • Hydroxyzine hydrochloride may be given (25 mg every 6 hours) to treat pruitia, if present.
  • Example 6 the procedure of Example 6 were again followed, except that the vector employed to generate vector particles and retroviral vector supernatant, was the IL-2-NeoR vector.
  • This vector was constructed from the vector LXSN (Miller, et al, 1989), and contains the Il-2 gene.
  • LXSN Miller, et al, 1989
  • primary human cells which may be genetically engineered in accordance with the present invention include, but are not limited to, endothelial cells, epithelial cells, keratinocytes, stem cells, hepatocytes, connective tissue cells, fibroblasts, mesenchymal cells, msothelial cells, and parenchymal cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Mycology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Toxicology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Primary human cells which are genetically engineered with DNA (RNA) encoding a marker or therapeutic which is expressed to be expressed in vivo. Such engineered cells may be used in gene therapy.

Description

This is a continuation of application Ser. No. 904,662, filed Sep. 8, 1992, now abandoned, which is a continuation-in-part of U.S. application Ser. No. 868,794, filed Apr. 15, 1992, abandoned, which is a continuation-in-part of U.S. Ser. No. 807,446, filed Dec. 13, 1991, abandoned, which is a continuation-in-part of U.S. Ser. No. 365,567, abandoned.
BACKGROUND OF THE INVENTION
1. Field of the Invention
This invention relates to the use of primary human cells as vehicles for human gene transfer. More particularly, this invention relates to the use of human cells (such as, for example, but not limited to, human blood cells) as vehicles for the transfer of human genes encoding therapeutic agents and/or genes encoding detectable markers.
2. Background Information
Retroviral-mediated gene transfer is a new therapeutic approach for the treatment of human disease (W. F. Anderson, Science 226:401 (1984)). Initial attention has centered on candidate diseases affecting the bone marrow such as the hemoglobinopathies and severe combined immunodeficiency. Early attempts at bone marrow gone transfer in large mammals and primates were only partially successful. As an additional approach, specific blood cells, for example lymphocytes, have been used. Lymphocytes have several features which make them potentially attractive cellular vehicles for gene therapy (K. Culver, et al., J. Cellular Biochemistry Suppl. 12B:171 (1988); R. M. Blaese, et al, Clin. Research 37:599A (1989)).
Lymphocytes are readily available from peripheral blood as a single cell suspensions and they are easily manipulated in tissue culture where the availability of recombinant growth-factors such as rIL-2 permits their expansion by thousands of fold. This adaptability to tissue culture allows serial attempts at gene insertion, selection procedures and time to test for gene expression and other properties of the gene-transduced cells prior to their return to the patient. Long-lived antigen-specific memory lymphocytes proliferate when exposed to their appropriate antigen and thus the population of gene-treated lymphocytes can be selectively and specifically expanded in vivo by immunization of the host. Finally, some populations of antigen-specific lymphocytes “target” to sites in the body containing deposits of antigen. Therefore, it is expected that gene-treated antigen-specific lymphocytes can be used to deliver specific gene products directly to the site of pathology, such as a tumor, in a treated patient. For example, clinical studies with systemically administered recombinant cytokines alone and in conjunction with tumor infiltrating lymphocytes of certain cancers (S. A. Rosenberg, et al, New Engl. J. Med. 318:889 (1987); S. A. Rosenberg, et al., J. Natl. Cancer Institute 80:1393-1397 (1988)). It is expected that TIL transduced with genes promoting secretion of such a cytokine and using the TILs wn unique antigen-specific recept rs to target them to deposits f tumor will permit greater antitumor effect with less systemic toxicity.
SUMMARY OF THE INVENTION
In accordance with an aspect of the present invention, there is provided primary human cells which are genetically engineered with DNA (RNA) which encodes a therapeutic agent of interest, and the genetically engineered cells are employed as a therapeutic agent. (The term “therapeutic,” as used herein, includes treatment and/or prophylaxis).
A primary cell is one which will not have indefinite growth in culture, i.e., the cells has not been manipulated or transformed to provide for indefinite growth in culture.
The DNA (RNA) which is used for transducing the human cells may be one whose expression product is secreted from the cells. In another embodiment, the DNA (RNA) cures a genetic deficiency of the cells and the expression product is not secreted from the cells (for example, the expression product of DNA encoding ADA is not secreted from the cells).
The human cells may also be genetically engineering with DNA (RNA) which functions as a marker, as hereinafter described in more detail.
Thus, in accordance with another aspect of the present invention, there are provided primary human cells which are genetically engineered to include DNA which encodes a marker or therapeutic, with such cells expressing the encoded product in vivo.
In one embodiment, the invention is directed to a method of enhancing the therapeutic effects of human primary cells. For example, there is provided a method of enhancing the therapeutic effects of human primary cells which specifically “target” to a tissue site in a patient, whereby DNA (RNA) which encodes an agent that enhances the therapeutic effects is inserted in the cells. The tissue site can be, for example, a tumor. The DNA (RNA) produces the agent in the patient's body, and in accordance with such embodiment the agent is expressed at the tissue site itself. Similarly, as hereinabove indicated, primary human cells which are genetically engineered need not be targeted to a specific site and in accordance with the invention, such engineered primary human cells function as a systemic therapeutic; e.g., a desired therapeutic agent can be expressed and secreted from the cells systemically.
In one embodiment, the primary human cells may be primary human nucleated blood cells or primary human tumor cells.
In a preferred embodiment, the primarily human cells are primary human nucleated blood cells.
The primary human blood cell which can be used in the present invention include leukocytes, granulocytes, monocytes, macrophages, lymphocytes, immature forms of each of the previous cells (as well as immature erythroblasts) (CD34+cells) and totipotent stem cells.
In one embodiment, human cells, preferably blood cells, which are genetically engineering are cells which are targeted to a specific site; for example, the cells can be tumor infiltrating lymphocytes (TIL cells). In such case, the engineered cells function as a therapeutic at such site. In another aspect, the cells are n t cells which are targeted to a specific site and in such aspect such cells function as a systemic therapeutic.
The DNA carried by the cells can be any DNA having clinical usefulness, for example, any DNA that directly or indirectly enhances the therapeutic effects of the cells. Alternatively, the DNA carried by the cells can be any DNA that allows the cells t exert a therapeutic effect that the cells would not exert. Examples of suitable DNA, which can be used for genetically engineering, f r example, bl od cells, include those that encode cytokines such as tumor necrosis factor (TNF), interleukins (for example, interleukins 1-12), interferons (α,β,γ-interferons). T-cell receptor proteins and the Fc receptors for antigen-binding domains of antibodies, such as immunoglobulins.
In another embodiment, the inserted gases are marker genes which permit determination of the traffic and survival of the transformed cells in vivo. Examples of such maker genes include the neomycin resistance (neoR) gene, multi-drug resistant gene, thymidine kinase genes, β-galactosidase, dehydrofolate reductase (DHFR) and chloroamphenicol acetyl transferase.
Thus, in accordance with another embodiment, there is provided a process for treating a patient with a therapeutic agent by providing the patient with primary human cells, preferably nucleated blood cells, genetically engineered with DNA (RNA) encoding such therapeutic agent.
The cells may be genetically engineered in vitro or in vivo. For example, cells may be removed from a patient; genetically engineered in vitro with DNA (RNA) encoding the therapeutic agent, with such genetically engineered cells being readministered to the patient. Such a treating procedure is sometimes referred to as an ex vivo treatment.
Alternatively, DNA (RNA) encoding the therapeutic agent may be administered to the patient for delivery of the DNA in vivo to the targeted cells. Such may be accomplished by the use of a variety of delivery systems, e.g., a retroviral or other viral vector; liposomes etc.
Thus, in accordance with another embodiment, a patient is provided with human primary cells which are genetically engineered with DNA (RNA) encoding a therapeutic agent whereby such therapeutic agent may be expressed in vivo. As hereinabove indicated, such genetically engineered cells may be provided by administering to the patient cells which have been genetically engineered ex vivo or by administering the DNA (RNA) as part of a delivery system for genetically engineering targeted cells in vivo.
In accordance with another embodiment, there are provided primary human cells genetically engineered with DNA (RNA) encoding a therapeutic agent.
In accordance with a further aspect of the present invention, there is provided a composition comprising (i) primary human cells genetically engineered with DNA (RNA) encoding a therapeutic agent and (ii) a pharmaceutically acceptable carrier suitable for administration to a patient. Preferably, the primary human cells are primary human nucleated blood cells. The carrier may be a liquid carrier (for example, a saline solution) or a solid carrier, e.g., an implant. In employing a liquid carrier, the engineered cells may be introduced, e.g., intravenously, sub-cutaneously, intramuscularly, intraperitoneally, intralesionaly, etc.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1A dem nstrates by assay for functi nal enzyme that murine 14.1-T cells containing an inserted SAX vector express high levels of the neomycin resistant (neoR) gene product, aeomycin phosphotranferase (NPT). FIG. 1B demonstrates the expression f human adenosine deaminase (hADA), also encoded by the SAX vector inserted in the murine 14.1-T cells.
FIG. 2 shows the proliferative response of murine splenic lymphocytes and 14.1-T cells to stimulation with phytohemagglutinin and IL-2and the effect of G418 on this proliferati n.
FIG. 3A demonstrates that human N2-transduced TIL expresses sufficient NPT to become resistant to the toxic effects of G418 on cellular proliferation. FIG. 3B shows growth curves of human N2-transduced and untreated TIL for the 30 day period following gene insertions;
FIG. 4 demonstrates absolute Blood T Cell levels of patient # 1 receiving ADA gene therapy in accordance with the procedure of Example 4. The bar portion of the figure is the transduced T-cells infused at the day indicated; and
FIG. 5 demonstrates the ADA enzyme activity of patient # 1 during treatment.
DETAILED DESCRIPTION OF THE INVENTION
As indicated above, the present invention relates to the use of primary human cells as cellular vehicles for gene transfer. The genes can be any gene having clinical usefulness, for example, therapeutic or marker genes. Preferably, the primary human cells are blood cells. The term “blood cells” as used herein is meant to include all forms of nucleated blood cells as well as progenitors and precursors thereof, as hereinabove described.
In one embodiment, the invention is directed to a method of enhancing the therapeutic effects of human primary cells, preferably blood cells, that are infused in a patient, comprising: (i) inserting into the human primary cells of a patient a DNA (RNA) segment encoding a product that enhances the therapeutic effects of the human primary cells; and (ii) introducing cells resulting from step (1) into the patient. The gene can be inserted into the human primary cells using any gene transfer procedures, for example, retroviral-mediated gene transfer, electroporation, calcium phosphate mediated transfection, microinjection or proteoliposomes. Other vectors can be used besides retroviral vectors, including those derived from DNA viruses and other RNA viruses. As should be apparent when using an RNA virus, such as virus includes RNA which encodes the desired agent, with the human primary cells which are genetically engineered with such RNA virus thus being provided with DNA encoding the agent.
More specifically, there is provided a method of enhancing the therapeutic effects of blood cells, that are infused in a patient, comprising: (i) inserting into the blood cells of a patient a DNA (RNA) segment encoding a product that enhances the therapeutic effects of the blood cells; and (ii) introducing cells resulting from step (i) into the patient under conditions such that the cells resulting from step (i) “target” to a tissue site. In the alternative, as previously described the cells are not “targeted” and function as a systemic therapeutic. The genes are inserted in such a manner that the patient's transformed blood cell will produce the agent in the patient's body. In the case of antigen-specific blood cells which are specific for an antigen present at the tissue site, the specificity of the blood cells f r the antigen is n t lost when the cell produces the product.
Alternatively, as hereinbefore indicated, DNA (RNA) may be inserted into the blood cells of a patient, in vivo, by administering such DNA (RNA) in a vehicle which targets such blood cells.
The method of the invention can-be used, for example, in the treatment of cancer in a human by inserting into human primary cells, such as, f r example, blood cells, which specifically “target” to a tumor and which have been removed from a cancer patient and expanded in culture, genes that enhance the anti-tumor effects of the blood cells. The blood cells can be expanded in number before or after insertion of the genes. The method of gene transfer in the blood cells is as described above. Thus, the procedure is performed in such a manner that upon injection into the patient, the transformed blood cells will produce the agent in the patient's body, preferably at the site of the tumor itself.
The gene carried by the blood cells can be any gene which directly or indirectly enhances the therapeutic effects, of the blood cells. The gene carried by the blood cells can be any gene which allows the blood cells to exert a therapeutic effect that it would not ordinarily have, such as a gene encoding a clotting factor useful in the treatment of hemophilia. The gene can encode one or more products having therapeutic effects. Examples of suitable genes include those that encode cytokines such as TNF, interleukins (interleukins 1-12), interferons (α, β, γ-interferons), T-cell receptor proteins and Fc receptors for antigen-binding domains of antibodies, such as immunoglobulins.
Additional examples of suitable genes include genes that modify blood cells to “target” to a site in the body to which the blood cells would not ordinarily “target,” thereby making possible the use of the blood cell's therapeutic properties at that site. In this fashion, blood cells such as TIL can be modified, for example, by introducing a Fab portion of a monoclonal antibody into the cells, thereby enabling the cells to recognize a chosen antigen. Likewise, blood cells having therapeutic properties can be used to target, for example, a tumor, that the blood cells would not normally target to. Other genes useful in cancer therapy can be used to encode chemotactic factors which cause an inflammatory response at a specific site, thereby having a therapeutic effect. Other examples of suitable genes include genes encoding soluble CD4 which is used in the treatment of AIDS and genes encoding α-antitrypsin, which is useful in the treatment of emphysema caused by α-antitrypsin deficiency.
The gene therapy of the present invention is useful in the treatment of a variety of diseases including but not limited to adenosine deaminase deficiency, sickle cell anemia, thalassemia, hemophilia, diabetes, α-antitrypsin deficiency, brain disorders such as Alzheimer's disease, and other illnesses such as growth disorders and heart diseases, for example, those caused by alterations in the way cholesterol is metabolized and defects of the immune system.
In still another embodiment, there is prodded a method of detecting the presence of human primary cells, such as, for example, blood cells containing a marker present in a patient, comprising (i) inserting into human primary cells removed from the patient a DNA (RNA) segment encoding the marker under conditions such that the marker is present in the blood cells; (ii) introducing cells resulting from step (i) into the patient; (iii) removing from the patient an aliquote f tissue (which can be, for example, normal tissue, cancerous tissue, blood, lymph nodes, etc.) including cells resulting from step (ii) and their progeny; and (iv) determining the quantity of the cells resulting from step (ii) and their progeny, in said aliquot.
M re specifically, the present invention comprises a method f detecting the presence f blood cells containing a marker, present at a site in a patient, to which site the blood cells “targets”, c mprising (i) inserting into blood cells removed from the patient that are specific for an antigen (either known or unknown) present at the site and that target to site, a DNA segment encoding the marker under conditions such that the marker is present in the blood cells, (ii) introducing cells resulting from step (i) into the patient under conditions such that cells resulting from step (ii) can be found at the site; (iii) removing from the patient an aliquot of tissue from the site, which aliquote includes cells resulting from step (ii) and their progeny; and (iv) determining the quantity of the cells resulting from step (ii) and their progeny, in said aliquot. The marker gene can be, for example, inserted into blood cells which specifically “target” to a tumor in order to determine the traffic and survival of the transformed blood cells in vivo. Alternatively, the transformed blood cells circulate through the body, rather than targeting to a particular tissue site.
The marker gene can be any gene which is different from the genes in the blood cell into which the marker gene is inserted. Examples of such marker genes include neoR, multi-drug resistant gene, thymidine kinase gene, β-galactosidase, dehydrofolate reductase (DHFR) and chloroamphenicol acetyl transferase.
The marker gene can be inserted into a human primary cell, such as a blood cell, together with a therapeutic gene or separately. The marker gene and the therapeutic gene may also be one and the same.
To initially test the feasibility of using lymphocytes for gene transfer, the retroviral gene transfer vector SAX (P. W. Kantoff, et al, PNAS 83:6563 (1986)) was used to insert the genes for human adenosine deaminase (hADA) and neomycin resistance (neoR) into the murine T cell line 14.1 SAX is a moloney virus based vector with the neoR gene promoted from the retroviral LTR and the hADA gene promoted from an internal SV40 promoter. The 14.1 T cell line was derived from the draining lymph nodes of a BIO.D2 mouse previously immunized with sperm whale myoglobin (SWM) by repeated cycles of antigen stimulation of these lymphocytes in tissue culture (I. Berkower, et al., J. Immunol. 135:2628 (1985)). These 14.1 T-cells proliferate when challenged with SWM in the presence of histocompatible antigen-presenting cells (without exogenous IL-2) or when stimulated with PHA and rIL-2 in vitro. An average of 20% of the 14.1-T cells achieved stable insertion of the SAX vector after a supernatant infection protocol (P. W. Kantoff, et al, PNAS 83:6563 (1986)). The population of SAX-transduced 14.1-T cells was selected in the neomycin analog G418 so that all cells were expressing the introduced genes and were then expanded by repeated cycles of antigen stimulation to obtain large numbers of transduced cells. FIG. 1A demonstrates by assay for functional enzyme that the cells containing the inserted SAX vector are expressing high levels of the neoR gene product, neomycin phosph transferase (NPT). FIG. 1B demonstrates expression of human adenosine deaminase (hADA), also encoded by the SAX vector inserted in these T cells. The SAX-transduced, expanded population of 14.1 cells (14.1SAX) were functionally unchanged except f r the gene insertion and its expressi n which provides for G418 resistance, the cells remained responsive to SWM in vitro.
A thymic nude mice were then injected intraperitoneally with 20×106 14.1SAX T-cells and immunized with 100 μg soluble SWM. Athymic recipients were chosen for these experiments because they have very little endogenous Te cell activity and therefore any T cells recovered from these mice at a later date should represent the original injected 14.1 SAX cells or their progeny.
Thirty-seven days after intraperitoneal injection of the 14.1SAX-T cells, spleens from recipient athymic mice were removed and tested to determine if any cells expressing the introduced genes could be detected by demonstrating G418 resistance and hADA. FIG. 2 shows the proliferative response of splenic lymphocytes and 14.1 cells to stimulation with phytohemagglutinin and IL-2 and the effect of G418 on this proliferation. While control nude mouse splenocytes did proliferate modesty to PHA+IL-2, this response was abolished by low concentrations of G418. By contrast, PHA and IL-2 responsive lymphocytes recovered from the spleens of nude mice injected with 14.1SAX-T cells 37 days earlier remained resistant to G418, indicating persistence of the transferred 14.1 cells as well as continued expression of the introduced neoR gene. These G418-resistant recovered T cells were further expanded in IL-2, tested for expression of the hADA isozymes by cellulose acetate electrophoresis and were found to be expressing the product of this introduced gene as well. Thus, antigen-specific T cells maintained in tissue culture and transduced with foreign genes by retroviral-mediated gene transfer were able to survive and express the introduced genes for at least 37 days when reinjected into an intact host animal.
Because of the success in demonstrating that cultured T lymphocytes carrying inserted foreign genes could survive for prolonged periods of time in mice and continue to express the inserted genes, situations were investigated where the technique might be used in mass. S. A. Rosenberg, et al, Science 223:1318 (1986) have shown that the treatment of experimental cancer with T lymphocytes which have been isolated directly from the tumor and expanded in culture with rIL-2 can cause tumor regression . Techniques for growing the tumor infiltrating lymphocytes (TIL) from human cancers have been developed and the functional and phenotypic properties of these cells have been extremely characterized (L. M. Muul, et al, J. Immunol. 138:989 (1987); S, Topalian et al, J. Immunol. Meth. 102:127 (1987); A. Belldegrum, et al, Cancer Res. 48:206 (1988)). In the initial clinical studies using human TIL along with intravenous infusion of rIL-2 in 20 patients with advanced metastatic melanoma, objective remissions with at least 50% reduction of tumor burden were seen in 55% of the patients (S. A. Rosenberg et al, N. Engl. J. Med. 319:1676 (1988)). These were patients who had failed air conventional forms of cancer treatment and thus TIL therapy is expected to open a significant new direction in the treatment of cancer.
At this state in the development of TIL therapy, many issues remain to be clarified including a more complete understanding of the features of individual TIL preparations which correlate with clinical antitum r effects, the in vivo distribution and time of persistence f TTL after infusion, and whether distinct TIL functional phenotypes localize in the body in ways which will permit prediction of their clinical efficacy.
Trafficking studies using radionuclides as cell labels have shown that TIL from some patents do appear to specifically accumulate in areas of tumor within the first few days, alth ugh these studies have been limited by the short half-life of the clinically acceptable isotopes (2.8 days for In) and the high rate f spontaneous release of the isotope from the TIL (B. Fisher, et al, J. Clin. Oncol. 7:250 (1989)). Genes used as labels could potentially solve several of the problems associated with radionucleotide labels. Genes become stably integrated into the genome of the target cell and then will be completely destroyed when the cell dies so that any detected label should only be associated with the original marked cell or its progeny. In contrast to radionuclide tags which become diluted as cell proliferate, a gene label will also equally mark progeny cells derived by continued proliferation of the originally labelled cell population as occurs when TIL proliferation is driven by IL-2 infusions given to the patient. An important characteristic that is unique to the use of genes as cell labels in their ability to introduce an entirely new functional property to the target cell. With the gene neoR, for example, the target cell acquires resistance to the neomycin analog G418 which permits, as we have shown for mouse 14.1SAXT-cells, the selective recovery of the gene-marked cells by regrowth of recovered cells in the presence of G418. With recovery, the cells from different sites could then be analyzed for their functional phenotypes and potential correlations with clinical efficacy.
To explore the possibility that inserted genes might be used to label TIL, cultured TIL from 6 patients with metastatic malignant melanoma were studied using the retroviral vector N2, which vector was described by (D. Armentano, et al, J. Virology 61:1647 (1987)). N2 contains the gene neoR promoted by the retroviral LTR and has been used extensively by the inventors in studies of bone marrow gene-transfer in mide and primates during the past 5 years (M. E. Egitis, et al, Science 230:1396 (1985); D. Kohn, et al, Blood Cells 13:285-298 (1987)); P. W. Kantoff, et al, J. Exp. Med. 166:219 (1987)). Human TIL were efficiently transduced with N2 by exposure to viral supernatant produced by the amphotrophic packaging cell line PA317 which package cell line was described by (A. D. Miller, et al, Molecular Cellular Biol. 6:2895 (1986)). The N2-TRANSDUCED TIL expressed sufficient neomycin phosphotransferase (NPT) to become resistant to the toxic effects of G418 on cellular proliferation as demonstrated in FIG. 3A. On average, 10-15% of the N2-treated TIL population grew in G418 while >99% of the untreated TIL died. After 10 days of selection in 0.3 mg/ml G418, the surviving N2-treated population was G418 resistant. FIG. 3B shows growth curves of N2-transduced and untreated TIL for the 30 day period following gene insertion. Growth of the N2-transduced TIL population as a whole slows initially when G418 is added to the culture (not shown, but resumes exponential expansion as growth of the NeoR-expressing G418-resistant subpopulation becomes established. After completion of selection, the N2-transduced cells as a rule grew at a rate comparable to that of TIL not transduced with the neoR gene.
To further characterize these cells and to study possible differences between th gene-transduced and non-transduced TIL, studies of the cell membrane phenotype and the constitutive cytotoxic properties f the tw cell rpus have been performed (see Table 1 below).
TABLE 1
CHARACTERISTICS OF N2-TRANSDUCED AND
NON-TRANSDUCED TIL
N2-transduced and G418
Untreated TIL Selected
Phenotype Cytotoxicity Phenotype Cytotoxicity
% Positive % 56Cr Release % Positive % 56Cr Release
Patient CD4 CD8 Auto NK CD4 CD8 Auto NK
1 0 97.9 0 0 0 98.7 0 0
2 81.8 3.9 0 0 88.6 1.4 0 0
3 30.7 60.5 26.4 0 6.9 82.2 26.1 4.7
4 76.5 13.7 0 0 30.5 68.6 0 0
5 0.1 97.7 22.5 0 0.7 98.2 25.5 0
6 8.9 94.7 4 0 5.4 95.0 2 0
TIL transduced with N2 and selected with 0.3 mg/ml G418 for 10-14 days were compared with non-transduced TIL from 6 patients with methastatic malignant melanoma. These populations were analyzed for their surface membrane phenotype as revealed by FACS analysis of 1×106 TIL stained monoclonal antibodies. Antibodies to CD4(Leu 3) and CD8 (Leu 2) were used on populations and the percentage of the cells positive for each determinant are shown. Constitutive cytotoxic function of each TIL population was determined for autologous tumor targets (auto) and the NK sensitive target K562 (NK) as well as for allogeneic tumor and sensitive target cells (not shown) by a standard 4 hour 56Cr release assay at E:T ratios of 40:1, 15:1, and 4.5:1. Data shows target lysis at the 40:1 E:T ratio. TIL isolated from different individual donors have different phenotypic profiles as determined by various cell membrane determinants (CD4, CD8, etc.) and different levels of cytotoxic activity toward autologous tumor and other target cell types. Human TIL of both major phenotypic subgroups (CD4 and CD8) were readily transduced by the N2 vector and expressed G418 resistance. Some transduced and selected TIL populations (patients 3 and 4) showed more drift in their phenotypic composition than others, and as expected with more prolonged culture, all of the TIL populations became progressively oligoclonal whether or not they had been transduced with N2 (not shown). TIL are often cytolytic for autologous tumor cells but not other target cell types in vitro. Of this group of six patients, TIL isolated from two were cytolytic to autologous tumor cells, but not alleogeneic melanoma tumor cells, NK (K562) or LAK (Daudi) sensitive targets. The functional cytotoxic activity profile of the TIL was unaffected by N2 transduction and G418 selection, even in those TIL populations showing some drift in T cell subset composition measured by surface membrane antigen phenotype.
The present invention will be illustrated in detail in the following examples. These examples are included for illustrative purposes and should not be considered to limit the present invention.
EXAMPLE 1
The following experiment demonstrates that murine 14.1-T cells containing an inserted SAX vector express high levels of the neoR gene product, NPT, as well as expressing hADA.
Long term cultures of the murine T cell line 14.1 were maintained by repeated cycles of stimulation with 4.0 μM SWM in the presence of fresh irradiated (3000 R) B10.D2 spleen cells as antigen-presenting cells. Four days after antigen stimulation, the cultures were fed with complete fresh medium and placed n irradiated syngeneic spleen cells f r a 10 day rest phase. No ex gen us IL-2 was used with this T cell line. T introduce ex genous genes int these 14.1-T cells, 3 days after antigen stimulation the proliferating activated 14.1 cells were exposed to SAX supernatant (MOI=5) in the presence of 8 μg/ml polybrene for two sequential 4 hour incubation.
Twenty-four hours later the SAX-transdermal 14.1 cells were placed under selection with 0.3 mg/ml G418 for 14 days. The SAX-transduced G418 selected cells were then expanded by repeated 14 day cycles of antigen stimulation and rest on syngeneic feeder cells. Lysates from the G418-selected 14.1 population were prepared and assayed for the presence of NPT by measuring the phosphorylation of kanamycin with P32 labelled ATP after separation of NPT from endogenous phosphotransferases on a non-denaturing polyacrylamide gel (B. Reiss, et al, Gene 30:211 (1984)).
As shown in FIG. 1A, the 14.1 cells containing the inserted SAX vector expresses high levels of NPT.
Lysates of the 14.1 cells were separated by electrophoresis on cellulose acetate (B. Lira, et al, Molecular Cellular Biol. 7:3459 (1987)) which was then stained for the presence of the murine and human isozymes of ADA (FIG. 1B). The human T cell line CEM was included as a control for the migration of hADA.
The results are shown in FIG. 1B which demonstrates the expression of hADA.
EXAMPLE 2
The following experiment was performed in order to demonstrate the continued expression of a transferred gene in gene modified T-cells. Specifically, the effect of G418 on the proliferative response of murine splenic lymphocytes and 14.1-T cells to stimulation with phytohemagglutinin and IL-2 was studied.
Cultures of 14.1-T cells, 14.1SAX transduced T cells, splenic lymphocytes from nude mice and splenic lymphocytes recovered from nude mice that had been given an intraperitioneal injection of 20×10614.1SAX T cells 37 days earlier were established with 2×105 cells/ml in flat bottom microculture trays using medium and culture conditions previously described (L. Berkower, et al, J. Immunol. 135:2628 (1985) Proliferation in response to stimulation with 5 μg/ml PHA (Wellcome) and 100 n/ml rIL-2(Cetus Corporation) was measured as the incorporation of 3H thymidine into cellular DNA following an overnight pulse with 0.4 μCl added after 72 hours of culture. G418 prepared as the concentrations of active drug indicated (FIG. 2) was added at the initiation of culture. Cultures were harvested for scintillation counting using a Sakron TM cell harvesting system.
FIG. 2 shows, as the mean of triplicate cultures, the proliferative response of the murine splenic lymphocytes and 14.1-T cells to stimulation with phytohemaglutimin and IL-2 and the effect on G418 on this proliferation. A review of the figure shows that while control nude in use splenic lymphocytes proliferated modestly to PHA+IL-2, this response was abolished by low concentrati ns of G418. By contrast, PHA and IL-2 responsively lymphocytes recovered from the spleens of nude mice injected with 14.1SAX-T cells 37 days earlier remained resistant to G418, indicating persistence of the transferred 14.1 cells as well as continued expression of the introduced neoR gene.
EXAMPLE 3
The experiment described bel w was perf rmed in order to verify that human N2-transduced TIL expresses sufficient NPT to become resistant to the toxic effects of G418 on cellular proliferation.
TIL grows from a tumor biopsy obtained from a patient with metastatic malignant melanoma were exposed to two sequential 4 hour exposure to N2 supernatant (average viral titer 106/ml, MOI=2) in the presence of 5 μg/ml protamine sulfate (K. Kornetta, et al, J. Viorol. Meth. 23:187 (1989)). Twenty-four hours later an aliquot was removed and placed under selection in 0.3 mg/ml G418 for 10 days. The cells were then to grown without G418 for two weeks and then were tested for the effect of various concentrations of G418 on their proliferative response to 1000 u/ml IL-2.
The untreated TIL (no vector) was compared with TIL which had been transduced with N2 and selected with G418 (N2+G418) and with an N2-transduced TIL population which had not been further selected in G418 (N2). Proliferative was measured by 3H-thymidine incorporation as described in Example 2.
The results are shown in FIG. 3A, wherein it may be seen that, on average, 10-15% of the N2-treated TIL population grew in G418 while >99% of the untreated TIL died After 10 days of selection in 0.3 mg/ml G418, the surviving N2 treated population was G418 resistant.
FIG. 3B shows the growth curves of the N2-transduced and the untreated TIL for the 30 day period following gene insertion. These results were obtained using the following procedure.
TIL from patent 1 were grown from a tumor biopsy as described (Tobalian et al, J. Immune Method 138:4006-4011 (1987)) and then an aliquot was transduced with the N2 vector by two 4-hour exposures to viral supernatant (note: LNL 6 is identical to N2 except that LNL6 has several additional safety features). Forty-eight hours later the transduced population was placed under selection for 10 days with 0.3 mg/ml G418. Non-transduced TIL exposed to this selection protocol were all dead by day 10. Growth of the N2-transduced TIL population allowed when placed in G418, but resumed at an exponential rate while still under selection as the cells expressing the neoR gene became dominant by day 10. TIL cultures were fed and split as required (1-2 times per week) and the cumulative cell total calculated when the running total reached >106 cells to that larger numbers of cells would not have to be maintained in culture.
In this example, about 15% of the N2 - treated TIL population expressed the inserted gene sufficiently to permit growth in 0.3 mg/ml G418. After completion of selection, the N2-transduced cells at a rule grew at a rate comparable to that of TIL not transduced with the neoR gene.
EXAMPLE 4—GENE MODIFIED T-CELL CLINICAL PROTOCOL
The TNF-NeoR vector is constructed by modifying the Monkey murine leukemia vector.
Construction f TNF vector
The TNF gene containing water, LT125N, was constructed from the vector LXSN (Miller AD and Rossman G., 1989, Biotechniques 7:980-990.), usign the entire 233 amino acid sequence encoding cDNA f the natural TNF genes (Pennies, et al, 1984, Nature 312, 724-729, Wang, et al, Science 228, 149-154). The ribosome binding site used upstread of the TNF gene was a syntehtic one, a consensus sequence f r translation initiation based on Kozak's rules (Kozak, Nucle. Acids Res. 12, 857-872 (1984) and having the nucleotide sequence 5′ TTCCGCAGCAGCCCGCCACC3′. The vectors construct was packaged using the PA317 packaging cell line (Miller et al., Mol. Cell Biol. 6, 2895-2902 (1986)).
Retroviral vector supernatant is produced by harvesting the cell culture medium from the PA317 packaging line developed by Dr. A. Dusty Miller. This line has been extensively characterized and was used by us in our previous studies of the infusion of TIL modified by the N2 vector. The TNF-Neo vector preparations from PA317 are extensively tested to assure that no detectable replication competent virus is present. Tests for replication-competent virus are conducted on both the vector supernatant and on the TIL after transduction. The follwoing tests are run on the producer line and/or the viral supernatant:
1) The viral titer is determined on 3T3 cells. Viral preparations with titers greater than 5×104 colony forming units/ml are used.
2) Southern blots are run on the producer line to detect the TNF gene.
3) TNF production by the product line is measured and should be significantly above baseline control values. TNF is assayed using standard biologic assays on the L929 amino cell line (Asher et al. J. Immunol 138:963-974, 1987) or by ELISA assay (R&D Systems, Minneapolis, Minn.).
4) Sterility of the producer line and the supernatant is assured by testing for aerobic and anaerobic bacteria, fungus and for mycoplasma.
5) Viral testing is performed including:
    • a. MAP test
    • b. LCM virus
    • c. Thymic agent
    • d. S+L—assay for ecotropic virus
    • e. S+L—for xenotropic virus.
    • f. S+L—for amphotropic virus.
    • g. 3T3 amplification.
6) Electron microscopy is performed to assure the absence of adventitions agents.
Following transduction and growth of the TIL population the following tests are performed on the TIL prior to infusion into patients.
1) Cell viability is greater than 70% as tested by trypan blue dye exclusion.
2) Cytologic analysis is performed on over 200 cells prior to infusion to assure that tumor cells are absent.
3) Sterility is assured by testing for aerobic anaerobic bacteria, fungus and mycoplasma.
4) S+L—assay including 3T3 amplification must be negative.
5) PCR assay for the absence of 4070A envelope gene must be negative.
6) Reverse transcriptase assay must be negative.
7) S uthern blots run on the transduced TIL to assure that intact provirus is present.
8) TNF protein assay to assure the production of TNF. Cells must be producing at least 100 pg TNF/106 cells/24 hours.
9) IL-2 will be withdrawn from the culture medium of an aliqu t f r at least one week to assure that cells do n t exhibit auton mous growth in the absence f IL-2.
10) Cyt toxicity against the autol gous and at least tw other targets are tested. The phenotype of the cells as tested by fluorescence activated cell sorting analysis.
At least tw days prior to surgery, peripheral blood lymphocytes are collected by leukapheresis for four hours. These are Ficoll-Hypaque separated and the mononuclear cells from the interface, washed in saline, and placed in culture in roller bottles at 106 cells/ml. Half are placed into AIM-V (a serum free medium, Gibco Laboratories) with 6000 IU/ml IL-2 (Cetus), and Half are placed into RPMI supplemented with 2% type-compatible human serum, penicillin (unless the patient is allergic), gentamicin, and 600 IU/ml IL-2. After 3 to 4days cells are centrifuged and the supernatants are collected and filtered. These are referred to as LAK supernatants.
Immediately upon tumor resection, the specimen(s) is transported to the laboratory in a sterile container and placed on a sterile dissection board in a laminar flow hood. A small reprensentative portion is taken for pathologic analysis, and the rest is minced into pieces roughly 4 mm in diameter. These are placed into an enzyme solution of collagenase, DNAse type I, and hyaluronidase type V for overnight digestion at room temperature. The resulting suspension is filtered through a wire mesh to remove any large debris, washed in saline, and placed on Ficoll-Hypaque gradients. The interface containing viable lymphocytes and tumor cells is collected and washed in saline, and a portion is frozen for subsequent use as targets.
TIL cultures are initiated at 5×105 ml viable cells (tumor plus lymphocytes) in 80% fresh medium/20% LAK supernatants. For half the cells, the fresh medium is AIM-V supplemented with penicillin, fungizone, and 6000 IU/ml IL-2; for the other half, the fresh medium is RPMI supplemented with 10% human serum, penicillin, gentamicin, fungizone, and 6000 IU/ml IL-2. The cultures are placed into 6-well tissue culture dishes and incubated at 37° in humidified incubators with 5% CO2.
Usually the lymphocyte density is not much increased at the end of seven days in culture, and the cultures are collected, centrifugal, and resuspended at 5×105 total viable cells/ml in newly prepared 80%/20% medium mixtures of the same type. Occasionally a culture will have increased lymphocyte density and need medium replenishment prior to seven days. After this first passage, TIL are subcultured by dilution when the density is between 1.5×106 and 2.5×106 cells per ml; densities of subcultures are established between 3×105 and 6×105 ml. Cultures are kept in 6-well dishes when the volume is less than 1 liter, and transferred to 3 liter polyolefins bags (Fenwall) when the volume reaches one liter. The subculture from bags are accomplished with Fluid Fill/Weight Units (Fenwall), which are programmed to pump prescribed weights of TIL culture and fresh medium into a new bag. When subculture volumes exceed 3 liters, the fresh medium used in AIM-5. Cultures growing in serum-containing medium are thus diluted into AIMW, and no further LAK supernatant is added to cultures growing in serum-containing or serum-free medium.
Tumor-infiltrating lymphocytes are transduced when the total number of lymphocytes is about 1-5×108 or higher. Up t one-half f the TIL culture is centrifuged, the medium is saved, and the cells are resuspended in Viral Supernatant with 5 ug/ml protamine. Multiplicities of infection are about 1.5 to 10. The cells in Vital Supernatant are placed int 800 ml tissue culture flasks at 200 ml/flask and incubated at 37° f r 2 hours. During incubati n, the flasks are agitated every 15 minutes to resuspend the cells. The original medium is centrifuged to remove any remaining cells and decanted into new containers. At the end of 2 hours, the cells are centrifuged and resuspended in the original cleared medium. If the density is such that subculturing is necessary, the cells are diluted slightly to a density of about 106/ml and placed into fresh 6-well tissue dishes for continued incubation. The following day, the above transduction procedure is repeated. If the cell density at the conclusion of this second transduction is such that subculturing is necessary, the cells are diluted to 5×105/ml for continued incubation.
When TIL are to be selected to G418, the TIL are cultured for 3 to 5 days after the second transduction and then G418 is added directly to the culture bags to a final concentration of 300 ug/ml G418. After 10 to 20 days the cells are washed and resuspended at 3 to 6×105 cells/ml in fresh medium not containing G418 and then cultured as described above.
When the total TILs for a patient are ready for harvest, 5×106 cells are taken for cytological examination. Cytospins are examined for the presence of remaining tumor. At least 200 cells are studied and therapy proceeds only when no tumor cells are found. Other TIL samples are taken for characterization of cells surface markers and for assessment of cytotoxicity. Briefly, TILs are stained with fluorescent-labeled antibodies (Leu2, Leu3, Leu4, Lue7, Leu11, Leu15, Leu19, LeuM3, HLADR, and Tac). Chromium release assays are performed with K562, Daudi, autologous tumor, and allogeneic tumor targets.
When the total cell number reaches about 2×1011 cells the TILs are collected in two or more batches by continuous flow centrifugation. Some of the TILs are then cryopreserved for future use in 1010 cell aliquots.
For infusion TIL are reharvested. At the time of cell collection, one liter of saline for injection is pumped through the collection chamber and the centrifuge is stopped. TILs are resuspended in the collection bag, the centrifuge is started again, and another liter of saline is pumped through to fully wash the TILs free of tissue culture medium components. The cells are then filtered through a platelet administration set into 600 ml transfer packs (Fenwall) and 50 ml of 25% albumin and 450,000 IU of IL-2 are added to the 200 to 300 ml volume of cells in saline. The TIL are infused over 30 to 60 minutes through a central venous catheter.
Patients receive either LNL-6 modified TIL or TNF-modified TIL (TNF-TIL).
EXAMPLE 5 ADA CLINICAL PROTOCOL
Fresh peripheral blood mononuclear cells (MNCS) are separated from the red cells and neutrophils by Ficoll-Hypaque density gradient centrifugation. The MNCs are then washed, counted and cultured at approximately 1×106 cells/well in 24-well tissue culture plates in AIM-V which consists of AIM-V (GIBCO) with 2 mM glutamine, 50 U/ml penicillin, 50 μg/ml streptomycin, 2.5 μg/ml Fungizone and 25-1,000 U/ml f IL-2 (Cetus). At the initial plating, 10 ng/ml OKT3 (Ortho) monoclonal antibody is added to each well. The cells are cultured at 37° C. in a humidified incubator with 5% CO.
Growth, Transduction and Selection of ADA-Deficient T Lymphocytes
Once the T lymphocytes have begun to proliferate (usually 24-96 hr after initial n at the culture) the cells are transduced by use of LASN vector (Blood, V L 72(2) pages 876-81) 2 ml. LASN vector-containing supernatant (contianing protamine 5-10 μg/ml and up to 1,000 U/ml IL-2) are added to the wells after aspirating off the top half of the medium. This is repeated 1-2 times daily for a period of up to 7 days. After the final exposure to retroviral vector the cells are fed with fresh AIM-V and cultured another 2-7 days to permit the cultures to return to exponential growth. Approximately 80% of the culture 0.1-2.5×1010 T cells are infused into the patient and the remaining cells cryopreserved for future use or returned to culture for studying growth and selection procedures, phenotype analysis, T cell repertoire analysis and percentage of cells demonstrating vector integration.
An aliquot of the cells infused into the patient is saved for subsequent Southern analysis on the DNA from the cultured cells after digestion with a restriction endonuclease which does not cut within the vector sequence to determine whether the gene-modified cells are polyclonal with respect to retroviral insertion.
Reinfusion of hADA Transduced T Lymphocytes
The transduced cells are harvested, washed, and resuspended in normal saline. The final cell preparation is filtered through a platelet filter and transferred into a syrinige or transfusion pack for infusion. A test dose of 5% of the total volume is infused by peripheral vein followed by an observation period of 5-10 min.
Transduced cells were administered to patient # 1 in the amounts and at the times shown in FIG. 4. The ADA enzyme levels of patient # 1 are shown in FIG. 5.
For purposes of completing this disclosure, the entire contents of the references cited herein are hereby incorporated by references and relied upon.
EXAMPLE 6 GENE-MODIFIED TUMOR CELL CLINICAL PROTOCOL
The TNF-NeoR vector was constructed as in Example 4, and retroviral vector supernatant is prepared and tested as disclosed therein.
A. Preparation of Gene-Modified Tumor Cells.
Tumor cell lines are established in tissue culture from tumor fragments or single cell suspensions using standard tissue culture techniques. (Topalian, et al., J. Immunol, Vol. 144, pgs. 4487-4495 (1990)). Tumor and normal tissue are obtained immediately after surgery.
The tumors were minced into 1 mm3 fragments and dissociated with agitation in serum free DMEM (Dulbecco Modified Eagle Medium)(Bio-fluids) containing 2 mM glutamine, 0.1 mg/ml hyaluronidase, 0.01 mg/ml DNase I an d0.1 mg/ml collagenase for 3 hours at room temperature. The cell suspension was then centrifuged at 800 g for 5 minutes and the pellet resuspended in a culture medium consisting of 5 ml of DMEM high glucose (4.5 g/l) with penicillin and glutamine supplemented with 10% fetal calf serum. The cells were either centrifuged prior to being frozen in 90% FCS, 10% DMSO at −80° C., or plated in appropriate dishes or culture flasks in culture medium. Plated cells were incubated at 37° C. in a humidified atmosphere of 5% CO2 and 95% air. Within 48 hrs, the culture medium was changed in order to remove all non-attached material. Subsequently, cultures were incubated for a period of 6 to 8 days without medium change. The tum rs grow as adherent monolayers in tissue culture flasks (Falcon #3028; 175 cm−2; 750 ml) containing about 50 ml of medium. When the cells are actively growing and not yet confluent the medium will be poured off and 30-50 ml of medium containing the retroviral supernatant with 5 μg/ml protamine will be added to the flask. Cornetta, et al., J. Virol. Meth., Vol. 23, Pgs. 186-194 (1989). The flasks will be incubated at 37° C. for six hours at which time the medium will be changed. This procedures will be repeated up to three times. After 24 to 48 hours medium containing 300 μg/ml G418 is added directly to the flask and the ells will be grown and subcultured for 7 to 14 days in G418 containing medium. The G418 concentrations may be raised to 1 mg/ml depending on the health of the culture.
B. Tests on the Transduced Tumor Population.
The following tests are performed on the tumor cells prior to injection into patients.
1) Call viability will be greater than 70% as tested by trypan dye exclusion.
2) Sterility will be assured by testing for aerobic and anaerobic bacteria, fungus and mycoplasma.
3) S+/L—assay must be negative.
4) Souther blot or PCR analysis will be run on the transduced tumor to assure that proviral sequences are present.
5) TNF protein assay to assure the production of TNF. Cells must be producing at least 100 pg TNF/106 cells/24 hours.
C. Injection of Tumor Cells.
Gene-modified tumor cells are harvested from the culture flasks by exposure to 0.25% versene (EDTA) for 10 minutes. The cells are washed three times by suspension in 50 mls normal saline and centrifugation. The final cell pellet will be suspended in normal saline and counted. 2×108 viable cells in 1 ml normal saline are injected subcutaneously just beneath the skin in the anterior mid thigh and the overlying skin marked with a tatoo dot. If 2×108 cells are not available, fewer may be given but not less than 2×107 cells will be injected. About 3 cm lateral or vertical to this injection the patient will receive two intradermal injections (separated by 1 cm) of 2×107 gene modified tumor cells in 0.1 ml normal saline and these sites also marked by a tatoo dot. These sites are monitored weekly by a physician. At three weeks the patient undergoes excisional biopsy of superficial inguinal lymph nodes (without formal dissection) in the area draining the inoculation site for growth of lymphocytes. If tumor grows at any of these sites they will be excised when they reach 1 to 2 cm for growth of TIL. If no tumor growth is evident that the sites of tumor injection will be excisionally biopsied at 8 weeks after injection of pathologic analysis.
D. Growth of Lymphocytes.
At least two days prior to surgery, peripheral blood lymphocytes are collected by leukapheresis for four hours, are processed, and then cultured as described in Example 4.
If, during the growth TIL, a patient's condition has deteriorated to an unacceptable level, or if the patient has developed significant cardiac, renal, pulmonary, or hematologic dysfunction, then the patient will not receive the infusion of TIL or of IL-2 as described hereinbelow.
When the total lymphocytes f r a patient are ready f r harvest, 5×106 cells are taken from cyt 1 gical examination. Cytospins are examined f r the presence f remaining tum r. At least 200 cells are studied and therapy proceeds only when no tumor cells are found. Other lymphocyte samples are taken f r characterization of cell surface markers and for assessment of cytotoxicity. (Berd, et al., Cancer Res., Vol. 46, pgs. 2572-2577 (1986)). Briefly, lymphocytes are stained with fluorescent-labeled antibodies (Leu2, Leu3, Lue4, Lye7, Lue11, Leu5, Leu9, LeuM3, HLADDR and Tac). Chromium release assays are performed with K562, Daudi autologous tumor, and allogeneic tumor targets.
To infuse the lymphocytes, they are thawed and grown for one to three additional weeks using the same procedures hereinabove described. For infusion TIL are reharvested. At the time of cell collection, one liter of saline for injection is pumped through the collection chamber and the centrifuge is stopped. Lymphocytes are resuspended in the collection bag, the centrifuge is started again, and another liter of saline is pumped through to wash fully the TIL's free of tissue culture medium components. The cells are then filtered through a platelet administration set into 600 ml transfer packs (Fenwal), and 50 ml of 25% albumin and 450,000 IU of IL-2 are added to the 200 to 300 ml volume of cells in saline. The TIL are infused over 30 to 60 minutes through a central venous catheter.
E. Administration of Interleukin-2.
The recombinant IL-2 used in this trial is provided by the Division of Cancer Treatment, National Cancer Institute (supplied by Cetus Corporation, Emeryville, Calif.) and will be administered exactly as specified in Rosenberg, et al., New Engl. J. Med., Vol. 323, pgs. 570-578 (1990). The IL-2 is provided as a lyophilized powder and will be reconstituted with 1.2 ml/vial. Each vial contains approximately 1.2 mg of IL-2 (specific activity 18×106IV/mg). Less than 0.04 ng of endotoxin are present per vial as measured by the limulus amebocyte assay. Each vial also contains 5% mannitol and approximately 130-230 μg of sodium dodecyl sulfate/mg of IL-2. Following reconstitution the IL-2 is diluted in 50 ml of normal saline containing 5% human serum albumin, and is infused intravenously at a dose of 720,000 IU/kg over a 15 minute period every 8 hr, beginning from two to 24 hr after the TIL infusion. IL-2 will be given for up to five consecutive days as tolerated. Under no circumstances is more than 15 doses of IL-2 be administered. Doses may be skipped depending on patient tolerance. Doses will be skipped if patients reach grade III or grade IV toxicity. If this toxicity is easily reversed by supportive measures then additional doses may be given.
Patients may receive concomitant medications to control side effects. For example, the patients may be given acetaminophen (650 mg every 4 hours), indomethacin (50-75 mg every 6 hours), and ranitidine (150 mg every 12 hours) throughout the course of the treatment. Patients may also receive intranveous meperidine (25-50 mg) to control chills if they occur. Hydroxyzine hydrochloride may be given (25 mg every 6 hours) to treat pruitia, if present.
EXAMPLE 7
In this example, the procedure of Example 6 were again followed, except that the vector employed to generate vector particles and retroviral vector supernatant, was the IL-2-NeoR vector. This vector was constructed from the vector LXSN (Miller, et al, 1989), and contains the Il-2 gene. Upon construction f the IL-2NeoR vector and the generation f vector particles and retroviral vector supernatants, that protocols for preparing gene-modified tumor cells and for treatment of patients with such cells, described in Example 6, were then followed.
Although the present invention has been described in particular with respect to genetically engineering primary human nucleated blood cells, and primary human tumor cells, it is to be understood that within the scope of the present invention, one may genetically engineer other human primary cells. Other primary human cells which may be genetically engineered in accordance with the present invention include, but are not limited to, endothelial cells, epithelial cells, keratinocytes, stem cells, hepatocytes, connective tissue cells, fibroblasts, mesenchymal cells, msothelial cells, and parenchymal cells.
While the invention has been described with respect to certain specific embodiment, it will be appreciated that many modifications and changes may be made by those skilled in the art without departing from the spirit of the invention. It is intended, therefore, by the appended claims to cover all such modification and changes as fall within the true spirit and scope of the invention.

