WO1997022349A1 - Methods for in vivo t cell activation by antigen-pulsed dendritic cells - Google Patents

Methods for in vivo t cell activation by antigen-pulsed dendritic cells Download PDF

Info

Publication number
WO1997022349A1
WO1997022349A1 PCT/US1996/019954 US9619954W WO9722349A1 WO 1997022349 A1 WO1997022349 A1 WO 1997022349A1 US 9619954 W US9619954 W US 9619954W WO 9722349 A1 WO9722349 A1 WO 9722349A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
antigen
cell
human
antigens
Prior art date
Application number
PCT/US1996/019954
Other languages
French (fr)
Inventor
Edgar G. Engleman
Ronald Levy
Frank Hsu
Claudia Benike
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Priority to AU11523/97A priority Critical patent/AU1152397A/en
Publication of WO1997022349A1 publication Critical patent/WO1997022349A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56988HIV or HTLV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • 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
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • 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
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • 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
    • A61K39/464499Undefined tumor antigens, e.g. tumor lysate or antigens targeted by cells isolated from tumor
    • 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
    • A61K39/464838Viral antigens
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • G01N33/686Anti-idiotype
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Abstract

The present invention relates to methods of using isolated human dendritic cells to present exogenous antigens for the induction of immune responses in vivo. In particular, it relates to the isolation of dendritic cells from human blood, exposing the cells to lymphoma-derived immunoglobulins or to proteins derived from human immunodeficiency virus (HIV) as antigens, and infusing the antigen-pulsed dendritic cells into patients to induce and/or augment an antigen-specific immune response. The methods of the invention described herein have a wide range of applications, including, but not limited to, the clinical use of antigen-pulsed dendritic cells as vaccines and/or immunotherapeutics against cancer and infectious agents such as viruses.

Description

METHODS FOR JN VIVO T CELL ACTIVATION BY ANTIGEN-PULSED DENDRITIC CELLS
1. INTRODUCTION
5 The present invention relates to methods of using isolated human dendritic cells to present exogenous antigens for the induction of immune responses m vi vo In particular, it relates to the isolation of dendritic cells from human blood, τ_0 exposing the cells to lymphoma-derived immunoglobulins or proteins derived from human immunodeficiency virus (HIV) as antigens, and infusing the antigen-pulsed dendritic cells into patients to induce and/or augment an antigen-specific immune response The methods of τ_5 the invention described herein have a wide range of applications, including but not limited to, the clinical use of antigen-pulsed dendritic cells as vaccines and/or immunotherapeutICS against cancel and infectious agents such as viruses.
20
2. BACKGROUND OF THE INVENTION
2.1. GENERATION OF AN IMMUNE RESPONSE
The introduction of a foreign antigen into 25 an individual elicits an immune response consisting of two a]oι components, the cellular and humoral immune responses, mediated by two functionally distinct populations of lymphocytes known as T and B cells, respectively The T cells may be further divided into 30 two subsets by function and phenotype A subset of " cells responds to antigen stimulation by producing lymphokines which "help" or activate various other cell types m the immune system. Another T cell subset is capable of developing into antigen-specific 35 cytotoxic effector cells, being able to directly kill antigen-positive target cells On the other hand, th<~ B cell response is primarily carried out by secretory proteins, antibodies. Antibodies function by neutralizing antigens or m conjunction with other 5 effector cells of the immune system m mediating antibody-dependent cellular cytotoxicity.
Helper T cells (TH) can be distinguished from classical cytotoxic T lymphocytes (CTL) and B cells by their cell surface expression of a
10 glycoprocem marker termed CD4. Although the mechanism by which CD4" TH function has not been fully elucidated, the existence of functionally distinct subsets within the CD4* T cell compartment has been reported ( osmann and Coffman, 1989, Ann. Rev. τ_5 Immunol. 7:145-173) . In the mouse, type 1 helper T cells (THI) produce mterleukm-2 (IL-2) and γ- mterferon (γ-IFN) upon activation, while type 2 helper T cells (TH2) produce IL-4 and IL-5 Based on the profile of lymphokme production, THI appear to be
20 involved m promoting the activation and proliferation of other T cell subsets including CTL, whereas TH2 specifically regulate B cell proliferation and differentiation, antibody synthesis, and antibody class switching Some CD4+ T cells, like CD8* CTL,
25 appear to be capable of cytotoxic effector function as well
A second T cell subpopulation is the classical CTL which express the CD8 surface marker Unlike most TH, these cells display cytolytic activity
30 upon direct contact with target cells, although they are also capable of producing certain lymphokines. In vi vo , CTL function is particularly important in situations where an antibody response alone is inadequate . There is a preponderance of experimental
35 evidence that CTL rather than B cells and their - 3 -
antibody products play a principal role in the defense against viral infections and cancer.
A salient feature of both T and B cell 5 responses is their exquisite specificity for the immunizing antigen; however, the mechanisms for antigen recognition differ between these two cell types. B cells recognize antigens by antibodies, either acting as cell surface receptors or as secreted _o proteins, which bind directly to antigens on a solid surface or in solution, whereas T cells only recognize antigens that have been processed or degraded into small fragments and presented on a solid phase such as the surface of antigen-presenting cells (APC) .
-*_5 Additionally, antigenic fragments must be presented to T cells m association with major histocompatibility complex (MHC) -encoded class I or class II molecules. The MHC refers to a cluster of genes that encode proteins with diverse immunological functions. In
20 man, the MHC is known as HLA. MHC class I gene products are found on all somatic cells, and they were originally discovered as targets of major transplantation rejection responses. Class II gene products are mostly expressed on cells of various
25 hematopoietic lineages, and they are involved in cell-cell interactions in the immune system. Most importantly, MHC-encoded proteins have been shown to function as receptors for processed antigenic fragments on the surface of APC (B orkman et al . ,
30 1987, Nature 329: 506-512) .
2.2. ANTIGEN PRESENTING CELLS
The presentation of antigens to T cells is carried out by specialized cell populations referred 35 to as APC. Typically, APC include macrophages/monocytes , B cells, and bone marrow- derived dendritic cells (DC) . DC are sometimes also referred to as "professional" APC. APC are capable of internalizing exogenous antigens, cleaving them into 5 smaller fragments in enzyme-rich vesicles, and coupling the fragments to MHC-encoded class I or class II products for expression on the cell surface (Goldberg and Rock, 1992, Nature 357:375-379) . Since APC express both MHC-encoded class I and class II 0 glycoproteins, they can present antigenic fragments to both CD4+ and CD8* T cells for the initiation of an immune response.
By definition, APC not only can present antigens to T cells with antigen-specific receptors, 5 but can provide all the signals necessary for T cell activation. Such signals are incompletely defined, but probably involve a variety of cell surface molecules as well as cytokines or growth factors . Further, the factors necessary for the activation of o naive or unprimed T cells may be different from those required for the re-activation of previously primed memory T cells. The ability of APC to both present antigens and deliver signals for T cell activation is commonly referred to as an accessory cell function. 5 Although monocytes and B cells have been shown to be competent APC, their antigen presenting capacities in vi tro appear to be limited to the re-activation of previously sensitized T cells. Hence, they are not capable of directly activating functionally naive or 0 unprimed T cell populations.
Although it had been known for a long time that APC could process and present antigens to T cells, it was not shown until relatively recently that small antigenic peptides could directly bind to MHC- 5 encoded molecules (Babbit et al . , 1985, Nature 317: 359; Townsend et al . , 1986, Ce 44: 959) . However, it is believed that normally, complex antigens are proteolytically processed into fragments inside the APC, and become physically associated with the 5 MHC-encoded proteins intracellularly prior to trafficking to the cell surface as complexes. Two distinct pathways for antigen presentation have been proposed (Braciale et al . , 1987, Immunol. Rev. 98: 95-114) . It was thought that exogenous antigens were
-j_0 taken up by APC, processed and presented by the exogenous pathway to class II-restricted CD4* T cells, while the endogenous pathway processed intracellularly synthesized proteins, such as products of viral genes in virally-infected cells, for association with MHC τ_5 class I proteins and presentation to CD8* CTL.
Although the two pathways in antigen processing and presentation may still be correct in some respects, the distinction is blurred in light of recent findings that exogenously added antigens may also be presented
20 to class I-restricted CTL (Moore et al . , 1988, Cell 54 : 777) .
The term "dendritic cell" refers to a diverse population of morphologically similar cell types found in a variety of lymphoid and non-lymphoid
25 tissues (Steinman, 1991, Ann. Rev. Immunol.
9:271-296) . These cells include lymphoid DC of the spleen, Langerhans cells of the epidermis, and veiled cells in the blood circulation. Although they are collectively classified as a group based on their
30 morphology, high levels of surface MHC class II expression, and absence of certain other surface markers expressed on T cells, B cells, monocytes, and natural killer cells, it is presently not known whether they derive from a common precursor or can all
35 function as APC in the same manner. Further, since the vast majority of published reports have utilized DC isolated from the mouse spleen, results from these studies may not necessarily correlate with the function of DC obtained from other tissue types. (Inaba et al . , 1987, J. Exp. Med. 166:182-194; Hengel et al., 1987 J. Immunol., 139:4196-4202; Kast et al . , 1988, J. Immunol., 140:3186-3193; Romani et al . , 1989, J. Exp. Med. 169:1169-1178; Macatonia et al . , 1989, J. Exp. Med. 169:1255-1264; Inaba et al . , 1990, J. Exp. Med. 172:631-6640) . For example, despite high levels of MHC class II expression, mouse epidermal Langerhans cells, unlike splenic DC, are not active APC in mixed leucocyte reaction (MLR) , unless cultured with granulocyte-macrophage colony stimulating factor (GM-CSF) (Witmer-Pock et al . , 1987, J. Exp. Med. 166:1484-1498; Heufler et al . , 1988, J. Exp. Med. 167:700-705) . Most human Langerhans cells express the CD1 and CD4 markers, while freshly isolated blood DC express CD4 weakly, but not CD1. On the other hand, cultured peripheral blood DC express CDlc, but not CD4. Additionally, it has not been established the extent to which the functional characteristics observed with mouse DC are applicable to human DC, especially the DC obtained from non-splenic tissues; in part, due to inherent differences between the human and murine immune systems.
In addition, the biological activity of human DC in vi vo has not been studied prior to the present invention. Although murine DC exposed to idiotype proteins of mouse lymphoma cells have been reported to induce tumor immunity in vivo, such success has not been demonstrated in human patients (Bohlen et al . , 1991, International Publication No. 091/13632) . There is no indication in the art that such an approach is suitable for clinical use. In fact, it is well known in the art that cancer is a complicated disease, and therapy performed in animal models does not adequately predict its likelihood of success m humans Similarly, acquired immunodeficiency syndrome (AIDS) caused by HIV does not have a correlative rodent animal model that is useful for assessing anti-HIV therapeutics Both the efficacy and toxicity of a therapeutic regimen for human disorders can only be properly examined in a clinical setting where all variable factors are present .
3. SUMMARY OF THE INVENTION
The present invention relates to the use of isolated and antigen-pulsed human DC as APC in vi vo to induce and/or augment antigen-specific immune responses It also relates to pharmaceutical composition containing such cells
The invention is based, m part, on the Applicants' discovery that human DC partially purified from human blood by sequential density gradient centrifugation function as potent APC m vi vo in patients with B cell ly phomas As shown m Example 6, infra , when isolated autologous DC are pulsed with immunoglobulin "custom made" from four patients' lymphoma cells and re-infused into the patients, ldiotype-specific T cell proliferative responses are detected Most importantly, one patient's tumor undergoes a complete regression over the course of such treatment. This patient also developed a tumor- specific CTL response. In addition, Example 7, infra , further demonstrates that HLA-matched donor DC pulsed with a recombinant HIV envelope protein, gpl60, induce HIV-spec fic T cell proliferative and cytotoxic responses, upon their infusion into an HIV- seropositive recipient A wide variety of uses is encompassed by the invention described herein, including but not limited to, the in vi vo administration of antigen-pulsed DC- as 5 vaccines for priming primary immune responses and/or as immunotherapeutics for augmenting secondary immune responses . Such responses may be induced against any antigen of interest, ranging from tumor antigens such as lymphoma idiotypes, p53 tumor suppressor protein, -j_0 human prostate carcinoma antigen known as prostatic acid phosphatase, melanoma antigens MAGE and MART-1, and oncogene products to infectious agents, including viruses such as HIV and its encoded antigenic products .
15
4. BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 Peripheral blood lymphocytes were isolated by "FICOLL/HYPAQUE" gradient centrifugation of patient's blood drawn at the indicated
20 point in her therapy regimen. Cells were plated at 4 x 105 cells/microtiter well with KLH or autologous immunoglobulin idiotype in Iscove's modified Dulbecco's medium (IMDM) with 1% human AB serum. After 3 days of
25 culture, the cells were split and fed with
IMDM containing 5% FCS and 30 units/ml of IL-2. After a total of 5 days at 37°C in 5% C02 atmosphere, the cells were pulsed for 16 hours with 1 μci/well 3H-thymidine . Data are
30 expressed as mean counts/minute of quadruplicate cultures.
FIG.
2A-2D Contiguous computerized tomographic scans of the chest were obtained prior to treatment with immunoglobulin-pulsed DC and two months
35 following the fourth immunization (optional booster) . FIG. 2A and B demonstrate the regression of paraaortic adenopathy. FIG. 2C and D demonstrate the regression of a paracardiac mass and paraaortic adenopathy. Prior to treatment, these nodal masses were documented to be slowly progressing.