Claims (20)

1. A process for providing a human with a therapeutic protein comprising:
introducing human cells int a human, said human cells having been treated in vitro to insert therein a DNA segment encoding a therapeutic protein said human cells expressing in vivo in said human a therapeutically effective amount of said therapeutic protein.
2. The process of claim 1 wherein said cells are blood cells.
3. The process of claim 1 wherein said cells are leukocytes.
4. The process of claim 1 wherein said cells are lymphocytes.
5. The process of claim 1 wherein said cells are T-lymphocytes.
6. The process of claim 1 wherein said cells are TIL cells.
7. The process of claim 1 wherein said cells are B-lymphocytes.
8. The process of any one of claims 1-7 wherein said DNA segment has been inserted into said cells in vitro by a viral vector.
9. The process of claim 8 wherein said viral vector is a retroviral vector.
10. The process of any one of claims 1-7 wherein the DNA segment encodes a cytokine.
11. The process of claim 10 wherein the cytokine is TNF.
12. The process of claim 10 wherein the cytokine is an interleukin.
13. The process of claim 10 wherein said DNA segment has been inserted into said cells in vitro by a viral vector.
14. The process f claim 13 wherein said viral vector is a retroviral vector.
15. A process for providing a human with a therapeutic protein comprising:
introducing autologous human cells into a human, said autologous human cells having been treated in vitro to insert therein a DNA segment encoding a therapeutic protein, said autologous human cells expressing in vivo in said human a therapeutically effective amount of said therapeutic protein, wherein said therapeutic protein is an interleukin, wherein the autologous human cells are T lymphocytes.
16. The process of claim 15, wherein said cells are tumor infiltrating lymphocytes (TIL).
17. The process of claim 15 or 16, wherein said DNA segment has been inserted into said cells in vitro by a viral vector.
18. The process of claim 17, wherein said viral vector is a retroviral vector.
19. The process of claim 15, wherein the interleukin is selected from the group consisting of IL- 1, IL- 2, IL- 3, IL- 4, IL- 5, IL- 6, IL- 7, IL- 8, IL- 9, IL- 10, IL- 11, and IL- 12.
20. The process of claim 19, wherein the interleukin is IL- 2.
US10/701,022 1989-06-14 2003-11-04 Gene therapy Expired - Lifetime USRE39788E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/701,022 USRE39788E1 (en) 1989-06-14 2003-11-04 Gene therapy

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US36556789A 1989-06-14 1989-06-14
US80744691A 1991-12-13 1991-12-13
US86879492A 1992-04-15 1992-04-15
US90466292A 1992-09-08 1992-09-08
US08/220,175 US5399346A (en) 1989-06-14 1994-03-30 Gene therapy
US10/701,022 USRE39788E1 (en) 1989-06-14 2003-11-04 Gene therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/220,175 Reissue US5399346A (en) 1989-06-14 1994-03-30 Gene therapy

Publications (1)

Publication Number Publication Date
USRE39788E1 true USRE39788E1 (en) 2007-08-21

Family

ID=27502997

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/220,175 Ceased US5399346A (en) 1989-06-14 1994-03-30 Gene therapy
US10/701,022 Expired - Lifetime USRE39788E1 (en) 1989-06-14 2003-11-04 Gene therapy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/220,175 Ceased US5399346A (en) 1989-06-14 1994-03-30 Gene therapy

Country Status (1)

Country Link
US (2) US5399346A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080131415A1 (en) * 2006-11-30 2008-06-05 Riddell Stanley R Adoptive transfer of cd8 + t cell clones derived from central memory cells
US20080175845A1 (en) * 2006-06-15 2008-07-24 Targeted Genetics Corporation Methods for treating target joints in inflammatory arthritis using AAV vectors encoding a TNF antagonist
US20090123441A1 (en) * 2007-10-08 2009-05-14 Intrexon Corporation Engineered Dendritic Cells and Uses for the Treatment of Cancer
US11111493B2 (en) 2018-03-15 2021-09-07 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy

Families Citing this family (953)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3681787D1 (en) * 1985-07-05 1991-11-07 Whitehead Biomedical Inst EXPRESSION OF FOREIGN GENETIC MATERIAL IN EPITHELIC CELLS.
US6150328A (en) * 1986-07-01 2000-11-21 Genetics Institute, Inc. BMP products
US6048729A (en) * 1987-05-01 2000-04-11 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US6093699A (en) * 1987-07-09 2000-07-25 The University Of Manitoba Method for gene therapy involving suppression of an immune response
EP0633318A1 (en) * 1987-09-11 1995-01-11 Whitehead Institute For Biomedical Research Transduced fibroblasts and uses therefor
US5674722A (en) * 1987-12-11 1997-10-07 Somatix Therapy Corporation Genetic modification of endothelial cells
US6544771B1 (en) 1987-12-11 2003-04-08 Cell Genesys, Inc. Retroviral gene therapy vectors and therapeutic methods based thereon
US6001350A (en) * 1987-12-11 1999-12-14 Somatix Therapy Corp Genetic modification of endothelial cells
US6140111A (en) * 1987-12-11 2000-10-31 Whitehead Institute For Biomedical Research Retroviral gene therapy vectors and therapeutic methods based thereon
US5580776A (en) * 1988-02-05 1996-12-03 Howard Hughes Medical Institute Modified hepatocytes and uses therefor
EP0732397A3 (en) 1988-02-05 1996-10-23 Whitehead Institute For Biomedical Research Modified hepatocytes and uses therefor
US5716826A (en) * 1988-03-21 1998-02-10 Chiron Viagene, Inc. Recombinant retroviruses
US5662896A (en) 1988-03-21 1997-09-02 Chiron Viagene, Inc. Compositions and methods for cancer immunotherapy
US6569679B1 (en) 1988-03-21 2003-05-27 Chiron Corporation Producer cell that generates adenoviral vectors encoding a cytokine and a conditionally lethal gene
US5997859A (en) * 1988-03-21 1999-12-07 Chiron Corporation Method for treating a metastatic carcinoma using a conditionally lethal gene
US6133029A (en) * 1988-03-21 2000-10-17 Chiron Corporation Replication defective viral vectors for infecting human cells
US5650148A (en) * 1988-12-15 1997-07-22 The Regents Of The University Of California Method of grafting genetically modified cells to treat defects, disease or damage of the central nervous system
ATE219519T1 (en) * 1989-01-23 2002-07-15 Chiron Corp RECOMBINANT THERAPIES FOR INFECTIONS AND HYPERPROLIFERATIVE DISORDERS
EP0497922B1 (en) * 1989-10-24 2002-01-30 Chiron Corporation Infective protein delivery system
US6027722A (en) * 1990-10-25 2000-02-22 Nature Technology Corporation Vectors for gene transfer
US5747469A (en) * 1991-03-06 1998-05-05 Board Of Regents, The University Of Texas System Methods and compositions comprising DNA damaging agents and p53
WO1993002556A1 (en) * 1991-07-26 1993-02-18 University Of Rochester Cancer therapy utilizing malignant cells
US5529774A (en) 1991-08-13 1996-06-25 The Regents Of The University Of California In vivo transfer of the HSV-TK gene implanted retroviral producer cells
US20080070842A1 (en) * 1991-11-04 2008-03-20 David Israel Recombinant bone morphogenetic protein heterodimers, compositions and methods of use
KR100259827B1 (en) * 1991-11-04 2000-06-15 브루스 엠. 에이센, 토마스 제이 데스로저 Recombinant bone morphogenetic protein heterodimers
NZ245015A (en) * 1991-11-05 1995-12-21 Transkaryotic Therapies Inc Delivery of human growth hormone through the administration of transfected cell lines encoding human growth hormone, which are physically protected from host immune response; the transfected cells and their production
US6692737B1 (en) 1991-11-05 2004-02-17 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
US6063630A (en) 1991-11-05 2000-05-16 Transkaryotic Therapies, Inc. Targeted introduction of DNA into primary or secondary cells and their use for gene therapy
US6054288A (en) * 1991-11-05 2000-04-25 Transkaryotic Therapies, Inc. In vivo protein production and delivery system for gene therapy
WO1993010218A1 (en) * 1991-11-14 1993-05-27 The United States Government As Represented By The Secretary Of The Department Of Health And Human Services Vectors including foreign genes and negative selective markers
WO1993017715A1 (en) * 1992-03-05 1993-09-16 Board Of Regents, The University Of Texas System Diagnostic and/or therapeutic agents, targeted to neovascular endothelial cells
US5660827A (en) * 1992-03-05 1997-08-26 Board Of Regents, The University Of Texas System Antibodies that bind to endoglin
US6749853B1 (en) 1992-03-05 2004-06-15 Board Of Regents, The University Of Texas System Combined methods and compositions for coagulation and tumor treatment
US6093399A (en) * 1992-03-05 2000-07-25 Board Of Regents, The University Of Texas System Methods and compositions for the specific coagulation of vasculature
US6036955A (en) * 1992-03-05 2000-03-14 The Scripps Research Institute Kits and methods for the specific coagulation of vasculature
US6004555A (en) * 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for the specific coagulation of vasculature
US5965132A (en) * 1992-03-05 1999-10-12 Board Of Regents, The University Of Texas System Methods and compositions for targeting the vasculature of solid tumors
US6087323A (en) * 1992-04-03 2000-07-11 Cambridge Neuroscience, Inc. Use of neuregulins as modulators of cellular communication
US6531124B1 (en) 1992-07-10 2003-03-11 Transkaryotic Therapies, Inc. In vivo production and delivery of insulinotropin for gene therapy
US6670178B1 (en) 1992-07-10 2003-12-30 Transkaryotic Therapies, Inc. In Vivo production and delivery of insulinotropin for gene therapy
US5747323A (en) * 1992-12-31 1998-05-05 Institut National De La Sante Et De La Recherche Medicale (Inserm) Retroviral vectors comprising a VL30-derived psi region
ES2210249T3 (en) * 1993-02-17 2004-07-01 Sloan-Kettering Institute For Cancer Research ALOGENIC VACCINE AND METHOD FOR SYNTHESIS.
US5654186A (en) * 1993-02-26 1997-08-05 The Picower Institute For Medical Research Blood-borne mesenchymal cells
WO1994020608A1 (en) 1993-03-12 1994-09-15 Creighton University Improved vectors for gene therapy
US7393682B1 (en) 1993-03-19 2008-07-01 The Johns Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
US6340674B1 (en) 1993-03-26 2002-01-22 Thomas Jefferson University Method of inhibiting the proliferation and causing the differentiation of cells with IGF-1 receptor antisense oligonucleotides
US5645829A (en) * 1993-06-18 1997-07-08 Beth Israel Hospital Association Mesothelial cell gene therapy
US5830463A (en) * 1993-07-07 1998-11-03 University Technology Corporation Yeast-based delivery vehicles
AU7404994A (en) * 1993-07-30 1995-02-28 Regents Of The University Of California, The Endocardial infusion catheter
AU7477394A (en) * 1993-07-30 1995-03-27 University Of Medicine And Dentistry Of New Jersey Efficient gene transfer into primary lymphocytes
US6652850B1 (en) 1993-09-13 2003-11-25 Aventis Pharmaceuticals Inc. Adeno-associated viral liposomes and their use in transfecting dendritic cells to stimulate specific immunity
US6291206B1 (en) * 1993-09-17 2001-09-18 Genetics Institute, Inc. BMP receptor proteins
EP0729511A1 (en) 1993-11-18 1996-09-04 Chiron Corporation Compositions and methods for utilizing conditionally lethal genes
US5591625A (en) * 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
US6190655B1 (en) * 1993-12-03 2001-02-20 Immunex Corporation Methods of using Flt-3 ligand for exogenous gene transfer
CA2176942C (en) * 1993-12-07 2011-11-01 Anthony J. Celeste Bmp-12, bmp-13 and tendon-inducing compositions thereof
AU1513495A (en) * 1993-12-14 1995-07-03 University Of Pittsburgh Systemic gene treatment of connective tissue diseases
US20040161416A1 (en) * 1993-12-14 2004-08-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Systemic gene treatment of connective tissue diseases
WO1995019369A1 (en) * 1994-01-14 1995-07-20 Vanderbilt University Method for detection and treatment of breast cancer
US6451578B1 (en) 1994-02-14 2002-09-17 Abbott Laboratories Non-A, non-B, non-C, non-D, non-E hepatitis reagents and methods for their use
US5843450A (en) * 1994-02-14 1998-12-01 Abbott Laboratories Hepatitis GB Virus synthetic peptides and uses thereof
US6720166B2 (en) 1994-02-14 2004-04-13 Abbott Laboratories Non-a, non-b, non-c, non-c, non-d, non-e hepatitis reagents and methods for their use
US6558898B1 (en) 1994-02-14 2003-05-06 Abbott Laboratories Non-A, non-B, non-C, non-D, non-E hepatitis reagents and methods for their use
US6586568B1 (en) 1994-02-14 2003-07-01 Abbott Laboratories Non-A, non-B, non-C, non-D, non-E hepatitis reagents and methods for their use
US6156495A (en) * 1994-02-14 2000-12-05 Abbott Laboratories Hepatitis GB virus recombinant proteins and uses thereof
US5981172A (en) * 1994-02-14 1999-11-09 Abbott Laboratories Non-A, non-B, non-C, non-D, non-E Hepatitis reagents and methods for their use
US6051374A (en) * 1994-02-14 2000-04-18 Abbott Laboratories Non-A, non-B, non-C, non-D, non-E hepatitis reagents and methods for their use
GB9402857D0 (en) 1994-02-15 1994-04-06 Isis Innovation Targeting gene therapy
US7294331B2 (en) * 1994-03-07 2007-11-13 Immunex Corporation Methods of using flt3-ligand in hematopoietic cell transplantation
US6984379B1 (en) * 1994-04-08 2006-01-10 Children's Hospital of LosAngeles Gene therapy by administration of genetically engineered CD34+ cells obtained from cord blood
IL109558A (en) * 1994-05-04 2004-08-31 Yissum Res Dev Co Sv40-derived dna constructs comprising exogenous dna sequences
US5714353A (en) * 1994-05-24 1998-02-03 Research Corporation Technologies, Inc. Safe vectors for gene therapy
US5888814A (en) * 1994-06-06 1999-03-30 Chiron Corporation Recombinant host cells encoding TNF proteins
US5891633A (en) * 1994-06-16 1999-04-06 The United States Of America As Represented By The Department Of Health And Human Services Defects in drug metabolism
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US20030026782A1 (en) * 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US6350730B1 (en) 1994-08-17 2002-02-26 The Rockefeller University OB polypeptides and modified forms as modulators of body weight
US6124448A (en) * 1994-08-17 2000-09-26 The Rockfeller University Nucleic acid primers and probes for the mammalian OB gene
US6471956B1 (en) 1994-08-17 2002-10-29 The Rockefeller University Ob polypeptides, modified forms and compositions thereto
US6048837A (en) * 1994-08-17 2000-04-11 The Rockefeller University OB polypeptides as modulators of body weight
US6001968A (en) * 1994-08-17 1999-12-14 The Rockefeller University OB polypeptides, modified forms and compositions
US6124439A (en) * 1994-08-17 2000-09-26 The Rockefeller University OB polypeptide antibodies and method of making
US5827642A (en) * 1994-08-31 1998-10-27 Fred Hutchinson Cancer Research Center Rapid expansion method ("REM") for in vitro propagation of T lymphocytes
US6911216B1 (en) 1994-10-12 2005-06-28 Genzyme Corporation Targeted delivery via biodegradable polymers
US5714170A (en) 1994-11-16 1998-02-03 Thomas Jefferson University Method of inducing resistance to tumor growth
US5980886A (en) * 1994-12-14 1999-11-09 University Of Washington Recombinant vectors for reconstitution of liver
US6107027A (en) * 1994-12-14 2000-08-22 University Of Washington Ribozymes for treating hepatitis C
US5843742A (en) * 1994-12-16 1998-12-01 Avigen Incorporated Adeno-associated derived vector systems for gene delivery and integration into target cells
EP0801526A4 (en) * 1994-12-30 1999-12-01 Univ Jefferson Gene transduction system
US5766899A (en) * 1995-02-27 1998-06-16 Board Of Regents , The University Of Texas System Targeted nucleic acid delivery into liver cells
US5681744A (en) * 1995-03-17 1997-10-28 Greenstein; Robert J. Delivery and expression of heterologus genes using upstream enhancer regions of mammalian gene promoters
US6531455B1 (en) 1995-03-24 2003-03-11 The Regents Of The University Of California Delivery of polynucleotides by secretory gland expression
US5837693A (en) * 1995-03-24 1998-11-17 The Regents Of The University Of California Intravenous hormone polypeptide delivery by salivary gland expression
US5885971A (en) * 1995-03-24 1999-03-23 The Regents Of The University Of California Gene therapy by secretory gland expression
US5830755A (en) * 1995-03-27 1998-11-03 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services T-cell receptors and their use in therapeutic and diagnostic methods
WO1996034109A1 (en) * 1995-04-25 1996-10-31 Vical Incorporated Single-vial formulations of dna/lipid complexes
US7069634B1 (en) 1995-04-28 2006-07-04 Medtronic, Inc. Method for manufacturing a catheter
DE69634563T2 (en) * 1995-05-04 2006-02-16 The United States Of America As Representend By The Secretary Of The Navy IMPROVED METHODS FOR TRANSFECTION OF T CELLS
US6692964B1 (en) * 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US7067318B2 (en) * 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US5688915A (en) * 1995-06-01 1997-11-18 The University Of Medicine And Dentistry Of New Jersey Long term maintenance of lymphocytes in vitro
US5993800A (en) * 1995-06-05 1999-11-30 Bristol-Myers Squibb Company Methods for prolonging the expression of a heterologous gene of interest using soluble CTLA4 molecules and an antiCD40 ligand
US5759776A (en) * 1995-06-05 1998-06-02 California Pacific Medical Center Targets for breast cancer diagnosis and treatment
WO1998008090A1 (en) * 1995-06-06 1998-02-26 Epigen, Inc. Anti-idiotypic antibodies to an epiglycanin
EP0830368A1 (en) 1995-06-07 1998-03-25 Genta Incorporated Novel carbamate-based cationic lipids
US6511811B1 (en) 1995-06-07 2003-01-28 The Regents Of The University Of California Protein kinase C antagonist related to insulin receptor
US5869040A (en) * 1995-06-07 1999-02-09 Biogen, Inc Gene therapy methods and compositions
US5629159A (en) * 1995-06-07 1997-05-13 California Institute Of Technology Immortalization and disimmortalization of cells
US5990388A (en) * 1995-06-07 1999-11-23 Research Corporation Technologies, Inc. Resistance to viruses and viroids in transgenic plants and animals expressing dsRNA-binding protein
WO1996040212A1 (en) * 1995-06-07 1996-12-19 Sloan-Kettering Institute For Cancer Research Retrovirus vectors for expression of cii-ta and activation of hla class ii gene expression and uses thereof
CN1203632A (en) 1995-07-17 1998-12-30 德克萨斯州立大学董事会 P16 expression constructs and their application in cancer therapy
US7163925B1 (en) 1995-07-17 2007-01-16 Board Of Regents, The University Of Texas System p16 expression constructs and their application in cancer therapy
US20020182730A1 (en) * 1995-07-26 2002-12-05 Micheal L. Gruenberg Autologous immune cell therapy: cell compositions, methods and applications to treatment of human disease
AU6410596A (en) * 1995-07-31 1997-02-26 Centre De Recherche De L'hopital Sainte-Justine Cytidine deaminase cdna as a positive selectable marker for ene transfer, gene therapy and protein synthesis
US5807670A (en) * 1995-08-14 1998-09-15 Abbott Laboratories Detection of hepatitis GB virus genotypes
US5955318A (en) * 1995-08-14 1999-09-21 Abbott Laboratories Reagents and methods useful for controlling the translation of hepatitis GBV proteins
US5770720A (en) * 1995-08-30 1998-06-23 Barnes-Jewish Hospital Ubiquitin conjugating enzymes having transcriptional repressor activity
US6482803B1 (en) 1995-09-01 2002-11-19 Board Of Regents, The University Of Texas System Modification of mutated P53 gene in tumors by retroviral delivery of ribozyme A
GB9519299D0 (en) * 1995-09-21 1995-11-22 Farrar Gwyneth J Genetic strategy
US5922583A (en) * 1995-10-17 1999-07-13 Biostar Inc. Methods for production of recombinant plasmids
US20030190753A1 (en) * 1995-11-09 2003-10-09 Nature Technology Corporation Vectors for gene transfer
EA003195B1 (en) 1995-11-13 2003-02-27 Такара Сузо Ко., Лтд. Method for gene transfer into target cells with retrovirus
US5986170A (en) * 1995-11-13 1999-11-16 Corixa Corporation Murine model for human carcinoma
JP2000500654A (en) * 1995-11-14 2000-01-25 トーマス・ジェファーソン・ユニバーシティ Induced resistance to tumor growth by soluble IGF-1 receptor
US5888767A (en) * 1996-11-27 1999-03-30 The Johns Hopkins University School Of Medicine Method of using a conditionally replicating viral vector to express a gene
US5753490A (en) * 1995-11-28 1998-05-19 Clinical Technologies, Inc. Recombinant HIV and modified packaging cells and method for treating acquired immune deficiency syndrome
US6063374A (en) * 1995-11-28 2000-05-16 Clinical Technologies, Inc. Recombinant HIV and modified packaging cells and method for using
US6033674A (en) 1995-12-28 2000-03-07 Johns Hopkins University School Of Medicine Method of treating cancer with a tumor cell line having modified cytokine expression
US6991787B1 (en) * 1995-12-29 2006-01-31 Alg Company Methods of preparing bone marrow stromal cells for use in gene therapy
AU1754497A (en) * 1996-01-16 1997-08-11 Board Of Trustees Of The Leland Stanford Junior University Compositions and their uses for transfer of down-regulatory genes into cells associated with inflammatory responses
US6087129A (en) 1996-01-19 2000-07-11 Betagene, Inc. Recombinant expression of proteins from secretory cell lines
US6110707A (en) * 1996-01-19 2000-08-29 Board Of Regents, The University Of Texas System Recombinant expression of proteins from secretory cell lines
US6051218A (en) * 1996-02-02 2000-04-18 The Regents Of The University Of California Tumor radiosensitization using gene therapy
JP2001503963A (en) * 1996-02-06 2001-03-27 イーライ・リリー・アンド・カンパニー Diabetes treatment
US5891857A (en) * 1996-02-20 1999-04-06 Vanderbilt University Characterized BRCA1 and BRCA2 proteins and screening and therapeutic methods based on characterized BRCA1 and BRCA2 proteins
US5789201A (en) * 1996-02-23 1998-08-04 Cocensys, Inc. Genes coding for bcl-y a bcl-2 homologue
US20030069195A1 (en) * 1996-03-01 2003-04-10 Farrar Gwenyth Jane Suppression of polymorphic alleles
US6316257B1 (en) 1996-03-04 2001-11-13 Targeted Genetics Corporation Modified rapid expansion methods (“modified-REM”) for in vitro propagation of T lymphocytes
GB9606961D0 (en) 1996-04-02 1996-06-05 Farrar Gwyneth J Genetic strategy III
US8551970B2 (en) * 1996-04-02 2013-10-08 Optigen Patents Limited Genetic suppression and replacement
US7026116B1 (en) 1996-04-04 2006-04-11 Bio-Rad Laboratories, Inc. Polymorphisms in the region of the human hemochromatosis gene
US6140305A (en) * 1996-04-04 2000-10-31 Bio-Rad Laboratories, Inc. Hereditary hemochromatosis gene products
US6723531B2 (en) * 1996-04-05 2004-04-20 The Salk Institute For Biological Studies Method for modulating expression of exogenous genes in mammalian systems, and products related thereto
US5776683A (en) * 1996-07-11 1998-07-07 California Pacific Medical Center Methods for identifying genes amplified in cancer cells
AU2557097A (en) 1996-04-17 1997-11-07 Board Of Regents, The University Of Texas System Enhanced expression of transgenes
US5846220A (en) 1996-04-30 1998-12-08 Medtronic, Inc. Therapeutic method for treatment of Alzheimer's disease
US7189222B2 (en) 1996-04-30 2007-03-13 Medtronic, Inc. Therapeutic method of treatment of alzheimer's disease
WO1997044356A2 (en) 1996-05-08 1997-11-27 Biogen, Inc. RET LIGAND (RetL) FOR STIMULATING NEURAL AND RENAL GROWTH
US6027721A (en) * 1996-05-20 2000-02-22 Cytotherapeutics, Inc. Device and method for encapsulated gene therapy
US6849399B1 (en) 1996-05-23 2005-02-01 Bio-Rad Laboratories, Inc. Methods and compositions for diagnosis and treatment of iron misregulation diseases
US8323963B2 (en) * 1996-05-29 2012-12-04 University Of Southern California Construction and use of genes encoding pathogenic epitopes for treatment of autoimmune disease
US6274136B1 (en) * 1996-05-29 2001-08-14 University Of Southern California Construction and use of genes encoding pathogenic epitopes for treatment of autoimmune disease
US6015828A (en) * 1996-05-31 2000-01-18 Cuppoletti; John Chemical modification of chloride channels as a treatment for cystic fibrosis and other diseases
US6132989A (en) * 1996-06-03 2000-10-17 University Of Washington Methods and compositions for enhanced stability of non-adenoviral DNA
US20030198626A1 (en) * 2002-04-22 2003-10-23 Antigen Express, Inc. Inhibition of Ii expression in mammalian cells
US20060008448A1 (en) * 1996-06-11 2006-01-12 Minzhen Xu Inhibition of li expression in mammalian cells
US5869037A (en) * 1996-06-26 1999-02-09 Cornell Research Foundation, Inc. Adenoviral-mediated gene transfer to adipocytes
US6093816A (en) 1996-06-27 2000-07-25 Isis Pharmaceuticals, Inc. Cationic lipids
WO1998000544A2 (en) * 1996-06-28 1998-01-08 Dendreon Corporation Growth arrest gene compositions and methods
US5951975A (en) * 1996-06-28 1999-09-14 University Of Pittsburgh Induction of CTLs specific for natural antigens by cross priming immunization
US6277368B1 (en) 1996-07-25 2001-08-21 The Regents Of The University Of California Cancer immunotherapy using autologous tumor cells combined with cells expressing a membrane cytokine
US6057102A (en) * 1996-08-08 2000-05-02 The Aaron Diamond Aids Research Center HIV coreceptor mutants
EP0929318B1 (en) 1996-08-16 2004-11-24 The Johns Hopkins University School Of Medicine Melanoma cell lines expressing shared immunodominant melanoma antigens and methods of using same
US6544523B1 (en) 1996-11-13 2003-04-08 Chiron Corporation Mutant forms of Fas ligand and uses thereof
US5980898A (en) 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization
US20060002949A1 (en) * 1996-11-14 2006-01-05 Army Govt. Of The Usa, As Rep. By Secretary Of The Office Of The Command Judge Advocate, Hq Usamrmc. Transcutaneous immunization without heterologous adjuvant
US20060002959A1 (en) * 1996-11-14 2006-01-05 Government Of The United States Skin-sctive adjuvants for transcutaneous immuization
US6797276B1 (en) 1996-11-14 2004-09-28 The United States Of America As Represented By The Secretary Of The Army Use of penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response
US6087174A (en) * 1996-12-26 2000-07-11 Johns Hopkins University, School Of Medicine Growth medium for primary pancreatic tumor cell culture
EP1961820B1 (en) 1997-01-27 2012-11-28 Ludwig Institute for Cancer Research Ltd Vaccine composition comprising LAGE-1 tumor associated nucleic acids or LAGE-1 polypeptides
CA2196496A1 (en) 1997-01-31 1998-07-31 Stephen William Watson Michnick Protein fragment complementation assay for the detection of protein-protein interactions
WO1998038326A1 (en) 1997-02-28 1998-09-03 Nature Technology Corporation Self-assembling genes, vectors and uses thereof
US5969102A (en) 1997-03-03 1999-10-19 St. Jude Children's Research Hospital Lymphocyte surface receptor that binds CAML, nucleic acids encoding the same and methods of use thereof
US6463933B1 (en) 1997-03-25 2002-10-15 Morris Laster Bone marrow as a site for transplantation
US5866412A (en) * 1997-03-27 1999-02-02 Millennium Pharmaceuticals, Inc. Chromosome 18 marker
US5914394A (en) * 1997-03-27 1999-06-22 Millenium Pharmaceuticals, Inc. Methods and compositions for the diagnosis and treatment of neuropsychiatric disorders
US5939316A (en) * 1997-03-27 1999-08-17 Millennium Pharmaceuticals, Inc. Chromosome 18 marker
US5955355A (en) * 1997-03-27 1999-09-21 Millennium Pharmaceuticals, Inc. Chromosome 18 marker
US6218597B1 (en) 1997-04-03 2001-04-17 University Technology Corporation Transgenic model and treatment for heart disease
DE69838061T2 (en) * 1997-04-18 2008-03-13 Biogen Idec Ma Inc., Cambridge TYPE II TGF-BETA RECEPTOR / IMMUNOGLOBULIN CONSTANT DOMAIN FUSION PROTEINS
EP0979290A2 (en) * 1997-04-28 2000-02-16 Aventis Pharma S.A. Adenovirus-mediated intratumoral delivery of an angiogenesis antagonist for the treatment of tumors
US6004798A (en) * 1997-05-14 1999-12-21 University Of Southern California Retroviral envelopes having modified hypervariable polyproline regions
CA2205076A1 (en) 1997-05-14 1998-11-14 Jim Hu Episomal expression cassettes for gene therapy
US6541036B1 (en) 1997-05-29 2003-04-01 Thomas Jefferson University Treatment of tumors with oligonucleotides directed to insulin-like growth factor-I receptors (IGF-IR)
US6391852B1 (en) 1997-06-13 2002-05-21 Bio-Rad Laboratories, Inc. Methods and compositions for diagnosis and treatment of iron overload diseases and iron deficiency diseases
US6300488B1 (en) 1997-07-10 2001-10-09 The Salk Institute For Biological Studies Modified lepidopteran receptors and hybrid multifunctional proteins for use in transcription and regulation of transgene expression
AU8505898A (en) * 1997-07-22 1999-02-16 Genitrix, Llc Nucleic acid compositions and methods of introducing nucleic acids into cells
US6891082B2 (en) 1997-08-01 2005-05-10 The Johns Hopkins University School Of Medicine Transgenic non-human animals expressing a truncated activintype II receptor
US6331388B1 (en) 1997-10-17 2001-12-18 Wisconsin Alumni Research Foundation Immune response enhancer
US6875606B1 (en) * 1997-10-23 2005-04-05 The United States Of America As Represented By The Department Of Veterans Affairs Human α-7 nicotinic receptor promoter
US20040258703A1 (en) * 1997-11-14 2004-12-23 The Government Of The Us, As Represented By The Secretary Of The Army Skin-active adjuvants for transcutaneous immunization
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US6177410B1 (en) 1997-12-05 2001-01-23 Vanderbilt University Therapeutic methods for prostate cancer
ES2221717T3 (en) * 1997-12-08 2005-01-01 Emd Lexigen Research Center Corp. USEFUL HETERODIMERAS FUSION PROTEINS FOR DIRECTED IMMUNOTHERAPY AND GENERAL IMMUNOSTIMULATION.
US6331412B1 (en) * 1998-01-29 2001-12-18 University Of Ottawa Methods and compounds for modulating male fertility
US20030105294A1 (en) * 1998-02-25 2003-06-05 Stephen Gillies Enhancing the circulating half life of antibody-based fusion proteins
AU760794B2 (en) 1998-03-10 2003-05-22 Regents Of The University Of California, The Methods and tools for identifying compounds which modulate atherosclerosis by impacting LDL-proteoglycan binding
CA2245224A1 (en) * 1998-08-14 2000-02-14 Jiang-Hong Giong Chemokine receptor antagonists and chemotherapeutics
CA2305787A1 (en) * 2000-05-09 2001-11-09 The University Of British Columbia Cxcr4 antagonist treatment of hematopoietic cells
DE69914463T2 (en) * 1998-03-13 2004-11-11 The University Of British Columbia, Vancouver THERAPEUTIC CHEMOKINE RECEPTOR ANTAGONISTS
AUPP249298A0 (en) * 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
US7157083B2 (en) * 1998-04-17 2007-01-02 Surrogate Pharmaceutical Pathways, Llc Compositions and methods for treating retroviral infections
US6461606B1 (en) * 1998-04-24 2002-10-08 University Of Florida Research Foundation Materials and methods for gene therapy
US6225456B1 (en) 1998-05-07 2001-05-01 University Technololy Corporation Ras suppressor SUR-5
US6548290B1 (en) 1998-05-13 2003-04-15 President And Fellows Of Harvard College Geminin gene and protein
US6333318B1 (en) 1998-05-14 2001-12-25 The Salk Institute For Biological Studies Formulations useful for modulating expression of exogenous genes in mammalian systems, and products related thereto
US6506889B1 (en) 1998-05-19 2003-01-14 University Technology Corporation Ras suppressor SUR-8 and related compositions and methods
ES2324540T3 (en) 1998-05-27 2009-08-10 Genzyme Corporation AAV VECTORS FOR THE MANUFACTURE OF MEDICINES FOR THE ADMINISTRATION POTENTIATED BY CONVECTION.
US6197947B1 (en) 1998-06-01 2001-03-06 The Rockefeller University Translation initiation factor 4AIII and methods of use thereof
US5973119A (en) 1998-06-05 1999-10-26 Amgen Inc. Cyclin E genes and proteins
US20040247662A1 (en) * 1998-06-25 2004-12-09 Dow Steven W. Systemic immune activation method using nucleic acid-lipid complexes
US20030022854A1 (en) 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
US6693086B1 (en) * 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes
AU764686B2 (en) 1998-08-28 2003-08-28 Duke University Adenoviruses deleted in the IVa2, 100K, polymerase and/or preterminal protein sequences
DK1115862T3 (en) 1998-09-23 2009-11-16 Zymogenetics Inc Cytokine receptor Zalpha11
US6344541B1 (en) * 1998-09-25 2002-02-05 Amgen Inc. DKR polypeptides
US6135976A (en) 1998-09-25 2000-10-24 Ekos Corporation Method, device and kit for performing gene therapy
US6221349B1 (en) 1998-10-20 2001-04-24 Avigen, Inc. Adeno-associated vectors for expression of factor VIII by target cells
US6200560B1 (en) * 1998-10-20 2001-03-13 Avigen, Inc. Adeno-associated virus vectors for expression of factor VIII by target cells
US6468793B1 (en) 1998-10-23 2002-10-22 Florida State University Research Foundation CFTR genes and proteins for cystic fibrosis gene therapy
AU1331100A (en) 1998-10-29 2000-05-22 Dana-Farber Cancer Institute Cancer immunotheraphy and diagnosis using universal tumor associated antigens, including htert
US7399751B2 (en) * 1999-11-04 2008-07-15 Sertoli Technologies, Inc. Production of a biological factor and creation of an immunologically privileged environment using genetically altered Sertoli cells
US20030148929A1 (en) * 1998-11-18 2003-08-07 Hisamitsu Pharmaceutical Co., Inc. Nucleic acid carriers and pharmaceutical compositions for gene therapy
US6773911B1 (en) * 1998-11-23 2004-08-10 Amgen Canada Inc. Apoptosis-inducing factor
WO2000034474A2 (en) 1998-12-07 2000-06-15 Zymogenetics, Inc. Growth factor homolog zvegf3
US6441156B1 (en) 1998-12-30 2002-08-27 The United States Of America As Represented By The Department Of Health And Human Services Calcium channel compositions and methods of use thereof
WO2000040614A2 (en) 1998-12-30 2000-07-13 Beth Israel Deaconess Medical Center, Inc. Characterization of the soc/crac calcium channel protein family
US7063959B1 (en) * 1998-12-30 2006-06-20 Beth Israel Deaconess Medical Center, Inc. Compositions of the SOC/CRAC calcium channel protein family
JP2002533124A (en) 1998-12-31 2002-10-08 カイロン コーポレイション Improved expression of HIV polypeptide and generation of virus-like particles
US7935805B1 (en) 1998-12-31 2011-05-03 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV Type C polypeptides, polypeptides and uses thereof
US6727224B1 (en) * 1999-02-01 2004-04-27 Genetics Institute, Llc. Methods and compositions for healing and repair of articular cartilage
AU773954B2 (en) 1999-02-03 2004-06-10 Amgen, Inc. Novel Polypeptides involved in immune response