FIG. 3 Peripheral blood mononuclear cells (105 per well) from an HIV-infected individual with a CD4 count of 480 CD4 cells per μl. were cultured with purified HIV-l MN gpl60 (Immuno AG, Vienna, Austria) for 7 days. [3H] -thymidine was added during the last 12 hours of culture and [3H] -thymidine incorporation determined using standard methods. The patient was infused monthly with DC pulsed with gpl60 (arrows) . Peripheral blood mononuclear cells were collected one week after each infusion, and at one and two weeks before the first infusion. Results are expressed as stimulation index (S.I.) ; counts per minute with antigen/counts per minute without antigen.
FIG. 4 Autologous Epstein Barr virus-transformed B cells were pulsed with vaccinia constructs that carried the HIV env gene or the β galactosidase gene and used as target cells. Peripheral blood mononuclear cells were isolated from the HIV-infected individual and then incubated with the target cells infected with vaccinia virus . Standard chromium release assays were performed one week after each infusion, and at one and two weeks before the first infusion. Patient was treated with gpl60 pulsed-DC at monthly intervals (arrows) . Background lysis of target cells pulsed with control vaccinia 5 virus expressing the β galactosidase gene was subtracted. Standard error of the mean never exceeded 5% of the mean.
5. DETAILED DESCRIPTION OF THE INVENTION lO Immunoglobulin or antibody molecules are polypeptides composed of heavy and light chains, which possess highly specific variable regions at their amino termini . The variable regions of heavy and light chains collectively form the unique antigen¬ ic recognition site of the immunoglobulin protein. These variable regions contain determinants that can themselves be recognized as antigens by the immune system, and they are referred to as idiotypes .
B-cell malignancies (B cell lyτnphomas, 2o leukemias, and myelomas) are products of clonal proliferation of tumor cells synthesizing a single immunoglobulin molecule (or monoclonal :, . ibody) with unique variable regions in the heavy and light chains. Since such tumors are monoclonal in origin, the 25 immunoglobulin expressed by all cells of a given tumor in a patient is identical, and can be distinguished from normal B cells by virtue of its unique idiotype. B-cell lymphomas are tumors of mature lymphocytes, which generally express ir
Figure imgf000012_0001
on their cell 30 surface. The idiotypic de .rminants of the surface immunoglobulin of a B-cell /mphoma can thus serve as a tumor-specific marker fo. he malignant clone since it is not expressed by any ^iher tissues in the body. Studies in animals as well as in humans have 35 demonstrated the usefulness of the immunoglobulin idiotype as a tumor-specific antigen and as a target for passive immunotherapy in vivo (Campbell et al . , 1990, J. Immunol. 145:1029; Campbell et al . , 1988, J. Immunol. 141:3227) . Active immunization against 5 idiotypic determinants on malignant B-cells has produced resistance to tumor growth in several animal models of syngeneic tumors, as well as specific antitumor therapy against established tumors (Campbell et al. , 1988, J. Immunol. 141:3227; George et al . ,
10 1988, J. Immunol. 141:2168) . Moreover, preclinical studies in non-human primates have demonstrated that optimal immunization with immunoglobulin derived from human lymphomas requires conjugation of the protein to a strongly immunogenic carrier protein such as
-j_5 keyhole-limpet hemocyanin (KLH) and emulsification in an adjuvant .
In a recent study, several lymphoma patients with minimal disease activity were repeatedly injected with idiotype protein coupled to KLH, emulsified in an
20 experimental adjuvant formulation (Kwak et al . , 1992, New Eng. J. Med. 327:1209) . Optimal immunization conditions required the use of both an immunogenic protein such as KLH and an adjuvant. In the absence of adjuvant, negligible idiotype-specific immune
25 responses were observed. However, several patients treated with the combination of KLH-idiotype and adjuvant demonstrated an anti-idiotypic immune response in vi tro and two patients with measurable disease experienced objective tumor regressions. The
30 use of an immunologic adjuvant was also associated with toxicity. Furthermore, an approved human adjuvant, alum, may not be effective in inducing anti- idiotypic responses.
The present invention, demonstrates that
35 patients with non-Hodgkin' s, B cell lymphoma who are not eligible for bone marrow transplantation can tolerate the infusion of autologous DC following m vi tro incubation of these cells with purified immunoglobulin idiotype isolated from autologous tumor cells . The patients are shown to generate immune responses to the tumor-associated ldiotypes and regression of tumor burden. Since the immunogenicity of an isolated antigen such as immunoglobulin may be poor, and its induction of successful immunity requires its conjugation to an immunogenic carrier and the use of an immunogenic adjuvant, antigen-pulsed DC may replace these two requirements m presenting antigens m vivo . Although the specific procedures and methods described herein are exemplified using DC isolated from human blood and lymphoma-derived immunoglobulin as antigens, they are merely illustrative for the practice of the invention Analogous procedures and techniques are equally applicable. In this connection, similarly prepared DC pulsed with an HIV-encoded protein are also shown to induce HIV-specific immune responses in vivo . Therefore, DC may be isolated from any source where they are found using variants of the procedure described herein, pulsed with any antigens or fragments thereof, and remfused nto pat- nts containing cells or tissues that express the antigens for the lmmunologic destruction of such cells and tissues .
5.1. ISOLATION OF HUMAN DENDRITIC CELLS
The present invention relates to an antigen presentation system using DC for the activation of T cells m vivo . Due to their presence m low numbers m most tissues, DC must first be isolated and enriched. Although DC are found m both lymphoid and non-lymphoid tissues, a natural and easily accessible source of DC in man is the peripheral blood, which contains an estimate of fewer than 1 DC per 100 white blood cells. The potency of the accessory cell function of DC in antigen presentation allows for the use of these cells in relatively small numbers when enriched, and absolute purity is not necessary for the generation of a T cell response in vivo . However, it is most preferable that a highly purified DC population (>90%) be used for in vivo administration.
DC may be isolated from a normal human or from a patient suffering from a disease. Additionally, such individuals may be treated with colony stimulating factors to increase their number of DC prior to isolation. For example, GM-CSF ("LEUKINE", Immunex Corporation, Seattle, WA) may be infused into an individual at 250 mcg/m2/day for several days up to three weeks intravenously prior to obtaining the peripheral blood mononuclear leukocytes (PBML) for the purification of DC. This procedure may increase the yield of DC for antigen pulsing and subsequent infusion.