AU779387B2 (en) * 1999-02-09 2005-01-20 Lexicon Pharmaceuticals, Inc. Human uncoupling proteins and polynucleotides encoding the same
US6495376B1 (en) 1999-02-18 2002-12-17 Beth Israel Deaconess Medical Center Methods and compositions for regulating protein-protein interactions
AU3731400A (en) 1999-03-05 2000-09-21 Trustees Of University Technology Corporation, The Methods and compositions useful in inhibiting apoptosis
US6849605B1 (en) * 1999-03-05 2005-02-01 The Trustees Of University Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of viral infections
WO2000051623A2 (en) 1999-03-05 2000-09-08 The Trustees Of University Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of nitric oxide-induced clinical conditions
WO2000051625A1 (en) * 1999-03-05 2000-09-08 The Trustees Of University Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of herpes viruses
ATE377076T1 (en) 1999-03-09 2007-11-15 Zymogenetics Inc HUMAN CYTOKINE AS A LIGAND OF THE ZALPHA RECEPTOR AND ITS USES
AU3770800A (en) 1999-03-26 2000-10-16 Amgen, Inc. Beta secretase genes and polypeptides
EP2368575B1 (en) * 1999-04-08 2014-10-01 Intercell USA, Inc. Dry formulation for transcutaneous immunization
US6451319B1 (en) * 1999-04-09 2002-09-17 Schering-Plough Veterinary Corporation Recombinant and mutant adenoviruses
US6299882B1 (en) 1999-04-09 2001-10-09 Schering Corporation UL54.5 of Marek's disease virus (MDV)
US6328762B1 (en) * 1999-04-27 2001-12-11 Sulzer Biologics, Inc. Prosthetic grafts
US7270969B2 (en) 1999-05-05 2007-09-18 Phylogica Limited Methods of constructing and screening diverse expression libraries
WO2000069913A1 (en) * 1999-05-19 2000-11-23 Lexigen Pharmaceuticals Corp. EXPRESSION AND EXPORT OF INTERFERON-ALPHA PROTEINS AS Fc FUSION PROTEINS
US7166573B1 (en) * 1999-05-28 2007-01-23 Ludwig Institute For Cancer Research Breast, gastric and prostate cancer associated antigens and uses therefor
EP1939300A1 (en) 1999-05-28 2008-07-02 Targeted Genetics Corporation Methods and compositions for lowering the level of tumor necrosis factor (TNF) in TNF-associated disorders
DE60040147D1 (en) * 1999-05-28 2008-10-16 Targeted Genetics Corp METHODS AND COMPOSITIONS FOR REDUCING TUMOR NEKROSIS FACTOR MIRROR (TNF) IN TNF ASSOCIATED DISEASES
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
US6346382B1 (en) 1999-06-01 2002-02-12 Vanderbilt University Human carbamyl phosphate synthetase I polymorphism and diagnostic methods related thereto
US9486429B2 (en) 1999-06-01 2016-11-08 Vanderbilt University Therapeutic methods employing nitric oxide precursors
US6696272B1 (en) 1999-06-02 2004-02-24 Hsc Research & Development Limited Partnership Products and methods for gaucher disease therapy
ES2364086T3 (en) 1999-07-07 2011-08-24 Zymogenetics, Inc. HUMAN CYTOKIN RECEPTOR.
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
CN1235911C (en) * 1999-08-09 2006-01-11 利思进药品公司 Multiple cytokine-antibody complexes
EP1916258B1 (en) 1999-08-09 2014-04-23 Targeted Genetics Corporation Enhancement of expression of a single-stranded, heterologous nucleotide sequence from recombinant viral vectors by designing the sequence such that it forms intrastrand base pairs
DE60039766D1 (en) * 1999-08-09 2008-09-18 Targeted Genetics Corp TEROLOGIC NUCLEOTIDE SEQUENCE FROM A RECOMBINANT VIRAL VECTOR BY STRUCTURE OF THE SEQUENCE IN A WAY THAT BASE COUNTERATIONS ARISE WITHIN THE SEQUENCE
EP2267029B1 (en) 1999-09-03 2016-06-15 The Brigham And Women's Hospital, Inc. Methods and compositions for treatment of inflammatory disease using Cadherin-11 modulating agents
US7459540B1 (en) 1999-09-07 2008-12-02 Amgen Inc. Fibroblast growth factor-like polypeptides
US6432409B1 (en) * 1999-09-14 2002-08-13 Antigen Express, Inc. Hybrid peptides modulate the immune response
US20030235594A1 (en) * 1999-09-14 2003-12-25 Antigen Express, Inc. Ii-Key/antigenic epitope hybrid peptide vaccines
US9289487B2 (en) * 1999-09-14 2016-03-22 Antigen Express, Inc. II-key/antigenic epitope hybrid peptide vaccines
US6900043B1 (en) * 1999-09-21 2005-05-31 Amgen Inc. Phosphatases which activate map kinase pathways
AU782326B2 (en) 1999-10-12 2005-07-21 Lexicon Pharmaceuticals, Inc. Human LDL receptor family proteins and polynucleotides encoding the same
DE60012557T2 (en) 1999-10-15 2005-08-04 Genetics Institute, LLC, Cambridge HYALURONIC ACID COMPOSITIONS FOR THE DISPOSAL OF OSTEOGENIC PROTEINS
US7057015B1 (en) 1999-10-20 2006-06-06 The Salk Institute For Biological Studies Hormone receptor functional dimers and methods of their use
US6558932B1 (en) 1999-11-05 2003-05-06 The General Hospital Corp. Gridlock nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
US20050202538A1 (en) * 1999-11-12 2005-09-15 Merck Patent Gmbh Fc-erythropoietin fusion protein with improved pharmacokinetics
WO2001036489A2 (en) 1999-11-12 2001-05-25 Merck Patent Gmbh Erythropoietin forms with improved properties
WO2001036431A1 (en) 1999-11-15 2001-05-25 Parker Hughes Institute Diamino platinum (ii) antitumor complexes
US6589992B2 (en) 1999-11-30 2003-07-08 Parker Hughes Institute Inhibiting collagen-induced platelet aggregation
US6241710B1 (en) 1999-12-20 2001-06-05 Tricardia Llc Hypodermic needle with weeping tip and method of use
EP1242600B1 (en) 1999-12-23 2010-03-03 ZymoGenetics, Inc. Cytokine zcyto18
AU4314801A (en) 2000-02-11 2001-08-20 Lexigen Pharm Corp Enhancing the circulating half-life of antibody-based fusion proteins
US6841362B1 (en) 2000-02-29 2005-01-11 The Trustees Of Columbia University In The City Of New York Melanoma differentiation associated gene-7 promoter and uses thereof
US7229822B1 (en) * 2000-02-29 2007-06-12 Univ Columbia Melanoma differentation associated gene-5 and vectors and cells containing same
US20050239061A1 (en) * 2000-03-01 2005-10-27 Marshall William S Identification and use of effectors and allosteric molecules for the alteration of gene expression
US20040002068A1 (en) 2000-03-01 2004-01-01 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US7514239B2 (en) 2000-03-28 2009-04-07 Amgen Inc. Nucleic acid molecules encoding beta-like glycoprotein hormone polypeptides and heterodimers thereof
GB0018307D0 (en) 2000-07-26 2000-09-13 Aventis Pharm Prod Inc Polypeptides
US20050059584A1 (en) * 2002-08-16 2005-03-17 Ahmed Merzouk Novel chemokine mimetics synthesis and their use
US7368425B2 (en) * 2006-03-24 2008-05-06 Chemokine Therapeutics Corp. Cyclic peptides for modulating growth of neo-vessels and their use in therapeutic angiogenesis
CA2335109A1 (en) * 2000-04-12 2001-10-12 Chemokine Therapeutics Corporation Cxcr4 agonist treatment of hematopoietic cells
US7378098B2 (en) * 2000-04-12 2008-05-27 The University Of British Columbia CXC chemokine receptor 4 agonist peptides
EP1702983A3 (en) 2000-04-13 2007-01-10 Medical University of South Carolina Tissue-specific and pathogen-specific toxic agents, ribozymes, DNAzymes and antisense oligonucleotides and methods of use thereof
AU2001255379A1 (en) 2000-04-14 2001-10-30 The Uab Research Foundation Poliovirus replicons encoding therapeutic agents and uses thereof
CA2407309C (en) * 2000-04-28 2011-08-02 Xiao Xiao Dna sequences encoding dystrophin minigenes and methods of use thereof
US20020115642A1 (en) * 2000-05-02 2002-08-22 Chan Ming Fai Beta-lactam antibiotics
ES2531552T3 (en) 2000-05-10 2015-03-17 Mayo Foundation For Medical Education And Research Human IgM antibodies with the ability to induce remyelination and diagnostic and therapeutic uses thereof, particularly in the central nervous system
US7585497B2 (en) * 2000-05-17 2009-09-08 Oregon Health & Science University Induction of apoptosis and cell growth inhibition by protein 4.33
US6790667B1 (en) 2000-05-30 2004-09-14 Lexicon Genetics Incorporated Human mitochondrial proteins and polynucleotides encoding the same
EP2258725A3 (en) 2000-06-26 2014-09-17 ZymoGenetics, L.L.C. Cytokine receptor zcytor17
DE60137223D1 (en) 2000-06-28 2009-02-12 Amgen Inc THYMUS-STROMA LYMPHOPOIETIN RECEPTOR MOLECULES AND THEIR USE
AU7172901A (en) * 2000-06-29 2002-01-14 Lexigen Pharm Corp Enhancement of antibody-cytokine fusion protein mediated immune responses by combined treatment with immunocytokine uptake enhancing agents
EP1303502A2 (en) * 2000-07-06 2003-04-23 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Tetrahydrobenzothiazole analogues as neuroprotective agents
US6719970B1 (en) * 2000-07-10 2004-04-13 Alkermes Controlled Therapeutics, Inc. Method of generating cartilage
WO2002031115A2 (en) 2000-10-11 2002-04-18 Viron Therapeutics, Inc. Nucleic acid molecules and polypeptides for immune modulation
EP1325751A4 (en) * 2000-10-11 2005-05-04 Daiichi Seiyaku Co Novel drugs for liver diseases
AU784975B2 (en) 2000-10-11 2006-08-10 Sumitomo Chemical Company, Limited DNA-binding protein YB-1-containing collagen accumulation inhibitors
US6949379B2 (en) * 2000-10-20 2005-09-27 Canji, Inc. Aptamer-mediated regulation of gene expression
EP1666595A1 (en) 2000-10-26 2006-06-07 Beth Israel Deaconess Medical Center, Inc. GAB2 (P97) gene and methods of use thereof
AU2002232593A1 (en) * 2000-10-27 2002-05-06 Oregon Health And Science University Novel mutant igbp-3 molecules that do not bind to igfs, but retain their ability to functionally bind igfbp-3 receptor
US7060442B2 (en) * 2000-10-30 2006-06-13 Regents Of The University Of Michigan Modulators on Nod2 signaling
US7385023B1 (en) 2000-11-15 2008-06-10 Trustees Of Boston University Cancer immunotherapy and diagnosis using cytochrome P450 1B1
JP4520631B2 (en) * 2000-11-22 2010-08-11 松本油脂製薬株式会社 Durable water permeability-imparting agent and its fiber
CA2429599A1 (en) 2000-11-28 2002-06-06 Amgen Inc. Use of b7rp1 antagonists in ige-mediated disorders
US20040034045A1 (en) * 2000-11-29 2004-02-19 Parker Hughes Institute Inhibitors of thrombin induced platelet aggregation
US20030082233A1 (en) * 2000-12-01 2003-05-01 Lyons Karen M. Method and composition for modulating bone growth
TWI255272B (en) 2000-12-06 2006-05-21 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
JP4394878B2 (en) 2000-12-08 2010-01-06 ライフ テクノロジーズ コーポレーション Methods and compositions for nucleic acid molecule synthesis using multiple recognition sites
DK1355918T5 (en) 2000-12-28 2012-02-20 Wyeth Llc Recombinant protective protein of streptococcus pneumoniae
WO2002053701A2 (en) 2000-12-29 2002-07-11 Vanderbilt University Epididymal lipocalin gene and uses thereof
CA2434546C (en) 2001-01-12 2012-09-11 Chiron Corporation Induction of immune response by a replication-defective venezuelan equine encephalitis-sindbis chimeric virus replicon particle encoding an antigen
EP1363676B1 (en) * 2001-01-22 2007-03-28 Biogen Idec MA Inc. Method of enhancing delivery of a therapeutic nucleic acid
WO2002064162A2 (en) * 2001-02-13 2002-08-22 Government Of The United States, As Represented By The Secretary Of The Army Vaccine for transcutaneous immunization
PT1366067E (en) * 2001-03-07 2012-11-29 Merck Patent Gmbh Expression technology for proteins containing a hybrid isotype antibody moiety
US7723111B2 (en) * 2001-03-09 2010-05-25 The United States Of America As Represented By The Department Of Health And Human Services Activated dual specificity lymphocytes and their methods of use
FR2821947B1 (en) * 2001-03-12 2003-05-16 Canon Kk METHOD AND DEVICE FOR VALIDATING IMAGE DEFINING PARAMETERS
AU2002306709A1 (en) * 2001-03-14 2002-09-24 Replicon Technologies, Inc. Oncolytic rna replicons
US6613534B2 (en) 2001-03-20 2003-09-02 Wake Forest University Health Sciences MAP-2 as a determinant of metastatic potential
US20030026791A1 (en) * 2001-03-27 2003-02-06 Laurent Humeau Conditionally replicating vectors for inhibiting viral infections
US6992174B2 (en) 2001-03-30 2006-01-31 Emd Lexigen Research Center Corp. Reducing the immunogenicity of fusion proteins
CA2443493A1 (en) * 2001-04-13 2002-10-24 Wyeth Surface proteins of streptococcus pyogenes
US20070128229A1 (en) * 2002-04-12 2007-06-07 Wyeth Surface proteins of Streptococcus pyogenes
US7534444B2 (en) 2001-04-17 2009-05-19 Novattis Vaccines And Diagnostics, Inc. Molecular mimetics of meningococcal B epitopes which elicit functionally active antibodies
US7560424B2 (en) 2001-04-30 2009-07-14 Zystor Therapeutics, Inc. Targeted therapeutic proteins
US7629309B2 (en) 2002-05-29 2009-12-08 Zystor Therapeutics, Inc. Targeted therapeutic proteins
EP1974752B1 (en) 2001-04-30 2012-09-26 BioMarin Pharmaceutical Inc. Subcellular targeting of therapeutic proteins
MXPA03009924A (en) * 2001-05-03 2004-01-29 Merck Patent Gmbh Recombinant tumor specific antibody and use thereof.
EP2116259B1 (en) 2001-05-24 2012-01-25 ZymoGenetics, Inc. TACI-immunoglobulin fusion proteins
CA2449008A1 (en) * 2001-06-01 2002-12-12 Wyeth Compositions and methods for systemic administration of sequences encoding bone morphogenetic proteins
TWI267378B (en) * 2001-06-08 2006-12-01 Wyeth Corp Calcium phosphate delivery vehicles for osteoinductive proteins
AU2002320314A1 (en) 2001-07-05 2003-01-21 Chiron, Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
JP4302513B2 (en) 2001-07-05 2009-07-29 ノバルティス バクシンズ アンド ダイアグノスティックス,インコーポレーテッド Polynucleotides encoding antigenic type B HIV polypeptides and / or antigenic type C HIV polypeptides, polypeptides thereof and uses thereof
US20060275262A1 (en) * 2001-07-26 2006-12-07 Mathis James M Conditionally replicating viruses and methods for cancer virotherapy
WO2003015800A1 (en) 2001-08-09 2003-02-27 Ivoclar Vivadent, Inc. Tissue implants and methods for making and using same
US7419957B2 (en) * 2001-08-22 2008-09-02 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Peptides of melanoma antigen and their use in diagnostic, prophylactic and therapeutic methods
US20030134341A1 (en) * 2001-09-19 2003-07-17 Medcell Biologics, Llc. Th1 cell adoptive immunotherapy
US20030134415A1 (en) * 2001-09-19 2003-07-17 Gruenberg Micheal L. Th1 cell adoptive immunotherapy
US7582425B2 (en) * 2001-09-21 2009-09-01 The Regents Of The University Of Michigan Atlastin
US7108975B2 (en) * 2001-09-21 2006-09-19 Regents Of The University Of Michigan Atlastin
US20040023910A1 (en) * 2001-09-28 2004-02-05 Zhiming Zhang Use of cyr61 in the treatment and diagnosis of human uterine leiomyomas
AU2002343475A1 (en) 2001-10-03 2003-04-14 Selective Genetics, Inc. Traversal of nucleic acid molecules through a fluid space and expression in repair cells
AR045702A1 (en) 2001-10-03 2005-11-09 Chiron Corp COMPOSITIONS OF ASSISTANTS.
MX339524B (en) 2001-10-11 2016-05-30 Wyeth Corp Novel immunogenic compositions for the prevention and treatment of meningococcal disease.
ES2380612T3 (en) 2001-10-15 2012-05-16 Engeneic Molecular Delivery Pty Ltd. Intact mini cells as vectors for DNA transfer and gene therapy in vitro and in vivo
US20030072761A1 (en) * 2001-10-16 2003-04-17 Lebowitz Jonathan Methods and compositions for targeting proteins across the blood brain barrier
WO2003037376A1 (en) * 2001-11-02 2003-05-08 Kensuke Egashira Preventives and/or remedies for post-transplant arteriosclerosis as rejection of organ transplant
CN100534436C (en) 2001-11-21 2009-09-02 利兰·斯坦福青年大学托管委员会 Polynucleotide therapy
ES2381025T3 (en) * 2001-12-04 2012-05-22 Merck Patent Gmbh Immunocytokines with modulated selectivity
US7049121B2 (en) * 2001-12-20 2006-05-23 Applied Molecular Evolution Butyrylcholinesterase variant polypeptides with increased catalytic efficiency and methods of use
US6989261B2 (en) * 2001-12-20 2006-01-24 Eli Lilly And Company Butyrylcholinesterase variant polypeptides with increased catalytic efficiency and methods of use
RU2360923C2 (en) 2002-01-18 2009-07-10 Займоджинетикс, Инк. Novel ligand of cytokine receptor zcytor17
US7494804B2 (en) 2002-01-18 2009-02-24 Zymogenetics, Inc. Polynucleotide encoding cytokine receptor zcytor17 multimer
US20040009937A1 (en) * 2002-01-31 2004-01-15 Wei Chen Methods and composition for delivering nucleic acids and/or proteins to the respiratory system
US20040043003A1 (en) * 2002-01-31 2004-03-04 Wei Chen Clinical grade vectors based on natural microflora for use in delivering therapeutic compositions
US20050075298A1 (en) * 2002-01-31 2005-04-07 Wei Chen Methods and composition for delivering nucleic acids and/or proteins to the intestinal mucosa
US20040025194A1 (en) * 2002-02-22 2004-02-05 Board Of Trustees Of The University Of Illinois Beta chain-associated regulator of apoptosis
US7662924B2 (en) * 2002-02-22 2010-02-16 The Board Of Trustees Of The University Of Illinois Beta chain-associated regulator of apoptosis
US20030175272A1 (en) * 2002-03-07 2003-09-18 Medcell Biologics, Inc. Re-activated T-cells for adoptive immunotherapy
AU2003267949A1 (en) * 2002-03-15 2003-12-31 U.S. Government As Represented By The Department Of Veterans Affairs Methods and compositions using cellular asialodeterminants and glycoconjugates for targeting cells to tissues and organs
DE10213780A1 (en) * 2002-03-22 2003-11-27 Orthogen Ag Process and means for the production of therapeutically interesting blood compositions
JP2005530716A (en) * 2002-03-27 2005-10-13 アメリカ合衆国 Methods for treating cancer in humans
WO2003092594A2 (en) * 2002-04-30 2003-11-13 Duke University Adeno-associated viral vectors and methods for their production from hybrid adenovirus and for their use
EA008354B1 (en) * 2002-05-17 2007-04-27 Уайз Injectable solid hyaluronic acid carriers for delivery of osteogenic proteins
CA2484000A1 (en) 2002-05-24 2003-12-04 Schering Corporation Neutralizing human anti-igfr antibody
CN1671425B (en) * 2002-06-19 2013-07-10 大学保健网 ACE2 activation for treatment of heart, lung and kidney disease and hypertension
US20040009158A1 (en) * 2002-07-11 2004-01-15 Washington University Promotion of neovascularization using bone marrow-derived endothelial-progenitor cells
EP1563058A4 (en) * 2002-07-12 2005-12-28 Univ Singapore Hemangioblast progenitor cells
US20040052771A1 (en) * 2002-07-12 2004-03-18 Lim Sai Kiang Hemangioblast progenitor cells
US7785608B2 (en) * 2002-08-30 2010-08-31 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
US7179645B2 (en) * 2002-09-24 2007-02-20 Antigen Express, Inc. Ii-Key/antigenic epitope hybrid peptide vaccines
EP1556072B1 (en) 2002-09-17 2010-05-19 Antigen Express, Inc. Ii-KEY/ANTIGENIC EPITOPE HYBRID PEPTIDE VACCINES
AU2003275240A1 (en) * 2002-09-24 2004-04-23 Massachusetts Institute Of Technology Methods and compositions for soluble cpg15
WO2004032867A2 (en) * 2002-10-09 2004-04-22 Tolerrx, Inc. Molecules preferentially associated with effector t cells or regulatory t cells and methods of their use
AU2003269557A1 (en) 2002-10-18 2004-05-04 Lg Life Sciences Ltd. Gene families associated with cancers
US6863731B2 (en) * 2002-10-18 2005-03-08 Controls Corporation Of America System for deposition of inert barrier coating to increase corrosion resistance
US8076318B2 (en) * 2002-10-24 2011-12-13 Albert Einstein College Of Medicine Of Yeshiva University Caged ligands and uses thereof
TW200509968A (en) 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease
US8506959B2 (en) 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
NZ540194A (en) 2002-11-08 2008-07-31 Ablynx Nv Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
EP2267032A3 (en) 2002-11-08 2011-11-09 Ablynx N.V. Method of administering therapeutic polypeptides, and polypeptides therefor
CA2507249A1 (en) 2002-11-21 2004-06-10 Bayhill Therapeutics, Inc. Methods and immune modulatory nucleic acid compositions for preventing and treating disease
EP1581253A4 (en) * 2002-12-04 2007-02-14 Applied Molecular Evolution Butyrylcholinesterase variants that alter the activity of chemotherapeutic agents
WO2004055056A1 (en) * 2002-12-17 2004-07-01 Merck Patent Gmbh Humanized antibody (h14.18) of the mouse 14.18 antibody binding to gd2 and its fusion with il-2
WO2004060911A2 (en) * 2002-12-30 2004-07-22 Amgen Inc. Combination therapy with co-stimulatory factors
JP2006517789A (en) 2003-01-10 2006-08-03 アブリンクス エン.ヴェー. Therapeutic polypeptides, homologues thereof, fragments thereof, and use in modulating platelet-mediated aggregation
US20050048041A1 (en) * 2003-01-13 2005-03-03 Rao Mahendra S. Persistent expression of candidate molecule in proliferating stem and progenitor cells for delivery of therapeutic products
CA2802143C (en) 2003-01-14 2018-06-19 Dana-Farber Cancer Institute Sparc encoding polynucleotide as a cancer therapy sensitizer
CA2515708A1 (en) 2003-02-11 2004-08-26 Transkaryotic Therapies, Inc. Diagnosis and treatment of multiple sulfatase deficiency and other sulfatase deficiencies
US20070066548A1 (en) * 2003-03-14 2007-03-22 Albert Einstein College Of Medicine Of Yeshiva University Globin variant gene methods and compositions
WO2004094596A2 (en) * 2003-04-16 2004-11-04 Wyeth Holdings Corporation Novel immunogenic compositions for the prevention and treatment of meningococcal disease
US20040220242A1 (en) * 2003-05-02 2004-11-04 Leland Shapiro Inhibitors of serine protease activity, methods and compositions for treatment of nitric oxide induced clinical conditions
CN100581621C (en) 2003-05-16 2010-01-20 拉瓦勒大学 CNS chloride modulation and uses thereof
WO2006031210A1 (en) 2003-05-29 2006-03-23 Board Of Regents, The University Of Texas Systems Jabi as a prognostic marker and a therapeutic target for human cancer
AU2004245175C1 (en) 2003-06-10 2010-03-18 Biogen Ma Inc. Improved secretion of neublastin
EP1641491B1 (en) * 2003-06-11 2008-01-02 University of Chicago Increased t-cell tumor infiltration by mutant light
US7811983B2 (en) * 2003-06-11 2010-10-12 The University Of Chicago Increased T-cell tumor infiltration and eradication of metastases by mutant light
WO2005017109A2 (en) * 2003-06-30 2005-02-24 Massachusetts Institute Of Technology Nucleic acids and polypeptides required for cell survival in the absence of rb
US20050118145A1 (en) * 2003-07-03 2005-06-02 Jannette Dufour Compositions containing sertoli cells and myoid cells and use thereof in cellular transplants
ATE478963T1 (en) 2003-07-03 2010-09-15 Univ New Jersey Med GENES AS DIAGNOSTIC TOOLS FOR AUTISM
US20050013812A1 (en) * 2003-07-14 2005-01-20 Dow Steven W. Vaccines using pattern recognition receptor-ligand:lipid complexes
US20060035242A1 (en) 2004-08-13 2006-02-16 Michelitsch Melissa D Prion-specific peptide reagents
EP3192872A1 (en) 2003-08-26 2017-07-19 The Regents of the University of Colorado, a body corporate Inhibitors of serine protease activity and their use in methods and compositions for treatment of bacterial infections
US20050069521A1 (en) * 2003-08-28 2005-03-31 Emd Lexigen Research Center Corp. Enhancing the circulating half-life of interleukin-2 proteins
PL1675608T3 (en) * 2003-09-12 2007-11-30 Wyeth Corp Injectable calcium phosphate solid rods for delivery of osteogenic proteins
US20050187175A1 (en) * 2003-09-30 2005-08-25 Elly Nedivi Methods and compositions for CPG15-2
NZ547185A (en) 2003-10-20 2009-03-31 Nsgene As In vivo gene therapy of parkinson's disease
US20130266551A1 (en) 2003-11-05 2013-10-10 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1bb stimulatory signaling domain
US7435596B2 (en) 2004-11-04 2008-10-14 St. Jude Children's Research Hospital, Inc. Modified cell line and method for expansion of NK cell
DE60334645D1 (en) 2003-11-07 2010-12-02 Ablynx Nv CAMELIDAE HEAVY CHAIN ANTIBODIES VHHS AGAINST EPIDERMAL GROWTH FACTOR RECEPTOR (EGFR) AND ITS USE
EP3279328A1 (en) 2003-11-14 2018-02-07 Children's Medical Center Corporation Self-cleaving ribozymes and uses thereof
WO2005063820A2 (en) 2003-12-30 2005-07-14 Merck Patent Gmbh Il-7 fusion proteins
CN1902222A (en) * 2003-12-31 2007-01-24 默克专利有限公司 Fc-erythropoietin fusion protein with improved pharmacokinetics
DK1706428T3 (en) 2004-01-22 2009-11-30 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US7432057B2 (en) * 2004-01-30 2008-10-07 Michigan State University Genetic test for PSE-susceptible turkeys
US20050244400A1 (en) * 2004-02-10 2005-11-03 Zystor Therapeutics, Inc. Acid alpha-glucosidase and fragments thereof
US20050181035A1 (en) * 2004-02-17 2005-08-18 Dow Steven W. Systemic immune activation method using non CpG nucleic acids
AU2005214091B2 (en) * 2004-02-24 2010-08-12 Abbvie B.V. Method for determining the risk of developing a neurological disease
WO2005095450A2 (en) 2004-03-30 2005-10-13 Nsgene A/S Therapeutic use of a growth factor, nsg33
EP1758999B1 (en) 2004-06-22 2011-03-09 The Board Of Trustees Of The University Of Illinois METHODS OF INHIBITING TUMOR CELL PROLIFERATION WITH FOXM1 siRNA
US20060013772A1 (en) * 2004-06-30 2006-01-19 University Of Vermont And State Agricultural College Method and device to recover diagnostic and therapeutic agents
WO2006014422A2 (en) 2004-07-06 2006-02-09 The Trustees Of Columbia University In The City Of New York Polynucleotide encoding a trim-cyp polypeptide, compositions thereof, and methods of using same
US7604798B2 (en) * 2004-07-15 2009-10-20 Northwestern University Methods and compositions for importing nucleic acids into cell nuclei
CA2579574A1 (en) 2004-07-23 2007-01-04 The University Of North Carolina At Chapel Hill Methods and materials for determining pain sensitivity and predicting and treating related disorders
WO2006013462A2 (en) * 2004-07-30 2006-02-09 Nsgene A/S Growth factors nsg28, nsg30, and nsg32
US20090035784A1 (en) * 2004-07-30 2009-02-05 Mount Sinai School Of Medicine Of New York University Npc1l1 and npc1l1 inhibitors and methods of use thereof
US20060063208A1 (en) 2004-08-02 2006-03-23 Woolf Clifford J DRG11-responsive (DRAGON) gene and uses thereof
MX2007001679A (en) 2004-08-09 2007-05-23 Elan Pharm Inc Prevention and treatment of synucleinopathic and amyloidogenic disease.
EP1786473A4 (en) * 2004-08-11 2008-11-19 Cedars Sinai Medical Center Treatment of parkinson's disease and related disorders
EP2338524B1 (en) 2004-08-12 2013-05-22 Cedars-Sinai Medical Center Combined gene therapy for the treatment of macroscopic gliomas
WO2006024020A2 (en) 2004-08-27 2006-03-02 Novartis Vaccines And Diagnostics Inc. Hcv non-structural protein mutants and uses thereof
EP1802326A2 (en) * 2004-09-09 2007-07-04 Stryker Corporation Methods for treating bone tumors using bone morphogenic proteins
EP1846559A4 (en) 2004-09-11 2008-02-27 Joseph D Mosca Tumor-derived biological antigen presenting particles
US20060063187A1 (en) * 2004-09-15 2006-03-23 Hotamisligil Gokhan S Modulation of XBP-1 activity for treatment of metabolic disorders
US7635754B2 (en) * 2004-09-22 2009-12-22 Aerovance, Inc. Interleukin-9 and interleukin-4 chimeric antagonist muteins and methods of using same
BRPI0518151A2 (en) * 2004-10-13 2009-06-16 Ablynx Nv polypeptides against amyloid-beta, nucleic acid encoding such polypeptide, composition comprising such polypeptide, method for producing a polypeptide and use thereof
WO2006044984A1 (en) * 2004-10-18 2006-04-27 Mount Sinai School Of Medicine Of New York University Inhibition of tumor growth and metastasis by atf2-derived peptides
WO2006050394A2 (en) 2004-11-01 2006-05-11 Novartis Vaccines And Diagnostics Inc. Combination approaches for generating immune responses
CA2591297C (en) * 2004-12-09 2015-01-13 Stephen D. Gillies Il-7 variants with reduced immunogenicity
TW200714289A (en) * 2005-02-28 2007-04-16 Genentech Inc Treatment of bone disorders
WO2006099574A2 (en) * 2005-03-16 2006-09-21 Tai June Yoo Cockroach allergen gene expression and delivery systems and uses
KR100775958B1 (en) * 2005-03-30 2007-11-13 김정문 Non-activated Polypeptides Having a Function of Tissue Regeneration and Method for Preparing the Same
EP1863518A2 (en) * 2005-03-30 2007-12-12 Wyeth Methods for stimulating hair growth by administering bmps
EP1863850B1 (en) * 2005-03-30 2009-09-23 Jung Moon Kim Non-activated polypeptides having a function of tissue regeneration and method for preparing the same
WO2006118547A1 (en) 2005-04-29 2006-11-09 Agency For Science, Technology And Research Hyperbranched polymers and their applications
SI1888641T1 (en) 2005-05-18 2012-05-31 Ablynx Nv Serum albumin binding proteins
WO2006122825A2 (en) 2005-05-20 2006-11-23 Ablynx Nv Single domain vhh antibodies against von willebrand factor
US20090209621A1 (en) 2005-06-03 2009-08-20 The Johns Hopkins University Compositions and methods for decreasing microrna expression for the treatment of neoplasia
US20100129288A1 (en) * 2005-06-28 2010-05-27 Elior Peles Gliomedin, Fragments Thereof and Methods of Using Same
US7858590B2 (en) * 2005-08-11 2010-12-28 Cedars-Sinai Medical Center Treatment of parkinson's disease and related disorders
US20070104689A1 (en) * 2005-09-27 2007-05-10 Merck Patent Gmbh Compositions and methods for treating tumors presenting survivin antigens
PL1948798T3 (en) 2005-11-18 2015-12-31 Glenmark Pharmaceuticals Sa Anti-alpha2 integrin antibodies and their uses
EP1969001A2 (en) 2005-11-22 2008-09-17 Novartis Vaccines and Diagnostics, Inc. Norovirus and sapovirus antigens
US20090074733A1 (en) * 2005-12-09 2009-03-19 Medin Jeffrey A Thymidylate kinase mutants and uses thereof
US20090068158A1 (en) * 2005-12-09 2009-03-12 Medin Jeffrey A Thymidylate kinase mutants and uses thereof
US20080124303A1 (en) * 2005-12-12 2008-05-29 Cavit Sciences, Inc Methods and compositions for treatment of viral infections
WO2007076933A1 (en) 2005-12-30 2007-07-12 Merck Patent Gmbh Interleukin-12p40 variants with improved stability
ES2426468T3 (en) 2005-12-30 2013-10-23 Merck Patent Gmbh Anti-CD19 antibodies with reduced immunogenicity
US20090191597A1 (en) * 2006-01-20 2009-07-30 Asklepios Biopharmaceutical, Inc. Enhanced production of infectious parvovirus vectors in insect cells
TW200738752A (en) 2006-01-31 2007-10-16 Bayer Schering Pharma Ag Modulation of MDL-1 activity for treatment of inflammatory disease
WO2007095113A2 (en) * 2006-02-10 2007-08-23 Massachusetts Institute Of Technology Cpg15 and cpg15-2 compounds and inhibitors as insulin receptor and insulin-like growth factor receptor agonists and antagonists
AU2007219615B2 (en) 2006-03-03 2013-11-28 Promis Neurosciences Inc. Methods and compositions to treat and detect misfolded-SOD1 mediated diseases
WO2007120811A2 (en) 2006-04-14 2007-10-25 Advanced Cell Technology, Inc. Hemangio-colony forming cells
JP2009538924A (en) 2006-06-01 2009-11-12 エラン ファーマシューティカルズ,インコーポレイテッド A neuroactive fragment of APP
KR100954322B1 (en) 2006-06-14 2010-04-21 주식회사 엘지생명과학 Gene familyLBFL313 associated with pancreatic cancer
WO2008042436A2 (en) 2006-10-03 2008-04-10 Biogen Idec Ma Inc. Biomarkers and assays for the treatment of cancer