Human DC may be isolated from any tissues where they reside, using a variety of separation methods. Example 6, infra, presents variants of such methods as illustrations for isolating DC from the human peripheral blood. This procedure is principally designed to avoid the exposure of DC to antigens such as fetal calf serum, sheep red blood cells and murine monoclonal antibodies which have been used in the separation of peripheral blood leukocytes. Since DC may be able to present such proteins to T cells, even in the absence of other exogen usly added antigens, conventional methods of DC isolation may lead to activation of T cells not specific for the antigens of interest, thus potentially masking the response sough . In accordance with this aspect of the invention, human PBML may be isolated from blood samples, particularly buffy coats or leukocytes prepared by apheresis, by "FICOLL HYPAQUE" gradient centrifugation followed by "PERCOLL" discontinuous centrifugation (Markowicz and Engleman, 1990, J. Clin. Invest. 85:955) followed by "METRIZAMIDE" (2- [3- Acetamido-5-N-methyl-acetamido-2 ,4,6- triiodobenzamido] -2-deoxy-D-glucose) or "NYCOPREP 1.068" NYCODENZ, N.N' -Bis (2, 3 dihydroxypropl) -5- [N- (2,3-dihydroxypropyl) acetamido] -2,4, 6-trilodo- isophtalamide discontinuous centrifugation. The high buoyant density (HD) fraction contains γδ and atβ-T cells, B cells, and NK cells, whereas DC are in the low buoyant density (LD) fraction of the "METRIZAMIDE" or "NYCOPREP 1.068" gradient. The LD fraction can then be subjected to a second "METRIZAMIDE" or "NYCOPREP 1.068" gradient to obtain a further enriched population of DC. DC may also be further enriched using additional protocols, depending on the level of purity required. Isolated DC can be pulsed immediately with any antigen of interest. Alternatively, DC may be isolated by procedures involving repetitive density gradient centrifugation, positive selection, negative selection, or a combination thereof. However, the above-mentioned density gradient methods are preferred because they do not contain xenogeneic proteins in the form of mouse antibodies or sheep red blood cells which may be internalized and presented by DC prior to the addition of an exogenous antigen of interest. Positive selection methods may utilize affinity chromatography with antibodies directed to DC surface markers. Positive selection does not necessarily require the use of antibodies that recognize DC- specific determinants. For example, B cells and monocytes may be depleted first from the DC-containing fraction after density gradient centrifugation, plastic adhesion, and Fc receptor panning, then an antibody to MHC-Class II antigen can be used to positively select for DC. Negative selection includes modifications of the protocol disclosed herein, supra. In essence, a DC-containing cell preparation may be reacted with one or more antibodies directed at cell surface antigens not expressed by DC for the removal of non-DC. Antibodies to any T cell, B cell, "monocyte, and granulocyte markers may be used. Examples of such antibodies include anti-CD3, anti-CD4, anti-CD5, and anti-CD8 specific for T cells; anti-CD12, anti-CD19 and anti-CD20 specific for B cells; anti-CD14 specific for monocytes; and anti-CD16, and anti-CD56 specific for natural killer cells (Becton Dickinson, San Jose, CA and Ortho
Diagnostics, NJ) . These antibodies may be applied in any combination repeatedly or in a sequential manner for the enrichment of DC. Upon binding to the antibodies, the cells may be removed by adsorption to a solid surface coated with an anti-mouse antibody, as the majority of monoclonal antibodies directed at cell surface markers are of mouse origin, or if the antibodies are conjugated with biotin, the antibody-bound cells can be removed by an avidin or streptavidin-coated surface; or if the antibodies are conjugated to magnetic beads, the cells expressing antigens recognized by the antibodies can be removed in a magnetic field (Harlow and Lane, 1988, Antibodies: A Laboratory Manual,, Cold Spring Harbor Laboratory Press) . 5.2. USE OF DENDRITIC CELLS AS ANTIGEN PRESENTING CELLS
The initiation of an immune response is mediated by APC, which process complex antigens into smaller fragments by enzymatic degradation, and present them in association with MHC-encoded molecules to T cells. Although macrophages/monocytes have been studied most extensively as APC, murine DC have been shown to also possess potent accessory cell function.
The present invention demonstrates that DC isolated from human blood present antigens for the activation of antigen-specific T cells in vi vo .
5.2.1. ANTIGENIC SYSTEMS FOR
PRESENTATION BY DENDRITIC CELLS
The potent accessory cell function of DC provides for an antigen presentation system for virtually any antigenic epitopes which T and B cells are capable of recognizing through their specific receptors. Example 6, infra , demonstrates that human DC can present immunoglobulin idiotypes as antigens to T cells in vivo . T cell activation is manifested by T cell proliferation in response to antigen. Example 7, infra , shows that similarly prepared DC can present an HIV-encoded protein to HLA-matched T cells in a recipient to induce both T cell proliferative and cytotoxic responses against HIV-infected target cells. Hence, DC are useful in vi vo in presenting antigens encoded by infectious agents such as viruses, microorganisms and their products, as well as tumor antigens expressed by cancer cells (Urban and Schreiber, 1992, Ann. Rev. Immunol. 10: 637-644) .
Infectious agents against which the present invention may be applicable include, but are not limited to, bacteria, parasites, fungi, and viruses. The multitudes of antigens encoded by these agents, which may be processed and presented by DC include but are not limited to, external surface proteins, and structural proteins including internal enzymes . For example, antigens encoded by any genes of the HIV genome including the env, gag, pol , nef , vif , rev, and tat genes may all be presented by DC in vi vo . In addition, peptide fragments which are degradative products of proteins within DC may also be directly added to DC during pulsing. A variety of other infectious agents including hepatitis B virus, hepatitis C virus, cytomegalovirus, herpes simplex virus, varicella zoster, Staphylococcal species and Mycobacterium species are encompassed within the scope of the invention.
In addition to immunoglobulin idiotypes as tumor-specific antigens, a large number of human tumor-associated antigens have been identified by monoclonal antibodies (Reisfeld and Cheresh, 1987, Adv. Immunol. 40: 323-377) . Although these cellular- antigens are selectively expressed in higher quantities by certain tumor cells, it has not been established that they naturally elicit an immune response in cancer patients or can be used effectively to induce such a response. Unlike animal tumor models in which tumor-reactive T and B cells can be induced through hyperimmunization with tumor cells or tumor antigens, intact human tumor cells or oncogenic proteins may not be easily tested in humans without an approved clinical protocol. Thus, most human studies have utilized lymphocytes obtained from cancer patients whose cells presumably have been exposed to antigens expressed by their autologous tumor cells in vivo . However, it has been shown in some systems that tumor development is accompanied by a down-regulation of tumor specific immune responsiveness mediated by suppressor cells, and if so, T cells isolated from cancer patients may have already come under the influence of such suppression m vivo so as to not function in a manner similar to that of T cells obtained from tumor-immune hosts. Moreover, these attempts to activate human tumor-reactive T cells have generally used monocytes as APC, which have been shown to be much less effective than DC, especially if the T cells have not been primed adequately in vi vo against the tumor antigens .
The antigen-pulsed DC described herein may be used as a cellular adjuvant for presenting tumor antigens in vivo . The potent accessory cell function of DC can present tumor antigens to T cells of cancer patients m vivo, whose immune response is apparently inadequate to eliminate the tumors m vivo . Whole tumor cells in viable or irradiated form, tumor membrane preparations, and tumor antigens purified from natural sources or expressed as recombinant products or chemically synthesized as peptides may be used to pulse DC m vi tro
Recently, oncogene products have been shown to be capable of inducing murine T cell activities. For example, oncogenic forms of the ras gene product p21, and the fusion product p210 of the bcr-abl gene induce T cell proliferative responses, when used to immunize mice (Peace et al , 1991, J Immunol. 146: 2059-2065; Chen et al . , 1992, Proc. Natl. Acad. Sci. USA 89 1468-1472) . Thus, oncogenic proteins which are different from their normal cellular counterparts as a result of ammo acid substitutions may possess new immunogenic determinants that are recognizable by T cells It is not necessary that such proteins be expressed naturally on the cell surface, as cytoplasmic and nuclear proteins may be processed, attached to MHC-encoded products intracellularly, and translocated to the cell surface in a complex form (Gould et al . , 1989, J. Exp. Med. 170: 1051-1056) . Since oncogene products are expressed in a variety of tumor types including colon cancer, leukemia and lymphoma, antigen-pulsed DC may be used to activate T cells in vi vo against such cancers. Other molecules which are associated with various types of cancer are also encompassed by the present invention, and they include, but are not limited to, HER-2/neu gene product (United States Patent No. 4,968,603) , estrogen receptor, milk fat globulin, p53 tumor suppressor protein (Levine, 1993, Annu. Rev. Biochem. 62:623) , mucin (Taylor-Papadimitriou, 1990, International Publication No. W090/05142) , telo erases, nuclear matrix proteins, MART-1, MAGE-1, MAGE-2, MAGE-3 (van der Bruggen et al . , 1991, Science 254:1643; Celis et al . , 1994, Proc. Natl. Acad. Sci. USA 91:2105) , GP100 (Bakker et al . , 1994, J. Exp. Med. 179:1005) , prostatic acid phosphatase, carcinoembryonic antigen, tyrosinase and papilloma viral antigens.
Bacterial, parasitic, fungal, viral, and tumor antigens of cellular or viral origin may be introduced to DC by addition to DC cultures followed by incubation, by the osmotic lysis of pinosomes after pinocytotic uptake (Moore et al . , 1988, Cell 54: 777- 785) , or by uptake in antigen-containing liposomes. Antigens may be used as purified naturally occurring whole polypeptides, purified recombinant whole polypeptides, whole organisms or cells in viable or dead forms, protein fragments generated by enzymatic digestion, or synthetic peptides produced by solid phase chemical methods (Creight-on, 1983, Protein Structures and Molecular Principles, W.H. Freeman and Co. , N.Y. pp 50-60) . The amount of antigens necessary for pulsing DC may vary depending on the nature, size, and purity of the molecules. In general, polypeptides may be used at 1-100 μg/ml, and small peptides at 1-50 μg/ml . Introduction by osmotic lysis of pinosomes requires larger amounts of proteins in the range of 200-500 μg/106 APC. Alternatively, exogenous genes encoding specific antigens of interest or expression vectors containing such genes or portions thereof may be incorporated into DC in expression vectors using conventional methods, including transfection, recombinant vaccinia viruses and retroviruses (Sambrook et al . , 1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press) . This approach causes the continual expression of integrated genes, leading to MHC occupancy by the gene products. Any of the aforementioned methods for introducing exogenous antigens into DC as well as any others commonly used by those skilled in the art are hereinafter collectively referred to as antigen pulsing of DC.
5.2.2. INDUCTION OF PRIMARY AND
SECONDARY T CELL RESPONSES JN VIVO
The present invention relates to a method of activating an immune response in a human patient to an antigen. The method includes the isolation of human DC, preferably from the peripheral blood, pulsing the cells with an antigen in vi tro , and then administering the antigen-pulsed cells into a patient whose cells or tissues express the antigen.
The ability of DC to process and retain antigenic fragments for several days permits their use as potent immunogens in vivo . DC may be pulsed with antigens according to the various methods described in Section 5.2.1, supra , washed, and administered in vivo as vaccines and/or immunotherapeutics for the elicitation or augmentation of a pre-existing but weak T cell response. In an effort to increase the number of isolated DC for antigen pulsing and subsequent in vivo administration, the isolated cells may be first expanded in number prior to incubation with antigens. It has been shown that GM-CSF and tumor necrosis factor-of induce the differentiation of human hematopoietic progenitor cells into DC (Caux et al . , 1992, Nature 360:258) . Thus, isolated DC may be expanded by treatment with such cytokines in culture.
Immunization with antigen-pulsed DC may increase both the magnitude and the specificity of a response. It may be desirable to repeat such immunizations at time intervals of days or weeks. The potency of DC as APC may alleviate the need of using conventional adjuvants to augment the response, although it does not preclude the use of adjuvants to further enhance immune reactivity. In addition, the antigen-pulsed DC may be administered in combination with cytokines that can maintain their number and activity for prolonged periods in vi vo . Such cytokines include colony stimulating factors such as GM-CSF and interleukins such as IL-12. The antigen-pulsed DC may be suspended in any known physiologic saline at a concentration sufficient to induce an immune response as detected by assays which measure T cell proliferation, T cell cytotoxicity, antibody production or reduction of the number of antigen-positive cells or tissues. A patient usually receives the total number of isolated DC pulsed with antigen. Depending on the yield of DC following each isolation procedure, a patient may be infused with several million up. to several hundred million DC. The cells may be infused into autologous, HLA-matched allogeneic or even HLA-mismatched allogeneic patients.
EXAMPLE: B-CELL LYMPHOMA REDUCTION INDUCED BY AUTOLOGOUS IMMUNOGLOBULIN IDIOTYPE-PULSED HUMAN DENDRITIC CELLS
6.1. MATERIALS AND METHODS
6.1.1. CELL SEPARATION
Human DC were obtained from buffy coats of lymphoma patients after leukophoresis . PBML were isolated by "FICOLL-HYPAQUE" gradient centrifugation (Boyum, 1968, Scand. J. Clin. Lab. Invest: 21:21-29) . In brief, a buffy coat was diluted with Dulbecco's PBS without divalent ions such as Ca2* or Mg2* (referred to as DPBS) up to 10 ml. 10 ml of "FICOLL" was gently underlaid into each tube and centrifuged at 1000 x g for 35 minutes at room temperature. The interface was collected and washed with DPBS three times.
To further separate various cell populations from the remaining mononuclear cell fraction, the preparation was further fractionated over a four layer discontinuous "PERCOLL" gradient (30%, 40%, 50.5% and 75%) (Pharmacia, Uppsala, Sweden) (Markowicz and Engleman, 1990, J. Clin. Invest. 85:955) . Original "PERCOLL" density was prepared at 1.130g/ml DPBS and 15 ml of a 50.5% "PERCOLL" solution was made and shaken in a conical polypropylene tube to create a foam on a surface of the "PERCOLL" solution. The tube was gently underlaid with about 6.5 ml of 75% "PERCOLL" . The tube was then slowly overlaid with 3 to 3.5 ml of 40% "PERCOLL" dropwise along the side of the tube which was being slowly rotated, followed by an overlay of 2.5ml of 30% "PERCOLL" in the same manner. The gradients were kept on ice for use within about 4 hours .