PL2068921T3 (en) 2006-10-19 2014-12-31 Csl Ltd High affinity antibody antagonists of interleukin-13 receptor alpha 1
US8613925B2 (en) 2006-10-19 2013-12-24 Csl Limited Anti-IL-13Rα1 antibodies and their uses thereof
EP2099523A2 (en) * 2006-11-13 2009-09-16 ZyStor Therapeutics , Inc. Methods for treating pompe disease
US9156914B2 (en) 2006-12-19 2015-10-13 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the ADAM family and polypeptides comprising the same for the treatment of ADAM-related diseases and disorders
EP2115004A2 (en) 2006-12-19 2009-11-11 Ablynx N.V. Amino acid sequences directed against gpcrs and polypeptides comprising the same for the treatment of gpcr-related diseases and disorders
US20090048146A1 (en) * 2006-12-21 2009-02-19 Alcon, Inc. Use of agents that upregulate crystallin expression in the retina and optic nerve head
AR064642A1 (en) * 2006-12-22 2009-04-15 Wyeth Corp POLINUCLEOTIDE VECTOR THAT INCLUDES IT RECOMBINATING CELL THAT UNDERSTANDS THE VECTOR POLYPEPTIDE, ANTIBODY, COMPOSITION THAT UNDERSTANDS THE POLINUCLEOTIDE, VECTOR, RECOMBINATING CELL POLYPEPTIDE OR ANTIBODY, USE OF THE COMPOSITION AND A COMPOSITION AND A METHOD
JP5829377B2 (en) * 2007-02-06 2015-12-09 ジュン ヨー,タイ Treatment and prevention of neurodegenerative diseases using gene therapy
WO2008098183A2 (en) * 2007-02-08 2008-08-14 The University Of Chicago Combination therapy for treating cancer
CA2678572C (en) 2007-02-16 2012-10-30 University Of Florida Research Foundation Inc. Mitochondrial targeting and import of a virus to deliver a nucleic acid
HUE043966T2 (en) 2007-02-23 2019-09-30 Prothena Biosciences Ltd Prevention and treatment of synucleinopathic and amyloidogenic disease
PL2118300T3 (en) 2007-02-23 2015-11-30 Prothena Biosciences Ltd Prevention and treatment of synucleinopathic and amyloidogenic disease
EP2139908A4 (en) * 2007-03-12 2011-02-16 Antigen Express Inc Li-rnai involved li suppression in cancer immunotherapy
CA2584494A1 (en) * 2007-03-27 2008-09-27 Jeffrey A. Medin Vector encoding therapeutic polypeptide and safety elements to clear transduced cells
EP2141997B1 (en) 2007-03-30 2012-10-31 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes
WO2008124768A1 (en) 2007-04-09 2008-10-16 The General Hospital Corporation Hemojuvelin fusion proteins and uses thereof
EP3492596A1 (en) 2007-04-09 2019-06-05 University of Florida Research Foundation, Inc. Raav vector compositions having tyrosine-modified capsid proteins and methods for use
WO2009014565A2 (en) 2007-04-26 2009-01-29 Ludwig Institute For Cancer Research, Ltd. Methods for diagnosing and treating astrocytomas
CA2723320C (en) 2007-05-04 2019-06-11 University Health Network Il-12 immunotherapy for cancer
CA2690281A1 (en) 2007-05-11 2008-11-20 The Johns Hopkins University Biomarkers for melanoma
ES2433967T3 (en) 2007-05-14 2013-12-13 The University Of Chicago Antibody-LIGHT fusion products as cancer therapeutic products
WO2008142693A2 (en) * 2007-05-22 2008-11-27 Yeda Research And Development Co. Ltd. Regulation of myelination by nectin-like (necl) molecules
GB0710529D0 (en) 2007-06-01 2007-07-11 Circassia Ltd Vaccine
US8889622B2 (en) * 2007-07-25 2014-11-18 Washington University Methods of inhibiting seizure in a subject
WO2009018250A2 (en) * 2007-07-30 2009-02-05 University Of Iowa Research Foundation Use of rpa4 and rpa4/rpa32 hybrid polypeptides for modulating cell proliferation
AU2008288283B2 (en) 2007-08-15 2013-01-31 Circassia Limited Peptides for desensibilization against allergens
US9040051B2 (en) 2007-10-02 2015-05-26 Universitaet Zu Koeln Marker genes for regulatory T cells from human blood
EA201000343A1 (en) 2007-10-04 2011-10-31 Займодженетикс, Инк. MEMBER OF THE FAMILY B7, zB7H6 AND RELATED COMPOSITIONS AND METHODS
WO2009078799A1 (en) 2007-12-17 2009-06-25 Marfl Ab New vaccine for the treatment of mycobacterium related disorders
EP2077119A1 (en) * 2007-12-21 2009-07-08 Apeiron Biologics Forschungs- und Entwicklungsgesellschaft M.B.H. Treatment of fibrosis and liver diseases
EP2237803B1 (en) 2007-12-28 2015-07-01 Prothena Biosciences Limited Treatment and prophylaxis of amyloidosis
US8591888B2 (en) 2008-01-11 2013-11-26 Synovex Corporation Cadherin-11 EC1 domain antagonists for treating inflammatory joint disorders
BRPI0906606A2 (en) * 2008-01-31 2015-07-14 Univ Vanderbilt Therapeutic treatment for lung conditions.
SG196863A1 (en) * 2008-01-31 2014-02-13 Univ Vanderbilt Methods and compositions for treatment for coronary and arterial aneurysmal subarachnoid hemorrhage
KR20110009095A (en) 2008-03-03 2011-01-27 더 유니버시티 오브 마이애미 Allogeneic cancer cell-based immunotherapy
EP2257569B1 (en) 2008-03-13 2014-10-01 Agriculture Victoria Services PTY Limited Vectors for expression of antimicrobial peptides in mammary gland
JP2011515399A (en) 2008-03-20 2011-05-19 ユニバーシティー オブ マイアミ Heat shock protein GP96 vaccination and method using the same
US8568709B2 (en) * 2008-03-20 2013-10-29 University Health Network Thymidylate kinase fusions and uses thereof
AU2009235467A1 (en) 2008-04-07 2009-10-15 Ablynx Nv Single variable domains against the Notch pathways
EP3062106B1 (en) 2008-04-16 2020-11-11 The Johns Hopkins University Method for determining androgen receptor variants in prostate cancer
CA2722693C (en) 2008-05-06 2023-03-21 Advanced Cell Technology, Inc. Hemangio colony forming cells and non-engrafting hemangio cells
CN102083960B (en) 2008-05-06 2014-12-03 先进细胞技术公司 Methods for producing enucleated erythroid cells derived from pluripotent stem cells
AU2009244148B2 (en) * 2008-05-07 2014-10-09 Biomarin Pharmaceutical Inc. Lysosomal targeting peptides and uses thereof
US20090291073A1 (en) * 2008-05-20 2009-11-26 Ward Keith W Compositions Comprising PKC-theta and Methods for Treating or Controlling Ophthalmic Disorders Using Same
US7914798B2 (en) 2008-06-20 2011-03-29 Wyeth Llc Compositions and methods of use of ORF1358 from beta-hemolytic streptococcal strains
US20120058105A1 (en) 2008-06-27 2012-03-08 Martin Kean Chong Ng Method of treatment of vascular complications
DK2153841T4 (en) 2008-08-15 2016-02-15 Circassia Ltd Vaccine comprising Amb a 1 peptides for use in the treatment of ambrosieallergi
EP2346904B1 (en) 2008-10-29 2017-04-12 China Synthetic Rubber Corporation Methods and agents for the diagnosis and treatment of hepatocellular carcinoma
JP2012507299A (en) * 2008-10-31 2012-03-29 バイオジェン・アイデック・エムエイ・インコーポレイテッド LIGHT target molecule and use thereof
ES2648231T3 (en) 2008-11-05 2017-12-29 Wyeth Llc Multi-component immunogenic composition for the prevention of beta hemolytic streptococcal disease (EBH)
US8835617B2 (en) 2008-11-06 2014-09-16 The Trustees Of Columbia University In The City Of New York Polynucleotides encoding a human TRIM-Cyp fusion polypeptide, compositions thereof, and methods of using same
AU2010203223B9 (en) * 2009-01-05 2015-10-08 Epitogenesis Inc. Adjuvant compositions and methods of use
EP2389191A2 (en) 2009-01-23 2011-11-30 NsGene A/S Expression of neuropeptides in mammalian cells
JP2012516357A (en) 2009-01-29 2012-07-19 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Methods for distributing high levels of therapeutic agents throughout the cortex to treat neurological disorders
PL2393830T3 (en) 2009-02-05 2015-07-31 Circassia Ltd Grass peptides for vaccine
WO2010093784A2 (en) 2009-02-11 2010-08-19 The University Of North Carolina At Chapel Hill Modified virus vectors and methods of making and using the same
EP2221066A1 (en) 2009-02-18 2010-08-25 Sanofi-Aventis Use of VgII3 activity modulator for the modulation of adipogenesis
CN105399828B (en) 2009-04-10 2021-01-15 埃博灵克斯股份有限公司 Improved amino acid sequences directed against IL-6R and polypeptides comprising the same for the treatment of IL-6R related diseases and disorders
HRP20212024T1 (en) 2009-05-02 2022-04-01 Genzyme Corporation Gene therapy for neurodegenerative disorders
NZ597314A (en) 2009-06-05 2013-07-26 Ablynx Nv Monovalent, bivalent and trivalent anti human respiratory syncytial virus (hrsv) nanobody constructs for the prevention and/or treatment of respiratory tract infections
US8785168B2 (en) 2009-06-17 2014-07-22 Biomarin Pharmaceutical Inc. Formulations for lysosomal enzymes
US8455191B2 (en) 2009-08-28 2013-06-04 Miami University Cell transdifferentiation into brown adipocytes
US20120283115A1 (en) 2009-08-31 2012-11-08 Ludwig Institute For Cancer Research Ltd. Seromic analysis of ovarian cancer
GB0915794D0 (en) 2009-09-09 2009-10-07 Ucl Business Plc Screening method and treatment
WO2011032100A1 (en) 2009-09-11 2011-03-17 Government Of The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Inhibitors of kshv vil6 and human il6
US20110117113A1 (en) 2009-10-09 2011-05-19 Gerald Beste Immunoglobulin single variable domain directed against human cxcr4 and other cell associated proteins and methods to generate them
EP2498825B1 (en) 2009-11-09 2017-03-29 Genepod Therapeutics Ab Novel viral vector construct for neuron specific continuous dopa synthesis in vivo
WO2011064382A1 (en) 2009-11-30 2011-06-03 Ablynx N.V. Improved amino acid sequences directed against human respiratory syncytial virus (hrsv) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
US9872905B2 (en) 2009-12-01 2018-01-23 President And Fellows Of Harvard College Modulation of NK cell antigen specific effector activity by modulation of CXCR6 (CD186)
US8962807B2 (en) 2009-12-14 2015-02-24 Ablynx N.V. Single variable domain antibodies against OX40L, constructs and therapeutic use
CN102802658A (en) 2009-12-21 2012-11-28 台湾东洋药品工业股份有限公司 Methods and compositions related to reduced met phosphorylation by leukocyte cell-derived chemotaxin 2 in tumor cells
WO2011083140A1 (en) 2010-01-08 2011-07-14 Ablynx Nv Immunoglobulin single variable domain directed against human cxcr4
WO2011088081A1 (en) 2010-01-12 2011-07-21 The University Of North Carolina At Chapel Hill Restrictive inverted terminal repeats for viral vectors
US9120855B2 (en) 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
GB201002559D0 (en) 2010-02-15 2010-03-31 Circassia Ltd Birch peptides for vaccine
GB201004475D0 (en) 2010-03-17 2010-05-05 Isis Innovation Gene silencing
EP2552962B1 (en) 2010-03-26 2016-03-23 Ablynx N.V. Immunoglobulin single variable domains directed against cxcr7
CA2805267C (en) 2010-05-04 2019-07-30 The Brigham And Women's Hospital, Inc. Detection and treatment of fibrosis
EP3546483A1 (en) 2010-05-20 2019-10-02 Ablynx N.V. Biological materials related to her3
WO2011146938A1 (en) 2010-05-21 2011-11-24 NanoOncology, Inc. Reagents and methods for treating cancer
EP2580238A1 (en) 2010-06-09 2013-04-17 Zymogenetics, Inc. Dimeric vstm3 fusion proteins and related compositions and methods
LT2593128T (en) 2010-07-15 2018-06-25 Adheron Therapeutics, Inc. Humanized antibodies targeting the ec1 domain of cadherin-11 and related compositions and methods
GB201014026D0 (en) 2010-08-20 2010-10-06 Ucl Business Plc Treatment
CA2808975C (en) 2010-08-23 2018-10-30 Wyeth Llc Stable formulations of neisseria meningitidis rlp2086 antigens
WO2012025759A2 (en) 2010-08-26 2012-03-01 Isis Innovation Limited Method
EP3056212B1 (en) 2010-09-10 2019-04-03 Wyeth LLC Non-lipidated variants of neisseria meningitidis orf2086 antigens
EP2621953B1 (en) 2010-09-30 2017-04-05 Ablynx N.V. Biological materials related to c-met
AU2011307488B2 (en) 2010-10-01 2015-08-20 Hoba Therapeutics Aps Use of meteorin for the treatment of allodynia, hyperalgesia, spontaneous pain and phantom pain
KR101470908B1 (en) * 2010-10-07 2014-12-09 가톨릭대학교 산학협력단 Anticancer immunotherapeutic agent
WO2012051301A1 (en) 2010-10-12 2012-04-19 President And Fellows Of Harvard College Methods for identifying modulators of triglyceride metabolism, for modulating triglyceride metabolism and for identifying subjects at risk for abnormal triglyceride metabolism
US20130273093A1 (en) 2010-10-18 2013-10-17 Peter Charles Leonard Beverley Method for Immunising a Subject against Mycobacterium Tuberculosis or Mycobacterium Bovis
US8815942B2 (en) 2010-10-20 2014-08-26 The Royal Institution For The Advancement Of Learning/Mcgill University Combination therapy and uses thereof for treatment and prevention of parasitic infection and disease
MX343085B (en) 2010-11-08 2016-10-24 Novartis Ag Cxcr2 binding polypeptides.
US9072766B2 (en) 2010-11-18 2015-07-07 Beth Israel Deaconess Medical Center, Inc. Methods of treating obesity by inhibiting nicotinamide N-methyl transferase (NNMT)
DK3214091T3 (en) 2010-12-09 2019-01-07 Univ Pennsylvania USE OF CHEMICAL ANTIGEN RECEPTOR MODIFIED T CELLS FOR TREATMENT OF CANCER
WO2012094511A2 (en) 2011-01-05 2012-07-12 Rhode Island Hospital Compositions and methods for the treatment of orthopedic disease or injury
WO2012095548A2 (en) 2011-01-13 2012-07-19 Centro De Investigación Biomédica En Red De Enfermedades Neurodegenerativas (Ciberned) Compounds for treating neurodegenerative disorders
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
WO2012109238A2 (en) 2011-02-07 2012-08-16 President And Fellows Of Harvard College Methods for increasing immune responses using agents that directly bind to and activate ire-1
US9409953B2 (en) 2011-02-10 2016-08-09 The University Of North Carolina At Chapel Hill Viral vectors with modified transduction profiles and methods of making and using the same
US20150158948A9 (en) 2011-03-28 2015-06-11 Francis Descamps Bispecific anti-cxcr7 immunoglobulin single variable domains
US20140155469A1 (en) 2011-04-19 2014-06-05 The Research Foundation Of State University Of New York Adeno-associated-virus rep sequences, vectors and viruses
UA117218C2 (en) 2011-05-05 2018-07-10 Мерк Патент Гмбх Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
US9880151B2 (en) 2011-05-23 2018-01-30 Phylogica Limited Method of determining, identifying or isolating cell-penetrating peptides
CN103717235A (en) 2011-06-24 2014-04-09 埃皮托吉尼西斯有限公司 Pharmaceutical compositions, comprising a combination of select carriers, vitamins, tannins and flavonoids as antigen-specific immuno-modulators
SI2751279T1 (en) 2011-08-31 2018-01-31 St. Jude Children's Research Hospital Methods and compositions to detect the level of lysosomal exocytosis activity and methods of use
CN104105501B (en) 2011-09-05 2017-10-20 霍巴治疗公司 Allodynia, hyperalgia, the treatment of spontaneous pain and phantom pain
JP2014528715A (en) 2011-09-15 2014-10-30 ロサンゼルス バイオメディカル リサーチ インスティテュート アットハーバー− ユーシーエルエー メディカル センター Immunotherapy and diagnosis of mucormycosis using CotH
CA2848740A1 (en) 2011-09-16 2013-03-21 Mingxing Wang Amphiphilic cationic polymers for the delivery of therapeutic agents
KR102130439B1 (en) 2011-09-19 2020-07-07 악손 뉴로사이언스 에스이 Protein-based therapy and diagnosis of tau-mediated pathology in alzheimer's disease
WO2013044225A1 (en) 2011-09-22 2013-03-28 The Trustees Of The University Of Pennsylvania A universal immune receptor expressed by t cells for the targeting of diverse and multiple antigens
CA2850261C (en) 2011-09-30 2021-04-20 Ablynx Nv C-met immunoglobulin single variable domains
CA3077804C (en) 2012-01-09 2023-07-25 Serpin Pharma, Llc Peptides and methods of using same
WO2013106358A1 (en) 2012-01-10 2013-07-18 Hussain M Mahmood Method of treating hyperlipidemia and atherosclerosis with mir-30c
MX2014010181A (en) 2012-02-22 2015-03-20 Univ Pennsylvania Use of icos-based cars to enhance antitumor activity and car persistence.
US9783591B2 (en) 2012-02-22 2017-10-10 The Trustees Of The University Of Pennsylvania Use of the CD2 signaling domain in second-generation chimeric antigen receptors
EP2817318A4 (en) 2012-02-22 2016-04-27 Univ Pennsylvania Compositions and methods for generating a persisting population of t cells useful for the treatment of cancer
CN107056901B (en) 2012-03-09 2021-05-07 辉瑞公司 Meningococcal compositions and methods thereof
SA115360586B1 (en) 2012-03-09 2017-04-12 فايزر انك Neisseria meningitidis compositions and methods thereof
JP2015516400A (en) 2012-04-24 2015-06-11 ユニバーシティー オブ マイアミUniversity Of Miami Perforin 2 protection against invasive and multi-drug resistant pathogens
US9328174B2 (en) 2012-05-09 2016-05-03 Novartis Ag Chemokine receptor binding polypeptides
US9050296B2 (en) 2012-07-03 2015-06-09 Maine Medical Center Methods for treating metabolic syndrome with Cthrc1 polypeptide
JP2015524255A (en) 2012-07-13 2015-08-24 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Method for enhancing the activity of CART cells by co-introducing bispecific antibodies
KR20210149195A (en) 2012-07-13 2021-12-08 더 트러스티스 오브 더 유니버시티 오브 펜실베니아 Methods of assessing the suitability of transduced T cells for administration
WO2014039513A2 (en) 2012-09-04 2014-03-13 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive t cell transfer
EP3738974A1 (en) 2012-09-28 2020-11-18 The University of North Carolina at Chapel Hill Aav vectors targeted to oligodendrocytes
NZ746914A (en) 2012-10-02 2020-03-27 Memorial Sloan Kettering Cancer Center Compositions and methods for immunotherapy
WO2014055771A1 (en) 2012-10-05 2014-04-10 The Trustees Of The University Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
AU2013204200B2 (en) 2012-10-11 2016-10-20 Brandeis University Treatment of amyotrophic lateral sclerosis
WO2014074785A1 (en) 2012-11-08 2014-05-15 Ludwig Institute For Cancer Research Ltd. Methods of predicting outcome and treating breast cancer
AU2013344375B2 (en) 2012-11-16 2017-09-14 Transposagen Biopharmaceuticals, Inc. Site-specific enzymes and methods of use
WO2014107739A1 (en) 2013-01-07 2014-07-10 Eleven Biotherapeutics, Inc. Antibodies against pcsk9
US9394368B2 (en) 2013-02-20 2016-07-19 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
EP2958942B1 (en) 2013-02-20 2020-06-03 Novartis AG Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
AU2014223601B9 (en) 2013-02-26 2020-04-23 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
US8957044B2 (en) 2013-03-01 2015-02-17 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
CA2903716C (en) 2013-03-08 2019-04-09 Pfizer Inc. Immunogenic fusion polypeptides
CA2940513C (en) 2013-03-11 2023-08-15 University Of Florida Research Foundation, Inc. Delivery of card protein as therapy for ocular inflammation
DK2968470T3 (en) 2013-03-12 2021-02-01 Massachusetts Gen Hospital MODIFIED MULER INHIBITANT SUBSTANCE (MIS) PROTEINS AND USES THEREOF FOR THE TREATMENT OF DISEASES
EP2968526A4 (en) 2013-03-14 2016-11-09 Abbott Lab Hcv antigen-antibody combination assay and methods and compositions for use therein
MX2015012825A (en) 2013-03-14 2016-06-10 Abbott Lab Hcv core lipid binding domain monoclonal antibodies.
JP2016512241A (en) 2013-03-14 2016-04-25 アボット・ラボラトリーズAbbott Laboratories HCVNS3 recombinant antigen for improved antibody detection and mutants thereof
MX2015012274A (en) 2013-03-14 2016-06-02 Univ Maryland Androgen receptor down-regulating agents and uses thereof.
EP3517612A1 (en) 2013-03-15 2019-07-31 The University of North Carolina At Chapel Hill Synthetic adeno-associated virus inverted terminal repeats
CA2907184C (en) 2013-03-15 2022-12-06 Sutter West Bay Hospitals Falz for use as a target for therapies to treat cancer
SG11201507688VA (en) 2013-03-15 2015-10-29 Sloan Kettering Inst Cancer Compositions and methods for immunotherapy
WO2014144229A1 (en) 2013-03-15 2014-09-18 The University Of North Carolina At Chapel Hill Methods and compositions for dual glycan binding aav vectors
UY35468A (en) 2013-03-16 2014-10-31 Novartis Ag CANCER TREATMENT USING AN ANTI-CD19 CHEMERIC ANTIGEN RECEIVER
EP2978442B1 (en) 2013-03-29 2020-03-18 The Regents of the University of Colorado, a body corporate Alpha 1 antitrypsin of use for preparing a subject for transplant
PT2981607T (en) 2013-04-03 2020-11-20 Memorial Sloan Kettering Cancer Center Effective generation of tumor-targeted t-cells derived from pluripotent stem cells
US9644215B2 (en) 2013-04-12 2017-05-09 The General Hospital Corporation AAV1-caspase gene therapy induced pyroptosis for the treatment of tumors
WO2014191630A2 (en) 2013-05-28 2014-12-04 Helsingin Yliopisto Non-human animal model encoding a non-functional manf gene
EP3008191A2 (en) 2013-06-13 2016-04-20 Shire Human Genetic Therapies, Inc. Messenger rna based viral production
WO2015031686A1 (en) 2013-08-30 2015-03-05 Amgen Inc. High titer recombinant aav vector production in adherent and suspension cells
RU2662968C2 (en) 2013-09-08 2018-07-31 Пфайзер Инк. Immunogenic composition for neisseria meningitidis (options)
CN105764926A (en) 2013-09-24 2016-07-13 梅迪塞纳医疗股份有限公司 Interleukin-4 receptor-binding fusion proteins and uses thereof
US10781242B2 (en) 2013-09-24 2020-09-22 Medicenna Therapeutics Inc. Interleukin-2 fusion proteins and uses thereof
WO2015048331A1 (en) 2013-09-25 2015-04-02 Cornell University Compounds for inducing anti-tumor immunity and methods thereof
AU2014331938A1 (en) 2013-10-09 2016-04-28 University Of Miami Perforin-2 activators and inhibitors as drug targets for infectious disease and gut inflammation
WO2015058018A1 (en) 2013-10-17 2015-04-23 National University Of Singapore Chimeric receptor that triggers antibody-dependent cell cytotoxicity against multiple tumors
JP6317440B2 (en) 2013-10-29 2018-04-25 プレジデント アンド フェローズ オブ ハーバード カレッジ Methods and compositions for inhibiting oxidative stress
US11135269B2 (en) 2013-12-11 2021-10-05 The General Hospital Corporation Use of mullerian inhibiting substance (MIS) proteins for contraception and ovarian reserve preservation
CA2931684C (en) 2013-12-19 2024-02-20 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
US11053291B2 (en) 2014-02-19 2021-07-06 University Of Florida Research Foundation, Incorporated Delivery of Nrf2 as therapy for protection against reactive oxygen species
WO2015124546A1 (en) 2014-02-19 2015-08-27 Fundación Centro Nacional De Investigaciones Cardiovasculares Carlos Iii- Cnic Aav vectors for the treatment of ischemic and non-ischemic heart disease
US20170335281A1 (en) 2014-03-15 2017-11-23 Novartis Ag Treatment of cancer using chimeric antigen receptor
EP3122872B1 (en) 2014-03-27 2019-07-03 The Salk Institute for Biological Studies Compositions and methods for treating type 1 and type 2 diabetes and related disorders
IL307423A (en) 2014-04-07 2023-12-01 Novartis Ag Treatment of cancer using anti-cd19 chimeric antigen receptor
AU2015259877B2 (en) 2014-05-15 2021-02-25 National University Of Singapore Modified natural killer cells and uses thereof
WO2015179404A1 (en) 2014-05-19 2015-11-26 The Johns Hopkins University Methods for identifying androgen receptor splice variants in subjects having castration resistant prostate cancer
EP4166148A1 (en) 2014-06-06 2023-04-19 Memorial Sloan-Kettering Cancer Center Mesothelin-targeted chimeric antigen receptors and uses thereof
EP3155128B1 (en) 2014-06-10 2019-05-15 Erasmus University Medical Center Rotterdam Methods for characterizing alternatively or aberrantly spliced mrna isoforms
WO2015189429A1 (en) 2014-06-13 2015-12-17 Universidade De Santiago De Compostela Nanoparticulate systems for use in gene transfer or gene delivery
NL2013661B1 (en) 2014-10-21 2016-10-05 Ablynx Nv KV1.3 Binding immunoglobulins.
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
EP4205749A1 (en) 2014-07-31 2023-07-05 Novartis AG Subset-optimized chimeric antigen receptor-containing cells
JP2017522895A (en) 2014-07-31 2017-08-17 ザ ボード オブ リージェンツ オブ ザ ユニヴァーシティ オブ オクラホマ High isomerohydrolase activity mutant of mammalian RPE65
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
CN112410363A (en) 2014-08-19 2021-02-26 诺华股份有限公司 anti-CD 123 Chimeric Antigen Receptor (CAR) for cancer therapy
JP6860476B2 (en) 2014-08-25 2021-04-14 ザ ジョンズ ホプキンズ ユニヴァーシティー Methods and Compositions for Prostate Cancer Treatment
KR102500531B1 (en) 2014-09-04 2023-02-17 메모리얼 슬로안 케터링 캔서 센터 Globin gene therapy for treating hemoglobinopathies
SG10201902168PA (en) 2014-09-09 2019-04-29 Unum Therapeutics Chimeric receptors and uses thereof in immune therapy
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016044707A1 (en) 2014-09-18 2016-03-24 Cedars-Sinai Medical Center Compositions and methods for treating fibrosis
RU2743657C2 (en) 2014-10-08 2021-02-20 Новартис Аг Biomarkers predicting a therapeutic response to therapy with a chimeric antigen receptor, and use thereof
NZ731673A (en) 2014-11-21 2024-07-26 Univ North Carolina Chapel Hill Aav vectors targeted to the central nervous system
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
KR20240130831A (en) 2014-12-05 2024-08-29 메모리얼 슬로안 케터링 캔서 센터 Chimeric antigen receptors targeting b-cell maturation antigen and uses thereof
JP7174522B2 (en) 2014-12-05 2022-11-17 メモリアル スローン ケタリング キャンサー センター Chimeric antigen receptor targeting Fc receptor-like 5 and uses thereof
SG11201704552TA (en) 2014-12-05 2017-07-28 Memorial Sloan Kettering Cancer Center Antibodies targeting g-protein coupled receptor and methods of use
SI3227339T1 (en) 2014-12-05 2022-02-28 Memorial Sloan-Kettering Cancer Center Chimeric antigen receptors targeting g-protein coupled receptor and uses thereof
JP6938377B2 (en) 2015-01-14 2021-09-22 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル Methods and compositions for targeting gene transfer
MA41349A (en) 2015-01-14 2017-11-21 Univ Temple RNA-GUIDED ERADICATION OF HERPES SIMPLEX TYPE I AND OTHER ASSOCIATED HERPES VIRUSES
GB201501004D0 (en) 2015-01-21 2015-03-04 Cancer Rec Tech Ltd Inhibitors
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
AU2016215124B2 (en) 2015-02-06 2020-08-06 The University Of North Carolina At Chapel Hill Optimized human clotting Factor VIII gene expression cassettes and their use
WO2016132294A1 (en) 2015-02-19 2016-08-25 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
JP6495468B2 (en) 2015-02-27 2019-04-03 ソーク インスティチュート フォー バイオロジカル スタディーズ Reprogramming precursor composition and method of use thereof
MA41629A (en) 2015-03-04 2018-01-09 Center For Human Reproduction COMPOSITIONS AND METHODS OF USE OF ANTI-MÜLLERIAN HORMONE FOR THE TREATMENT OF INFERTILITY
CN114478791A (en) 2015-04-03 2022-05-13 优瑞科生物技术公司 Constructs targeting AFP peptide/MHC complexes and uses thereof
SG11201708191XA (en) 2015-04-08 2017-11-29 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
EP4194001A1 (en) 2015-04-22 2023-06-14 Cedars-Sinai Medical Center Enterically delivered bitter oligopeptides for the treatment for type 2 diabetes and obesity
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
WO2016174652A1 (en) 2015-04-30 2016-11-03 Technion Research & Development Foundation Limited Chimeric antigen receptors and methods of their use
EP4088732A1 (en) 2015-05-01 2022-11-16 The Regents of The University of California Glycan-dependent immunotherapeutic molecules
DK3288379T3 (en) 2015-05-01 2022-02-07 Univ Michigan Regents PEPTIME COMPOSITIONS AND METHODS OF APPLICATION
CN114634943A (en) 2015-05-18 2022-06-17 T细胞受体治疗公司 Compositions and methods for reprogramming TCRs using fusion proteins
IL274151B (en) 2015-05-21 2022-07-01 Harpoon Therapeutics Inc Trispecific binding proteins and methods of use
KR102637402B1 (en) 2015-06-17 2024-02-15 포세이다 테라퓨틱스, 인크. Compositions and methods for inducing proteins to specific loci in the genome
US10550379B2 (en) 2015-06-29 2020-02-04 The Board Of Trustees Of The Leland Stanford Junior University Degron fusion constructs and methods for controlling protein production
EP3328994A4 (en) 2015-07-31 2019-04-17 Memorial Sloan-Kettering Cancer Center Antigen-binding proteins targeting cd56 and uses thereof
CA2992511A1 (en) 2015-08-03 2017-02-09 Myodopa Limited Systemic synthesis and regulation of l-dopa
CN113429455A (en) 2015-08-28 2021-09-24 赛品制药有限责任公司 Methods for disease treatment
EP3344996A2 (en) 2015-09-03 2018-07-11 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
US11242375B2 (en) 2015-09-04 2022-02-08 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use
CA2996420A1 (en) 2015-09-28 2017-04-06 The University Of North Carolina At Chapel Hill Methods and compositions for antibody-evading virus vectors
IL258405B (en) 2015-10-23 2022-09-01 Eureka Therapeutics Inc Antibody/t-cell receptor chimeric constructs and uses thereof
CA3006759A1 (en) 2015-11-30 2017-06-08 The Regents Of The University Of California Tumor-specific payload delivery and immune activation using a human antibody targeting a highly specific tumor cell surface antigen
MY197441A (en) 2015-12-04 2023-06-19 Memorial Sloan Kettering Cancer Center Antibodies targeting fc receptor-like 5 and method of use
US10590169B2 (en) 2015-12-09 2020-03-17 Virogin Biotech Canada Ltd Compositions and methods for inhibiting CD279 interactions
AU2016366226B2 (en) 2015-12-09 2023-06-01 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of using same
BR112018011881A2 (en) 2015-12-14 2018-12-04 The University Of North Carolina At Chapel Hill modified capsid proteins for increased release of parvovirus vectors
US11441146B2 (en) 2016-01-11 2022-09-13 Christiana Care Health Services, Inc. Compositions and methods for improving homogeneity of DNA generated using a CRISPR/Cas9 cleavage system
US11116850B2 (en) 2016-02-22 2021-09-14 The University Of North Carolina At Chapel Hill AAV-IDUA vector for treatment of MPS I-associated blindness
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
EP3426804A4 (en) 2016-03-11 2020-03-25 The Brigham and Women's Hospital, Inc. Compositions and methods for treating chemotherapy resistant cancer
CA3018382A1 (en) 2016-03-29 2017-10-05 University Of Southern California Chimeric antigen receptors targeting cancer
ES2933961T3 (en) 2016-04-15 2023-02-15 Memorial Sloan Kettering Cancer Center Transgenic T Cells and Chimeric Antigen Receptor T Cell Compositions and Related Methods
SI3443096T1 (en) 2016-04-15 2023-07-31 Novartis Ag Compositions and methods for selective expression of chimeric antigen receptors
EP4273248A3 (en) 2016-05-20 2024-01-10 Braingene AB Destabilising domains for conditionally stabilising a protein
WO2017201488A1 (en) 2016-05-20 2017-11-23 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
AU2017269364B2 (en) 2016-05-25 2023-08-31 Salk Institute For Biological Studies Compositions and methods for organoid generation and disease modeling
EP3464375A2 (en) 2016-06-02 2019-04-10 Novartis AG Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
RU2764919C2 (en) 2016-06-13 2022-01-24 Дзе Юниверсити Оф Норт Каролина Эт Чепел Хилл Optimized cln1 genes and expression cassettes, and their application