2.5-3X108 PBML in 5 to 10 ml of DPBS supplemented with 5% human serum were overlaid onto the four layer discontinuous "PERCOLL" gradient. Human serum was negative in licensed blood bank screening assays for antibodies to HIV, HTLV, hepatitis viruses, and treponema pal l idu . The serum was heat inactivated at 56°C for 1 hour and filtered through 0.2 μm filter prior to use. The cells were centrifuged at 1000 x g for 20-25 minutes at 4°C. The LD cells (monocytes) were collected from the interface over the "PERCOLL" 50.5% layer, whereas the lymphocytes and DC were collected from the interface between 75% and 50.5% layers. The collected cell fractions were diluted with DPBS at least 3 volumes and centrifuged at 1000 x g for 12 minutes at 4°C. The cells were washed twice with DPBS supplemented with 5% human serum at 400 x g for 5-6 minutes at 4°C. The HD cells (3-7 x 10β/50 ml of RPMI containing 10% pooled human serum) were then cultured overnight in teflon vessels at 37°C. Thereafter, the cultured cells were subjected to gradient centrifugation in "METRIZAMIDE" (15.5%) by overlaying the cells onto 10 ml of 15.5% (wt/vol) "METRIZAMIDE" (Sigma Chemical Co.) followed by centrifugation at 650 xg for 10 min at room temperature. This fraction was further depleted of contaminating monocytes by a solid phase absorption procedure for about 20 min. on human IgG-coated petri dishes. The IgG was commercial preparation tested and approved for intravenous human use . The DC were then enriched over a second "METRIZAMIDE" gradient (14%) . The HD cells from the first "METRIZAMIDE" gradient consisted of a mixture of atβ and γδ-T cells, B cells, and NK cells. The purity of the DC obtained using this procedure were 60-90%. Alternatively, the DC could be enriched 5 after overnight culture by centrifugation over a "NYCOPREP 1.068" discontinuous gradient (Nycomed Pharma AS, Oslo, Norway) . About 2.5 x 108 cells were suspended in 15-20 ml of a solution made up of 85% DPBS, 10% human serum and 5% EDTA. This was underlaid 0 sequentially with 4-5 ml of a solution of 50% human serum, 10% EDTA and 10% DPBS, followed by 4 ml of a solution of 75% "NYCOPREP 1.068", 24% DPBS and 1% human serum, followed by 8 ml of 100% "NYCOPREP 1.068". The cells were centrifuged at 400 xg for 13 5 minutes at room temperature. The interface and the pellet were collected and diluted with at least 3 volumes of DPBS containing 10% human serum, and centrifuged at 800 x g for 12 minutes at 20°C. The cells were washed twice with 10% human serum in RPMI o at room temperature, and DC occupied 30-40% of the total cell population. However, these cells could be further enriched by another round of "NYCOPREP" centrifugation to obtain a LD fraction of 80-90% DC. Alternatively, the LD cells after the first "NYCOPREP" 5 step could be negatively selected by incubation with antibody-coated petri dishes to remove CD3", CD14+, CD16* and CD20+ cells. The non-adherent cell population also contained 80-90% DC. However, density gradient centrifugation was preferred in order to 0 avoid the use of xenogeneic proteins in the form of antibodies against leukocyte markers. All procedures described herein could produce a yield of 1-2.5 x 106 cells from 400-500 ml of whole blood.
The purity of DC following each step of DC 5 enrichment was assessed by staining with an anti-HLA-DR (anti-MHC class II) antibody (CA141) conjugated to fluorescein, and phycoerythrin- conjugated anti-CD14 (anti-monocyte) .
Cytofluorographic analysis of the entire cell population was assessed by Fluorescence Activated Cell
Sorter. HLA-DR* but CD14" cells represented the DC population.
6.1.2. TUMOR IMMUNOGLOBULIN IDIOTYPE PRODUCTION
Ail patients had a histopathological diagnosis of non-Hodgkin' s lymphoma. Immunophenotypic studies of the tumors showed that they were of B-cell origin, with the surface expression of immunoglobulin molecules composed of both heavy and light chains in a monotypic pattern. Patients were selected who had low grade B cell lymphoma and were not eligible for bone marrow transplantation. The base-line studies used to evaluate clinical disease in all patients included complete physical examination, chest radiography, routine blood counts and chemistry tests, abdominal and pelvic CT scanning, with or without bipedal lymphangiography, which were all normally part of routine clinical care of such patients. All underwent re-staging during the course of the study according to the objective procedures used at base line.
In order to cause the B cell lymphoma to secrete their immunoglobulin for purification for use as antigens, a cell suspension obtained from the tumor biopsy specimen was washed in phosphate buffered saline (PBS) and mixed with the HAT sensitive heterohybridoma B5/K6H6 (B5) in a ratio of 1:4 or 1:5
(fusion partner:tumor cells) . B5 is a heterohybridoma that was produced by the fusion of the NS-1 mouse myeloma fusion partner with a human lymphoma cell.
The B5 clone has lost the ability to spontaneously secrete immunoglobulin. A subclone was identified which had retained the ability to secrete immunoglobulin when fused with human B-cells. This subclone was drug-marked so that it would not survive in HAT medium. Studies by Carroll et al . (1986, J. Immunol. Methods 89:61) show B5 to be a useful fusion partner for rescuing idiotype secretion from non- secreting human B-cell tumors. No mouse immunoglobulin is produced by B5 or hybridomas derived from i .
The mixture of tumor cells and fusion partner cells was washed 2 times in Hank's balanced salt solution (HBSS) . Supernatant was aspirated and the cell pellet was exposed to 40% polyethylene glycol (PEG) for 2 minutes. The PEG was diluted slowly by adding HBSS, and the cells were centrifuged. The cells were then resuspended in complete medium (RPMI Medium, 10% fetal bovine serum, glutamine 4 mM) to density of 2xlOG cells per ml, and 0.1 ml was then added to the wells of a microtiter plate. Twenty-four hours later after the cells had recovered from the PEG fusion, selection for rescue fusions was begun by changing the media to HAT media (complete media with the addition of hypoxanthine 10~4 M, thymidine 1.6 X 10~3 M and aminopterin 4 x 10"5 M) . The microtiter plates containing the cells were incubated for 10-20 days at 37°C in a humidified 5% C02 in air incubator. The supernatants were removed from the wells and the cells were fed with the medium described above but without the aminopterin (HT medium) .
An ELISA assay was used to determine which wells of the microtiter plate contained hybridomas which were secreting the rescued tumor immunoglobulin. The tumor biopsy specimen used for cell fusion contained a large predominance of malignant cells which expressed a single light chain type, either kappa or lambda. Wells secreting immunoglobulin with the heavy and light chain type corresponding to the known immunophenotype of the tumor specimen were selected and expanded.
Goat anti-human Ig heavy chain-specific reagents were diluted in 0.05 M sodium bicarbonate buffer (pH 9.5) . The 96-well microtiter plates were coated with this antibody in a volume of 50 microliters per well. The plates were incubated overnight at 4°C. The plates were then washed 5 times with normal saline containing 0.05% Triton X (wash buffer) . Nonspecific binding was blocked by incubating the wells with 5% non-fat milk in PBS for 1 hour and then washing 5 times with wash buffer.
Hybridoma supernatants were added (50 microliters per well) and incubated for 1 hour at room temperature. The plates were again washed and developed with 50 microliters per well of horseradish peroxidase conjugated to goat anti-human Ig light chain-specific reagents. The plates were incubated for 60 minutes at room temperature. Antibody reactivity was determined by added ABTS substrate and the optic density was read at 405 nm using a microtiter plate reader. As the hybridomas were grown, the supernatants were tested for immunoglobulin content using the ELISA described above. Dilutions of the supernatants were compared to a standard curve generated by using purified human immunoglobulin of the same isotype as that expressed on the patient's tumor. The idiotype protein was confirmed to be derived from the patient's lymphoma by cloning and comparing the DNA sequences of the variable region genes from both the lymphoma and rescue fusion. Hybridomas that constantly produced in excess of 2 micrograms per ml were expanded to large volumes (2000-5000 ml) . The cell density was allowed to increase until the media was fully spent and the supernatants were collected and pooled. 5 The rescued tumor idiotype protein was purified from bulk supernatant using affinity chromatography. For IgM class, a murine monoclonal anti-human IgM heavy chain-specific antibody was coupled to cyanogen bromide activated sepharose B4 or 0 an equivalent solid phase matrix at a concentration of 2-6 mg/ml of swollen beads (immunoadsorbent) . Rescued tumor idiotypes of the IgA class was purified using an immunoadsorbent column consisting of a goat or rabbit anti-human IgA-specific antibody similar]y coupled to 5 a solid phase matrix. Idiotypes of the IgG class were purified using a column of protein A coupled to a solid phase. The idiotype-containing supernatant was gravity flowed through the immunoadsorbent column. After absorption, the column was washed with 200 ml of o physiologic saline to remove the unbound protein. The idiotype was eluted with 0.1 M glycine-HCl buffer (pH 2.4) using absorption at 280 nm to monitor the protein effluent. The protein peak was collected and the immunoglobulin level measured using an ELISA technique 5 or absorption at 280 nm. The protein purity was determined by SDS-PAGE.
6.1.3. PREPARATION OF ANTIGENS
Keyhole Limpet Hemocyanin Megathura 0 crenulata (KLH) was obtained from Calbiochem, San Diego, California (Cat. #374805) . The KLH was supplied in a form containing more than 60% protein in BES buffer and magnesium sulfate. The protein purity, which was determined by the vendor, was greater than 5 90%. Prior to use, the KLH was dialyzed extensively against physiologic saline and then passed through a detoxigel column (Pierce Chemical Company, Rockford, Illinois, Cat. #20339) in PBS pH 7.4 or it was passed over a QAE Zeta Prep 15 disk (LKBm, Broma, Sweden, Cat. #224-202) using 25 mM Tris-HCl pH 8.0. A gradient of sodium chloride was used to remove the endotoxin which adhered to the column. Repeated passage of the KLH over the columns reduced the endotoxin to an acceptable level. The limulus amoebocyte lysate (LAL) assay was used to monitor endotoxin levels.
Purified idiotype specific protein and KLH were then dialyzed extensively against sterile physiologic saline. Each product was then concentrated or diluted to achieve a final concentration of 1 mg/ml in sodium chloride. The proteins were then sterile filtered through 0.45 μm filters. The final concentration was determined by measuring the absorption at 280 nm or performing an ELISA on a small, expendable aliquot of the final product . The final product was aseptically filled into sterile containers under a laminar flow hood. The vials were then frozen and stored at -20 C until needed. Sterility of the final product was confirmed by a standard 5-day bacterial culture assay performed by clinical laboratory.
6.1.4. PULSING OF DENDRITIC
CELLS WITH ANTIGENS
Enriched DC were resuspended in RPMI media at a concentration of approximately one million cells/ml . The DC were split into two independent cultures of five million cells into 24 well plates.
Each set of DC was then cultured with either sterile, purified idiotype immunoglobulin protein or KLH at a concentration of fifty micrograms/ml for 4-5 hours or overnight in a humidified 37° C incubator with 5% C02.
6.1.5. INJECTION OF ANTIGEN- PULSED DENDRITIC CELLS
DC pulsed with either idiotype protein or KLH were washed three times in physiologic, pyrogen- free, injection grade saline and resuspended into a volume of 100 ml in an intravenous injection administration bag. For each infusion, the patients received the total number of DC isolated from their own blood. Since each isolation yielded a different number of DC, each cell infusion contained between 5xl06 to lOOxlO6 antigen-pulsed DC. The patients were premedicated with acetaminophen and diphenhydramine, and received the transfusion of DC through a peripheral intravenous line or a central catheter (if available) over a period of 30-60 minutes. Vital signs were monitored continuously prior to and during the infusion. All therapy was monitored and supervised closely by a physician. Patients received similarly prepared antigen-pulsed DC for up to four times .
Any systemic reactions to the procedure were carefully noted. Systemic reactions were scored for temperature, blood pressure, and signs of bronchospasm, vasculitis, and/or immune complex formation. The risk of anaphylaxis was extremely low with the use of autologous cells.
6.1.6. ASSAY FOR IDIOTYPE-SPECIFIC PROLIFERATIVE RESPONSE
Proliferation assays were adapted from the techniques of Chain et al . (1987, J. Immunol. Methods 99:221) . Fresh PBML, prepared by "FICOLL-HYPAQUE" gradient centrifugation, were washed and plated at a concentration of 4xl05 cells per well in Iscove's modified Dulbecco's medium (IMDM) with 1 percent human AB serum (IMDM-1% AB) . KLH or autologous immunoglobulin idiotype at concentrations of 0 to 100 μg per milliliter in IMDM-1% AB preparation was added in quadruplicate. After 3 days, the cells were divided and fed with IMDM containing 5% FCS and 30 units/ml of IL-2. After the cells were incubated for a total of five days at 37°C in an atmosphere containing 5% carbon dioxide, they were pulsed for 16 to 20 hours with 3H-labeled thymidine (1 μCi per well) . Data are expressed as mean counts per minute of [3H] thymidine incorporation minus background.
6.1.7. ASSAY FOR ANTI-IDIOTYPIC ANTIBODIES
Microtiter plates were coated either with idiotype protein or with a panel of other immunoglobulins of the same isotype. Patient serum w s serially diluted. Pretreatment samples were used as a negative control. Binding of antibodies in the patient's serum to the test panel of immunoglobulins including the patient's idiotype was detected by goat anti-human light chain-HRP antibodies specific for the light chain opposite to the patient's idiotype or by a mixture of anti-human heavy chain isotype-specific-HRP antibodies that did not react with the patient's idiotype .