US10183058B2 (en) 2016-06-17 2019-01-22 Nymox Corporation Method of preventing or reducing the progression of prostate cancer
CA2971303A1 (en) 2016-06-21 2017-12-21 Bamboo Therapeutics, Inc. Optimized mini-dystrophin genes and expression cassettes and their use
MX2019000088A (en) 2016-06-27 2019-08-29 Broad Inst Inc Compositions and methods for detecting and treating diabetes.
WO2018001858A1 (en) 2016-06-27 2018-01-04 University Of Copenhagen Tailored assembly of a modular bud polypeptide
JP2018035137A (en) 2016-07-13 2018-03-08 マブイミューン ダイアグノスティックス エイジーMabimmune Diagnostics Ag Novel anti-fibroblast activated protein (FAP) binding agent and use thereof
US20190336504A1 (en) 2016-07-15 2019-11-07 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
AU2017299854A1 (en) 2016-07-18 2019-01-31 Helix Biopharma Corp. CAR immune cells directed to carcinoembryonic antigen related cell adhesion molecule 6 to treat cancer
RU2019103384A (en) 2016-07-26 2020-08-26 Дзе Юниверсити Оф Норт Каролина Эт Чепел Хилл VECTOR-MEDIATED IMMUNOLOGICAL EYE TOLERANCE
US10172910B2 (en) 2016-07-28 2019-01-08 Nymox Corporation Method of preventing or reducing the incidence of acute urinary retention
BR112019001570A2 (en) 2016-07-28 2019-07-09 Novartis Ag chimeric antigen receptor combination therapies and pd-1 inhibitors
EP3490590A2 (en) 2016-08-01 2019-06-05 Novartis AG Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
CA3032498A1 (en) 2016-08-02 2018-02-08 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018044866A1 (en) 2016-08-30 2018-03-08 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use for treating viral and other infections
US10532081B2 (en) 2016-09-07 2020-01-14 Nymox Corporation Method of ameliorating or preventing the worsening or the progression of symptoms of BPH
CN117866991A (en) 2016-10-07 2024-04-12 诺华股份有限公司 Chimeric antigen receptor for the treatment of cancer
JP7217970B2 (en) 2016-10-07 2023-02-06 ティーシーアール2 セラピューティクス インク. Compositions and methods for reprogramming T-cell receptors using fusion proteins
US10172933B2 (en) 2016-10-31 2019-01-08 The United States Of America, As Represented By The Secretary Of Agriculture Mosaic vaccines for serotype a foot-and-mouth disease virus
CA3044593A1 (en) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
CA3044729A1 (en) 2016-11-23 2018-05-31 Harpoon Therapeutics, Inc. Psma targeting trispecific proteins and methods of use
EA201991168A1 (en) 2016-11-23 2019-12-30 Харпун Терапьютикс, Инк. PROTEIN BINDING PROSTATIC SPECIFIC MEMBRANE ANTIGEN
CN110234343A (en) 2016-12-21 2019-09-13 T细胞受体治疗公司 Engineered T cell for treating cancer
EP3568470B1 (en) 2017-01-10 2022-07-06 Christiana Care Health Services, Inc. Methods for in vitro site-directed mutagenesis using gene editing technologies
EP3568468A4 (en) 2017-01-12 2020-12-30 Eureka Therapeutics, Inc. Constructs targeting histone h3 peptide/mhc complexes and uses thereof
EP3574005B1 (en) 2017-01-26 2021-12-15 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
CN110234658B (en) 2017-01-31 2024-03-12 辉瑞大药厂 Neisseria meningitidis compositions and methods of use thereof
CN108395482B (en) 2017-02-08 2021-02-05 西比曼生物科技(香港)有限公司 Construction of targeting CD20 antigen chimeric antigen receptor and activity identification of engineered T cell thereof
WO2018148667A1 (en) 2017-02-10 2018-08-16 Memorial Sloan-Kettering Cancer Center Reprogramming cell aging
US11597911B2 (en) 2017-02-27 2023-03-07 Life Technologies Corporation Expansion of populations of T cells by the use of modified serum free media
WO2018160754A2 (en) 2017-02-28 2018-09-07 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
WO2018161092A1 (en) 2017-03-03 2018-09-07 New York University Induction and enhancement of antitumor immunity involving virus vectors expressing multiple epitopes of tumor associated antigens and immune checkpoint inhibitors or proteins
EP3592368A1 (en) 2017-03-08 2020-01-15 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of use
IL268891B2 (en) 2017-03-15 2024-09-01 Univ North Carolina Chapel Hill Polyploid adeno-associated virus vectors and methods of making and using the same
CN117384929A (en) 2017-03-27 2024-01-12 新加坡国立大学 Polynucleotide encoding chimeric receptor expressed by cell
MX2019011570A (en) 2017-03-27 2019-11-18 Nat Univ Singapore Stimulatory cell lines for ex vivo expansion and activation of natural killer cells.
US10934336B2 (en) 2017-04-13 2021-03-02 The Trustees Of The University Of Pennsylvania Use of gene editing to generate universal TCR re-directed T cells for adoptive immunotherapy
US11613573B2 (en) 2017-04-26 2023-03-28 Eureka Therapeutics, Inc. Chimeric antibody/T-cell receptor constructs and uses thereof
US11447564B2 (en) 2017-04-26 2022-09-20 Eureka Therapeutics, Inc. Constructs specifically recognizing glypican 3 and uses thereof
US20200055948A1 (en) 2017-04-28 2020-02-20 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
BR112019023856A2 (en) 2017-05-12 2020-06-09 Harpoon Therapeutics Inc triespecific proteins targeting msln and methods of use
WO2018209298A1 (en) 2017-05-12 2018-11-15 Harpoon Therapeutics, Inc. Mesothelin binding proteins
WO2018220236A1 (en) 2017-06-02 2018-12-06 Merck Patent Gmbh Polypeptides binding adamts5, mmp13 and aggrecan
MX2019014448A (en) 2017-06-02 2020-02-10 Merck Patent Gmbh Mmp13 binding immunoglobulins.
BR112019024333A2 (en) 2017-06-02 2020-07-28 Merck Patent Gmbh adamts binding immunoglobulins
TW202413408A (en) 2017-06-02 2024-04-01 比利時商艾伯林克斯公司 Aggrecan binding immunoglobulins
EP3638295A1 (en) 2017-06-13 2020-04-22 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US20210145828A1 (en) 2017-06-29 2021-05-20 Rutgers, The State University Of New Jersey Compositions And Methods Targeting G12 Signaling For Bronchodilator Therapy
CN111032074A (en) 2017-07-03 2020-04-17 斯特拉斯堡大学 MTMR2-S polypeptide for use in the treatment of myopathy
GB201710973D0 (en) 2017-07-07 2017-08-23 Avacta Life Sciences Ltd Scaffold proteins
CN109456943A (en) 2017-09-06 2019-03-12 亘喜生物科技(上海)有限公司 Universal Chimeric antigen receptor T cell technology of preparing
WO2019055853A1 (en) 2017-09-15 2019-03-21 Life Technologies Corporation Compositions and methods for culturing and expanding cells
EP3684421A4 (en) 2017-09-18 2021-08-04 Children's Hospital Medical Center A strong insulator and uses thereof in gene delivery
MX2020003046A (en) 2017-09-19 2020-10-12 Univ British Columbia Anti-hla-a2 antibodies and methods of using the same.
CN111448216B (en) 2017-09-20 2023-11-07 英属哥伦比亚大学 Novel anti-HLA-A 2 antibodies and uses thereof
CN109554348A (en) 2017-09-27 2019-04-02 亘喜生物科技(上海)有限公司 It can induce the engineering immunocyte of secretion anti-cd 47 antibody
US20200271657A1 (en) 2017-10-04 2020-08-27 Opko Pharmaceuticals, Llc Articles and methods directed to personalized therapy of cancer
CR20200195A (en) 2017-10-13 2020-08-14 Harpoon Therapeutics Inc B cell maturation antigen binding proteins
EP3694529B1 (en) 2017-10-13 2024-06-26 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
EP4124658A3 (en) 2017-10-16 2023-04-19 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
US20200237799A1 (en) 2017-10-16 2020-07-30 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis (als)
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
EP3707265A4 (en) 2017-11-07 2021-09-01 The University of North Carolina at Chapel Hill Optimized aga genes and expression cassettes and their use
WO2019092251A1 (en) 2017-11-11 2019-05-16 Universite De Strasbourg Compositions and method for the treatment of x-linked centronuclear myopathy
JP2021502829A (en) 2017-11-14 2021-02-04 メモリアル スローン ケタリング キャンサー センター Immune-responsive cells secreting IL-36 and their use
EP3710040A1 (en) 2017-11-15 2020-09-23 Novartis AG Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
EP3717907A1 (en) 2017-11-30 2020-10-07 Novartis AG Bcma-targeting chimeric antigen receptor, and uses thereof
EP3720509A4 (en) 2017-12-06 2021-12-08 Memorial Sloan-Kettering Cancer Center Globin gene therapy for treating hemoglobinopathies
WO2019126578A1 (en) 2017-12-20 2019-06-27 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
US20210061875A1 (en) 2017-12-29 2021-03-04 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Compositions and methods for treating autoimmune disease
TW201930591A (en) 2018-01-08 2019-08-01 瑞士商諾華公司 Immune-enhancing RNAs for combination with chimeric antigen receptor therapy
CA3090249A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
WO2019152868A1 (en) 2018-02-01 2019-08-08 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating herpes simplex virus (hsv) related keratitis
BR112020016138A2 (en) 2018-02-11 2020-12-15 Memorial Sloan-Kettering Cancer Center T-CELL RECEPTORS NOT RESTRICTED TO HLA AND USES OF THE SAME
EP3752532A1 (en) 2018-02-12 2020-12-23 Diabetes-Free, Inc. Improved antagonistic anti-human cd40 monoclonal antibodies
WO2019160956A1 (en) 2018-02-13 2019-08-22 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
WO2019161133A1 (en) 2018-02-15 2019-08-22 Memorial Sloan Kettering Cancer Center Foxp3 targeting agent compositions and methods of use for adoptive cell therapy
US12097257B2 (en) 2018-03-05 2024-09-24 New York University Induction and enhancement of antitumor immunity involving Sindbis virus vectors expressing immune checkpoint proteins
MX2020009514A (en) 2018-03-14 2020-12-07 Beijing Xuanyi Pharmasciences Co Ltd Anti-claudin 18.2 antibodies.
WO2019175380A2 (en) 2018-03-16 2019-09-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from secretogranin v and uses thereof for the diagnosis and treatment of type 1 diabetes
WO2019175381A1 (en) 2018-03-16 2019-09-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from pcsk2 and uses thereof for the diagnosis and treatment of type 1 diabetes
US12098179B2 (en) 2018-03-16 2024-09-24 Inserm (Institut National De La Sante Et De La Recherche Medicale) Antigenic peptides deriving from urocortin 3 and uses thereof for the diagnosis and treatment of type 1 diabetes
SG11202009542PA (en) 2018-03-29 2020-10-29 Genentech Inc Modulating lactogenic activity in mammalian cells
SG11202009451VA (en) 2018-04-03 2020-10-29 Stridebio Inc Antibody-evading virus vectors
MX2020010464A (en) 2018-04-03 2021-01-29 Antibody-evading virus vectors.
CA3094465A1 (en) 2018-04-03 2019-10-10 Stridebio, Inc. Virus vectors for targeting ophthalmic tissues
CN116514995A (en) 2018-04-12 2023-08-01 上海赛比曼生物科技有限公司 Chimeric antigen receptor targeting BCMA and preparation method and application thereof
WO2019196087A1 (en) 2018-04-13 2019-10-17 Syz Cell Therapy Co. Methods of cancer treatment using tumor antigen-specific t cells
WO2019196088A1 (en) 2018-04-13 2019-10-17 Syz Cell Therapy Co. Methods of obtaining tumor-specific t cell receptors
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
US20210396739A1 (en) 2018-05-01 2021-12-23 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019217512A1 (en) 2018-05-08 2019-11-14 Life Technologies Corporation Compositions and methods for culturing and expanding cells
WO2019217837A1 (en) 2018-05-11 2019-11-14 Memorial Sloan-Kettering Cancer Center T cell receptors targeting pik3ca mutations and uses thereof
WO2019217831A1 (en) 2018-05-11 2019-11-14 Memorial Sloan-Kettering Cancer Center Methods for identifying antigen-specific t cell receptors
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019241324A1 (en) 2018-06-12 2019-12-19 The University Of North Carolina At Chapel Hill Synthetic liver-tropic adeno-associated virus capsids and uses thereof
JP7438988B2 (en) 2018-06-13 2024-02-27 ノバルティス アーゲー BCMA chimeric antigen receptor and its use
AU2019288136A1 (en) 2018-06-18 2021-01-07 Eureka Therapeutics, Inc. Constructs targeting prostate-specific membrane antigen (PSMA) and uses thereof
EP3814513A4 (en) 2018-06-28 2022-04-13 The University of North Carolina at Chapel Hill Optimized cln5 genes and expression cassettes and their use
WO2020010035A1 (en) 2018-07-02 2020-01-09 Voyager Therapeutics, Inc. Cannula system
AU2019299861A1 (en) 2018-07-02 2021-01-14 Voyager Therapeutics, Inc. Treatment of amyotrophic lateral sclerosis and disorders associated with the spinal cord
AU2019299439A1 (en) 2018-07-03 2021-01-21 Sotio, LLC Chimeric receptors in combination with trans metabolism molecules enhancing glucose import and therapeutic uses thereof
GB201811382D0 (en) 2018-07-11 2018-08-29 Taylor John Hermon Vaccine
BR112021001568A2 (en) 2018-08-10 2021-05-04 The University Of North Carolina At Chapel Hill optimized cln7 genes and expression cassettes and their use
AU2019321540A1 (en) 2018-08-14 2021-02-11 Sotio, LLC Chimeric antigen receptor polypeptides in combination with trans metabolism molecules modulating Krebs cycle and therapeutic uses thereof
EP3844192A1 (en) 2018-08-30 2021-07-07 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
AU2019346466A1 (en) 2018-09-25 2021-05-20 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
CN114957475B (en) 2018-09-26 2023-06-20 福州拓新天成生物科技有限公司 anti-B7-H3 monoclonal antibodies and their use in cell therapy
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
EP3856779A1 (en) 2018-09-28 2021-08-04 Novartis AG Cd22 chimeric antigen receptor (car) therapies
WO2020081929A1 (en) 2018-10-19 2020-04-23 University Of Rochester Immune modulators in combination with radiation treatment for advanced pancreatic cancer
KR20210082205A (en) 2018-10-22 2021-07-02 유니버시티 오브 로체스터 Genome editing by induced heterologous DNA insertion using a retroviral integrase-Cas9 fusion protein
WO2020102454A1 (en) 2018-11-13 2020-05-22 Regents Of The University Of Minnesota Cd40 targeted peptides and uses thereof
KR20210101235A (en) 2018-11-16 2021-08-18 메모리얼 슬로안 케터링 캔서 센터 Antibodies to mucin-16 and methods of use thereof
WO2020140007A1 (en) 2018-12-28 2020-07-02 University Of Rochester Gene therapy for best1 dominant mutations
KR20210126014A (en) 2019-01-14 2021-10-19 유니버시티 오브 로체스터 Nuclear RNA cleavage and polyadenylation targeted with CRISPR-Cas
GB201900858D0 (en) 2019-01-22 2019-03-13 Price Nicola Kaye Receptors providing targeted costimulation for adoptive cell therapy
US20220088075A1 (en) 2019-02-22 2022-03-24 The Trustees Of The University Of Pennsylvania Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
EP3773918A4 (en) 2019-03-05 2022-01-05 Nkarta, Inc. Cd19-directed chimeric antigen receptors and uses thereof in immunotherapy
RU2742000C2 (en) 2019-03-13 2021-02-01 Общество С Ограниченной Ответственностью "Анабион" Isolated alternative intracellular signalling domain of chimeric antigen receptor and chimeric antigen receptor including it
JP7379654B2 (en) 2019-03-15 2023-11-14 カーティザン セラピューティクス,インコーポレーテッド Anti-BCMA chimeric antigen receptor
US20220184176A1 (en) 2019-03-20 2022-06-16 Centre National De La Recherche Scientifique Amphiphysin / bin1 for the treatment of autosomal dominant centronuclear myopathy
AR118465A1 (en) 2019-03-21 2021-10-06 Stridebio Inc RECOMBINANT ADENO-ASSOCIATED VIRUS VECTORS
CN111793627A (en) 2019-04-08 2020-10-20 中国科学院上海生命科学研究院 RNA fixed-point editing by utilizing artificially constructed RNA editing enzyme and related application
EP3957325A4 (en) 2019-04-19 2023-01-04 Chugai Seiyaku Kabushiki Kaisha Chimeric receptor recognizing modification site of antibody
EP3959232B1 (en) 2019-04-24 2024-10-16 University of Massachusetts Aav capsid chimeric antigen receptors and uses thereof
EP3959320A1 (en) 2019-04-24 2022-03-02 Novartis AG Compositions and methods for selective protein degradation
AU2020263392A1 (en) 2019-04-26 2021-10-28 The University Of North Carolina At Chapel Hill Methods and compositions for dual glycan binding AAV2.5 vector
SG11202111130SA (en) 2019-04-30 2021-11-29 Senti Biosciences Inc Chimeric receptors and methods of use thereof
KR20220005050A (en) 2019-05-01 2022-01-12 팩트 파마, 인크. Compositions and methods for the treatment of cancer using TET2 engineered T cell therapy
US20220202864A1 (en) 2019-05-07 2022-06-30 Gracell Biotechnologies (Shanghai) Co., Ltd. Bcma-targeting engineered immune cell and use thereof
CN113784732B (en) 2019-05-07 2024-03-22 亘喜生物科技(上海)有限公司 Engineered immune cells targeting BCMA and uses thereof
EP4023230A4 (en) 2019-06-05 2023-11-15 Chugai Seiyaku Kabushiki Kaisha Antibody cleavage site-binding molecule
EP3990030A1 (en) 2019-06-27 2022-05-04 Pfizer Inc. Methods of treating duchenne muscular dystrophy using aav mini-dystrophin gene therapy
US20220380800A1 (en) 2019-07-02 2022-12-01 M6P Therapeutics (Switzerland) Llc Vector compositions and methods of using same for treatment of lysosomal storage disorders
WO2021016453A1 (en) 2019-07-23 2021-01-28 University Of Rochester Targeted rna cleavage with crispr-cas
CN112300997A (en) 2019-08-01 2021-02-02 上海赛比曼生物科技有限公司 Universal CAR-T cell and preparation and application thereof
EP4013788A1 (en) 2019-08-12 2022-06-22 Purinomia Biotech, Inc. Methods and compositions for promoting and potentiating t-cell mediated immune responses through adcc targeting of cd39 expressing cells
WO2021072218A1 (en) 2019-10-10 2021-04-15 Pact Pharma, Inc. Method of treating immunotherapy non-responders with an autologous cell therapy
TW202128775A (en) 2019-10-16 2021-08-01 英商阿法克塔生命科學有限公司 Pd-l1 inhibitor - tgfβ inhibitor bispecific drug moieties
CA3157700A1 (en) 2019-10-17 2021-04-22 Stridebio, Inc. Adeno-associated viral vectors for treatment of niemann-pick disease type c
CN110760007B (en) 2019-11-21 2022-08-26 博生吉医药科技(苏州)有限公司 CD7-CAR-T cell and preparation and application thereof
CN114945382A (en) 2019-11-26 2022-08-26 诺华股份有限公司 CD19 and CD22 chimeric antigen receptors and uses thereof
US20230078498A1 (en) 2020-02-07 2023-03-16 University Of Rochester Targeted Translation of RNA with CRISPR-Cas13 to Enhance Protein Synthesis
US20230073250A1 (en) 2020-02-07 2023-03-09 University Of Rochester Ribozyme-mediated RNA Assembly and Expression
EP4106806A4 (en) 2020-02-21 2024-07-24 Harpoon Therapeutics Inc Flt3 binding proteins and methods of use
EP4110823A1 (en) 2020-02-26 2023-01-04 A2 Biotherapeutics, Inc. Polypeptides targeting mage-a3 peptide-mhc complexes and methods of use thereof
WO2021170067A1 (en) 2020-02-28 2021-09-02 上海复宏汉霖生物技术股份有限公司 Anti-cd137 construct and use thereof
EP4110826A4 (en) 2020-02-28 2024-08-14 Shanghai Henlius Biotech Inc Anti-cd137 constructs, multispecific antibody and uses thereof
WO2021194915A1 (en) 2020-03-23 2021-09-30 The University Of North Carolina At Chapel Hill Aav-naglu vectors for treatment of mucopolysaccharidosis iiib
MX2022011805A (en) 2020-03-24 2023-01-11 Generation Bio Co Non-viral dna vectors and uses thereof for expressing factor ix therapeutics.
CN115667531A (en) 2020-03-24 2023-01-31 世代生物公司 Non-viral DNA vectors and their use for expressing Gaucher (Gaucher) therapeutics
CN115315512A (en) 2020-03-26 2022-11-08 基因泰克公司 Modified mammalian cells with reduced host cell proteins
EP4127170A1 (en) 2020-03-27 2023-02-08 University of Rochester Crispr-cas13 crrna arrays
WO2021195519A1 (en) 2020-03-27 2021-09-30 University Of Rochester Targeted destruction of viral rna by crispr-cas13
CN113527507A (en) 2020-04-16 2021-10-22 上海赛比曼生物科技有限公司 Chimeric antigen receptor targeting CD22 and preparation method and application thereof
WO2021226513A1 (en) 2020-05-08 2021-11-11 President And Fellows Of Harvard College Methods for treating inflammatory and autoimmune disorders
US20230212231A1 (en) 2020-05-26 2023-07-06 Institut National De La Santé Et De La Recherche Médicale (Inserm) Severe acute respiratory syndrome coronavirus 2 (sars-cov-2) polypeptides and uses thereof for vaccine purposes
EP4157881A4 (en) 2020-05-27 2024-10-09 Staidson Beijing Biopharmaceuticals Co Ltd Antibodies specifically recognizing nerve growth factor and uses thereof
WO2021247995A2 (en) 2020-06-04 2021-12-09 Voyager Therapeutics, Inc. Compositions and methods of treating neuropathic pain
EP4161552A1 (en) 2020-06-05 2023-04-12 The Broad Institute, Inc. Compositions and methods for treating neoplasia
GB202101299D0 (en) 2020-06-09 2021-03-17 Avacta Life Sciences Ltd Diagnostic polypetides and methods
KR20230024967A (en) 2020-06-11 2023-02-21 노파르티스 아게 ZBTB32 Inhibitors and Uses Thereof
KR20230026491A (en) 2020-06-24 2023-02-24 제넨테크, 인크. Apoptosis resistant cell lines
EP4182455A1 (en) 2020-07-15 2023-05-24 University of Rochester Targeted rna cleavage with dcasl3-rnase fusion proteins
US20230257713A1 (en) 2020-07-16 2023-08-17 Shanghai Jiaotong University Immunotherapy method of targeted chemokine and cytokine delivery by mesenchymal stem cell
IL299886A (en) 2020-07-17 2023-03-01 Instil Bio Uk Ltd Receptors providing targeted costimulation for adoptive cell therapy
AU2021310467A1 (en) 2020-07-17 2023-03-09 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
JPWO2022025220A1 (en) 2020-07-31 2022-02-03
EP4200408A1 (en) 2020-08-19 2023-06-28 Sarepta Therapeutics, Inc. Adeno-associated virus vectors for treatment of rett syndrome
WO2022093769A1 (en) 2020-10-28 2022-05-05 The University Of North Carolina At Chapel Hill Methods and compositions for dual glycan binding aav2.5 vector
AU2021369480A1 (en) 2020-10-28 2023-04-20 The University Of North Carolina At Chapel Hill Methods and compositions for dual glycan binding AAV2.5 vector
US20240033358A1 (en) 2020-11-13 2024-02-01 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022101463A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of the last c-terminal residues m31/41 of zikv m ectodomain for triggering apoptotic cell death
CN114525306A (en) 2020-11-23 2022-05-24 博生吉医药科技(苏州)有限公司 Preparation method and application of CD7-CAR-T cell
TW202237639A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
TW202237638A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
TW202241935A (en) 2020-12-18 2022-11-01 美商世紀治療股份有限公司 Chimeric antigen receptor system with adaptable receptor specificity
EP4271482A2 (en) 2020-12-31 2023-11-08 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
CN114763381A (en) 2021-01-13 2022-07-19 博生吉医药科技(苏州)有限公司 B7-H3 chimeric antigen receptor modified T cell and application thereof
TW202242124A (en) 2021-01-14 2022-11-01 美商史崔德生物公司 Aav vectors targeting t-cells
MX2023007788A (en) 2021-01-24 2023-11-17 Michael David Forrest Inhibitors of atp synthase - cosmetic and therapeutic uses.
GB2603166A (en) 2021-01-29 2022-08-03 Thelper As Therapeutic and Diagnostic Agents and Uses Thereof
CN118146395A (en) 2021-02-08 2024-06-07 浙江大学 Chimeric antigen receptor using endogenous protein molecules to replace single domain antibodies
KR20230167397A (en) 2021-04-08 2023-12-08 얀센 바이오테크 인코포레이티드 Materials and methods for engineering enhanced stem cell-like memory T cells
CA3216491A1 (en) 2021-04-16 2022-10-20 Asklepios Biopharmaceutical, Inc. Rational polyploid aav virions that cross the blood brain barrier and elicit reduced humoral response
KR20230173164A (en) 2021-04-19 2023-12-26 제넨테크, 인크. modified mammalian cells
KR20240000580A (en) 2021-04-23 2024-01-02 유니버시티 오브 로체스터 Genome editing and treatment method by direct non-homologous DNA insertion using retroviral integrase-Cas fusion protein
CN117881786A (en) 2021-04-27 2024-04-12 世代生物公司 Non-viral DNA vectors expressing therapeutic antibodies and uses thereof
WO2022232286A1 (en) 2021-04-27 2022-11-03 Generation Bio Co. Non-viral dna vectors expressing anti-coronavirus antibodies and uses thereof
KR20240006581A (en) 2021-05-06 2024-01-15 호바 세라퓨틱스 에이피에스 Prevention and treatment of chemotherapy-induced neuropathic pain
WO2022234003A1 (en) 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
JP2024521107A (en) 2021-05-21 2024-05-28 ジェネンテック, インコーポレイテッド Modified cells for producing recombinant products of interest
WO2022254337A1 (en) 2021-06-01 2022-12-08 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
CN115477704B (en) 2021-06-16 2024-02-23 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cells constructed based on LOX1
CN115477705B (en) 2021-06-16 2024-02-23 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cells constructed based on granzyme B
US11945876B2 (en) 2021-06-16 2024-04-02 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
EP4355785A1 (en) 2021-06-17 2024-04-24 Amberstone Biosciences, Inc. Anti-cd3 constructs and uses thereof
EP4363059A1 (en) 2021-06-29 2024-05-08 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
TW202317633A (en) 2021-07-08 2023-05-01 美商舒泰神(加州)生物科技有限公司 Antibodies specifically recognizing tnfr2 and uses thereof
CN115812082A (en) 2021-07-14 2023-03-17 舒泰神(北京)生物制药股份有限公司 Antibody specifically recognizing CD40 and application thereof
EP4370213A1 (en) 2021-07-16 2024-05-22 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2023049933A1 (en) 2021-09-27 2023-03-30 Sotio Biotech Inc. Chimeric receptor polypeptides in combination with trans metabolism molecules that re-direct glucose metabolites out of the glycolysis pathway and therapeutic uses thereof
CN115873802A (en) 2021-09-29 2023-03-31 亘喜生物科技(上海)有限公司 Chimeric antigen receptor immune cell and preparation method and application thereof
MX2024003778A (en) 2021-09-30 2024-04-10 Akouos Inc Compositions and methods for treating kcnq4-associated hearing loss.
WO2023057567A1 (en) 2021-10-07 2023-04-13 Avacta Life Sciences Limited Pd-l1 binding affimers
TW202332694A (en) 2021-10-07 2023-08-16 英商阿凡克塔生命科學公司 Serum half-life extended pd-l1 binding polypeptides
CA3234811A1 (en) 2021-10-20 2023-04-27 Steven Goldman Rejuvenation treatment of age-related white matter loss
CA3236365A1 (en) 2021-11-02 2023-05-11 University Of Rochester Tcf7l2 mediated remyelination in the brain
WO2023081813A1 (en) 2021-11-05 2023-05-11 St. Jude Children's Research Hospital, Inc. Zip cytokine receptors
WO2023081894A2 (en) 2021-11-08 2023-05-11 St. Jude Children's Research Hospital, Inc. Pre-effector car-t cell gene signatures
CN116102658A (en) 2021-11-09 2023-05-12 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cells constructed based on GAS6
CN118488969A (en) 2021-11-16 2024-08-13 舒迪安生物技术公司 Treatment of patients with myxoid/round cell liposarcoma
WO2023091954A2 (en) 2021-11-19 2023-05-25 The Trustees Of The University Of Pennsylvania Engineered pan-leukocyte antigen cd45 to facilitate car t cell therapy
EP4445911A1 (en) 2021-12-06 2024-10-16 Beijing SoloBio Genetechnology Co., Ltd. Bispecific antibody that specifically binds to klebsiella pneumoniae o2 and o1 antigens, and composition
AU2022405685A1 (en) 2021-12-10 2024-07-11 Hoba Therapeutics Aps Treatment of nociceptive pain
EP4456910A1 (en) 2021-12-28 2024-11-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139269A1 (en) 2022-01-21 2023-07-27 Mnemo Therapeutics Modulation of suv39h1 expression by rnas
GB202201137D0 (en) 2022-01-28 2022-03-16 Thelper As Therapeutic and diagnostic agents and uses thereof
TW202342759A (en) 2022-02-04 2023-11-01 美商史崔德生物公司 Recombinant adeno-associated virus vectors, and methods of use thereof
AU2023235112A1 (en) 2022-03-14 2024-10-17 Generation Bio Co. Heterologous prime boost vaccine compositions and methods of use
WO2023202967A1 (en) 2022-04-19 2023-10-26 F. Hoffmann-La Roche Ag Improved production cells
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023223292A1 (en) 2022-05-20 2023-11-23 Takeda Pharmaceutical Company Limited Methods of producing engineered immune cells
WO2023223291A1 (en) 2022-05-20 2023-11-23 Takeda Pharmaceutical Company Limited Methods of producing engineered immune cells
WO2023232961A1 (en) 2022-06-03 2023-12-07 F. Hoffmann-La Roche Ag Improved production cells
WO2023240182A1 (en) 2022-06-08 2023-12-14 St. Jude Children's Research Hospital, Inc. Disruption of kdm4a in t cells to enhance immunotherapy
TW202417503A (en) 2022-07-19 2024-05-01 美商舒泰神(加州)生物科技有限公司 Antibodies specifically recognizing b- and t-lymphocyte attenuator (btla) and uses thereof
WO2024040208A1 (en) 2022-08-19 2024-02-22 Sotio Biotech Inc. Genetically engineered immune cells with chimeric receptor polypeptides in combination with multiple trans metabolism molecules and therapeutic uses thereof
WO2024040207A1 (en) 2022-08-19 2024-02-22 Sotio Biotech Inc. Genetically engineered natural killer (nk) cells with chimeric receptor polypeptides in combination with trans metabolism molecules and therapeutic uses thereof
WO2024040222A1 (en) 2022-08-19 2024-02-22 Generation Bio Co. Cleavable closed-ended dna (cedna) and methods of use thereof
WO2024059787A1 (en) 2022-09-16 2024-03-21 St. Jude Children's Research Hospital, Inc. Disruption of asxl1 in t cells to enhance immunotherapy
WO2024062138A1 (en) 2022-09-23 2024-03-28 Mnemo Therapeutics Immune cells comprising a modified suv39h1 gene
WO2024079662A1 (en) 2022-10-11 2024-04-18 Meiragtx Uk Ii Limited Upf1 expression constructs
WO2024079530A2 (en) 2022-10-11 2024-04-18 Meiragtx Uk Ii Limited Nucleic acid regulatory elements for gene expression in the muscle and methods of use
WO2024083843A1 (en) 2022-10-18 2024-04-25 Confo Therapeutics N.V. Amino acid sequences directed against the melanocortin 4 receptor and polypeptides comprising the same for the treatment of mc4r-related diseases and disorders
WO2024091824A1 (en) 2022-10-26 2024-05-02 Ada Forsyth Institute, Inc. Differentiation and reprogramming of chondrocyte
WO2024123797A1 (en) 2022-12-06 2024-06-13 The Broad Institute, Inc. Compositions and methods for modulating neuronal excitability
WO2024124019A2 (en) 2022-12-07 2024-06-13 Ginkgo Bioworks, Inc. Aav vectors targeting hematopoietic stem cells
WO2024134434A1 (en) 2022-12-19 2024-06-27 Takeda Pharmaceutical Company Limited Method for producing car-t cells
WO2024133600A1 (en) 2022-12-20 2024-06-27 Nanocell Therapeutics B.V. Integrating and self-inactivating viral vectors and constructs and uses thereof
WO2024151958A1 (en) 2023-01-13 2024-07-18 The Broad Institute, Inc. Compositions and methods for treating sensory processing abnormalities
WO2024163747A2 (en) 2023-02-02 2024-08-08 University Of Rochester Competitive replacement of glial cells
WO2024196855A2 (en) 2023-03-17 2024-09-26 University Of Rochester Ribozyme-mediated rna assembly and expression
WO2024215989A1 (en) 2023-04-14 2024-10-17 Sotio Biotech Inc. ENGINEERED IMMUNE CELLS FOR TREATING CANCER IN COMBINATION WITH IL-2/IL-15 RECEPTOR βγ AGONISTS
WO2024215987A1 (en) 2023-04-14 2024-10-17 Sotio Biotech Inc. IMMUNE CELLS FOR TREATING CANCER IN COMBINATION WITH IL-15/IL-15Rα CONJUGATES