6.2. EXAMPLE
Four patients with follicular B cell lymphoma received a series of three or four infusions of immunoglobulin-pulsed DC. One patient entered the study with active disease manifested by easily discernible tumor masses on the x-ray computed tomography of the chest and abdomen. After providing informed consent the patient underwent leukapheresis at the Stanford University Blood Center on three occasions at four-week intervals. DC were isolated after each leukapheresis and equal aliquots (5 x 107 cells) were cultured overnight with either KliH or immunoglobulin idiotype which had been isolated from the patient's own tumor. The antigen-pulsed DC were extensively washed to remove any free immunoglobulin, suspended in isotonic saline and infused intravenously. Two weeks after each infusion, the patient received 0.5 mg of soluble idiotype protein in sterile physiologic saline subcutaneously as a boost, and a separate 0.5 mg boost of sterile KLH at a separate subcutaneous site. The cell infusions and boosts were well tolerated and two weeks following each infusion fresh lymphocytes isolated from peripheral blood were challenged in vi tro with graded doses of either KLH or idiotype, and their proliferative activity was assayed.
Prior to DC infusion, the patient failed to respond to either KLH or immunoglobulin idiotype; however, following DC infusion the patient's lymphocytes proliferated in response to both KLH and idiotype. As shown in FIG. 1, there was little or no response to these antigens prior to the infusion of antigen-pulsed DC, there were significant proliferative responses following the initial infusion, and the responses increased following the third infusion. Most importantly, the patient's tumor burden, which had been increasing for more than one year prior to infusion of DC was reduced by more than 50% when measured after the third infusion of antigen- pulsed DC and by 100% after the fourth treatment (FIG. 2A-D) . To date, this patient is in a clinical complete remission. In addition, when PBML of this patient were expanded in culture in the presence of the immunoglobulin, the cells were able to lyse autologous tumor hybridomas in chromium release assays, indicating the development of tumor-specific CTL in this patient .
The second patient had residual lymphoma in periaortic and parailiac regions prior to DC infusion. Despite a measurable anti-idiotypic proliferative response, this patient subsequently developed progressive disease four months after the primary series of infusions. Following a fourth immunization, the patient has remained stable and has not required further therapy. The third patient had equivocal radiographic findings of tumors, but showed evidence of disease in bone marrow and blood by the use of a sensitive assay involving polymerase-chain reaction for detecting tumor-specific immunoglobulin heavy chain gene rearrangement. The sensitivity of this technique allowed the detection of one tumor cell in 107 normal PBML. After completion of the first three immunizations, tumor was no longer detectable in biopsy samples by molecular analysis . The fourth patient had stage IV A follicular lymphoma with growing disease prior to DC therapy. This patient completed three cell infusions and has shown a minor response with regression of some peripheral lymph nodes. Furthermore, all patients developed tumor idiotype-specific proliferative responses during the course of DC infusion.
These results demonstrate the feasibility and safety of isolating DC, pulsing them with a tumor antigen, and infusing the cells into autologous patients at repeated intervals. They further suggest that such infusions are associated with the induction of clinically significant immune responses.
EXAMPLE: HIV-SPECIFIC T CELL RESPONSES INDUCED BY HIV ENVELOPE ANTIGEN-PULSED HUMAN DENDRITIC CELLS
7.1. MATERIALS AND METHODS
7.1.1. DENDRITIC CELL PREPARATION AND ANTIGEN PULSING
DC were enriched from the buffy coat of leukapheresed blood according to the procedure described in Section 6.1.1, supra . However, when the
HIV envelope antigen, gpl60, was used to pulse DC, the cells were incubated with recombinant protein for three times. Firstly, the cells obtained after
"PERCOLL" centrifugation were incubated in the teflon vessels at 107 cells/ml with 2μg/ml of HIV recombinant gpl60 (Immuno AG, Vienna, Austria) at 37°C overnight.
Thereafter, DC were further enriched by "METRIZAMIDE" centrifugation as described in Section 6.1.1, supra .
Then , 10-20X106 cells/ml were resuspended in RPMI/10% human serum plus 2 μg/ml of the same gpl60 antigen, and placed into teflon vessels at 37°C for 14-18 hours. Following incubation, the cells were washed by centrifugation in DPBS/5% human serum at room temperature for 10 minutes at 500xg. The pellet was resuspended in 15 ml of the same buffer and cell clusters allowed to disaggregate at room temperature for 15 min. The cells were then underlaid with 15 ml of 14% "METRIZAMIDE"/RMPI/10% human serum, centrifuged and washed three times in RPMI/10% human serum. About
106 cells were removed for phenotype analysis. Prior to infusion of antigen-pulsed DC in vivo, 10-20X106 cells/ml in RPMI/10% human serum were again incubated with 50 μg/ml of gpl60 in teflon vessels overnight. In cases where DC were pulsed with HIV peptides, the cells were pulsed at a concentration of 5μg of peptides with 106 cells/ml. Also, only one pulsing step was used for peptides, which were presumed to not require further processing. Several HLA-A2.1-restricted HIV-l peptides were used. The amino acid sequences, HLA binding affinities and CTL- inducing activities of these peptides are given in Table 1 below.
Table 1
Amino Acid HLA 2.1 CTL
Antigen Sequence Residues Affinity* Conservancy activity'
Env KLTPLCVTL 120-128 0.16 97% +
SLLNATDIAV 814-823 0.11 57% +
Gag SLYNTVATL 77-85 +++ 64% +
VLAEAMSQV 362-370 0.11 96% +
Pol LLWKGEGAW 956-964 0.04 100% +
I KEPVHGV 464-472 0.02 92% +
* Ratio to IC50 Standard Peptide
** CTL actively demonstrated against peptide-coated target cells
7.2. CELL PREPARATION FOR INFUSION
After the final antigen pulsing step, the enriched DC preparation was collected the next morning and centrifuged at room temperature for 10 minutes at 500 xg. The pellet was resuspended in DPBS and cell viability was examined by trypan blue exclusion. After two additional washing steps, the cells were placed into a 100 ml injection grade saline administration bag. Using a 10ml syringe and a 19 gauge needle, 8-10ml of saline was removed from the bag for use in resuspending the cell pellet. Thereafter, the cell suspension was brought up into the syringe and injected into the bag, followed by injection of 5 ml of previously frozen autologous serum.
7.3. EXAMPLE Asymptomatic HIV-seropositive patients generally exhibit a vigorous T cell response against HIV. However, such a response subsides as clinical symptoms of the disease begin to develop. In order to augment the T cell immunity against HIV, an HIV- seropositive patient was infused with DC prepared from an HLA-matched sibling that had been pulsed with HIV gpl60 in vi tro .
Donor DC cells were enriched from peripheral blood and pulsed with HIV-encoded gpl60 and administered into the patient three times at one month intervals. During the course of treatment, PBML were obtained from the patient and assayed for T cell proliferative and cytotoxic responses against HIV- infected cells. FIG. 3 demonstrates that shortly after the first cell infusion, T cells