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987000201A1 (en) 1985-07-05 1987-01-15 Whitehead Institute For Biomedical Research Epithelial cells expressing foreign genetic material

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987000201A1 (en) 1985-07-05 1987-01-15 Whitehead Institute For Biomedical Research Epithelial cells expressing foreign genetic material

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
Anderson et al., Human Gene Therapy, 1, 331-362 (1990).
Anderson, "Gene Therapy: The Best of Times the Worst of Times," downloaded from the internet on Apr. 5, 2001 at 17:49 hours.
Anderson, John W., The Nation, Apr. 10, 1989, "Scrambling for BioTech Bucks", pp. 476-478. *
Anderson, The Nation, pp. 476-478 (Apr. 10, 1989).
Buden, The Scientist, Jan. 23, 1989, "Controversy Surrounds Gene Therapy Effort", pp. 1-3. *
Buden, The Scientist, pp. 1-3 (Jan. 23, 1989).
Cline, Pharmac. Ther., 29, 69-92 (1985).
Culver et al., J. Cell Biochem., Supp 12(B), 171 (1988).
Eppstein List of Prior Art References, submitted in Interference No. 104,711.
Gura, Science, 291, 1692-1697 (2001).
Kohn et al., J. Cell Biochem., Supp. 12(B), 178 (1988).
Marshall, Science, 288, 951-957 (2000).
Morgan et al., Science, 237, 1476-1479 (1987).
Morgan List of Prior Art Reference, submitted in Interference No. 104,712.
Palmer et al., Proc. Natl. Acad. Sci. USA, 84, 1055-1059 (1987).
Rosenberg et al., New Engl. J. Med., 323, 570-578 (1990).
Rosenberg, "Gene Therapy of Patients with Advanced Cancer Using Tumor Infiltrating Lymphocytes Transduced with the Gene Coding for Tumor Necrosis Factor," Jul. 6, 1990.
Selden et al., N. Eng. J. Med., 318(20): 1337-1338.
Selden, N. Eng. J. Med., vol. 318, No. 20, pp. 1337-1338. *
Siegel, Los Angeles Times, Dec. 13, 1987, "Egos, Prizes on the Line for Scientists", pp. 1, 37-40. *
Siegel, Los Angeles Times, Dec. 14, 1987, "Desire to be First Colors Gene Studies" pp. 1,22-25. *
Siegel, Los Angeles Times, pp. 1, 22-25 (Dec. 14, 1987).
Siegel, Los Angeles Times, pp. 1, 37-40 (Dec. 13, 1987).
Thompson, Time, Jun. 7, 1993 "The First Kids With New Gene" pp. 50-53. *
Thompson, Time, pp. 50-53 (Jun. 7, 1993).
Weiss, "New Rule for Gene Therapy Tests Proposed," downloaded from the internet on Jan. 18, 2001 at 06:52 hours.

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080175845A1 (en) * 2006-06-15 2008-07-24 Targeted Genetics Corporation Methods for treating target joints in inflammatory arthritis using AAV vectors encoding a TNF antagonist
US20080131415A1 (en) * 2006-11-30 2008-06-05 Riddell Stanley R Adoptive transfer of cd8 + t cell clones derived from central memory cells
US10400215B2 (en) 2006-11-30 2019-09-03 City of Home Adoptive transfer of CD8+ T cell clones derived from central memory cells
US10968431B2 (en) 2006-11-30 2021-04-06 City Of Hope Adoptive transfer of CD8+ T cell clones derived from central memory cells
US20090123441A1 (en) * 2007-10-08 2009-05-14 Intrexon Corporation Engineered Dendritic Cells and Uses for the Treatment of Cancer
US11111493B2 (en) 2018-03-15 2021-09-07 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
US11421228B2 (en) 2018-03-15 2022-08-23 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy
US11608500B2 (en) 2018-03-15 2023-03-21 KSQ Therapeutics, Inc. Gene-regulating compositions and methods for improved immunotherapy

Also Published As

Publication number Publication date
US5399346A (en) 1995-03-21

Similar Documents

Publication Publication Date Title
USRE39788E1 (en) Gene therapy
JP5312721B2 (en) CD19-specific redirecting immune cells
US6544787B1 (en) Non-myeloablative/lymphoablative conditioning regimen to induce patient anti-donor unresponsiveness in stem cell transplantation
JP2020502048A (en) Anti-BCMA CAR T cell composition
JPH07503455A (en) Lymphokine gene therapy for cancer
Rosenberg et al. Immunization of Cancer Patients Using Autologous Cancer Cells Modified by Insertion of the Gene for Tumor Necrosis Factor (National Institutes of Health)
CN111918963A (en) CD3 negative cell population expressing chemokine receptor and cell adhesion molecule and its use and preparation method
CA2977836A1 (en) Compositions and methods of treating cancer
JPH10510842A (en) How to increase hematopoietic cells
WO2022222846A1 (en) Chimeric antigen receptor targeting cd19, preparation method therefor and use thereof
JP7046016B2 (en) HERV-E reactive T cell receptor and usage
AU719930B2 (en) Prevention of graft-versus-host disease with T-cells including polynucleotides encoding negative selective markers
JPH11514209A (en) Highly efficient ex vivo transduction of hematopoietic stem cells with recombinant retrovirus preparations
JP3541950B2 (en) Use of modified TALL-104 cells to treat cancer and viral diseases
US5702702A (en) Modified cytotoxic tall cell line and compositions and methods for manufacture and use thereof as therapeutic reagents for cancer
KR20030081056A (en) Activated lymphocytes by HLA-coincided others for prevention and treatment of tumors, infection symptoms and autoimmune diseases, treatment method using the lymphocytes, formulation containing the lymphocytes as main component, method of manufacturing the formulation, and kit for preparing the formulation
WO1991018972A1 (en) Culturing bone marrow cells for adoptive immunotherapy
US20020034495A1 (en) Gene therapy
Rosenberg et al. TNF/TIL Human Gene Therapy Clinical Protocol: Original Protocol, April 23, 1990
Lotze et al. The Treatment of Patients with Melanoma Using Interleukin-2, Interleukin-4 and Tumor Infiltrating Lymphocytes. University of Pittsburgh
JP2002506980A (en) Novel complementary receptor-ligand pairs and adoptive immunotherapy using the same
Freedman et al. Use of a Retroviral Vector to Study the Trafficking Patterns of Purified Ovarian Tumor Infiltrating Lymphocytes (TIL) Used in Intraperitoneal Adoptive Immunotherapy of Ovarian Cancer Patients. A Pilot Study. University of Texas MD Anderson Cancer Center, Houston, Texas
WO1995029704A1 (en) Cell lines obtained by in vivo migration and by fusion with autoimmune cells
Favrot et al. Treatment of Patients with Advanced Cancer Using Tumor Infiltrating Lymphocytes Transduced with the Gene of Resistance to Neomycin. Centre Léon Bérard, Lyon, France
Klein Novel cellular therapies

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY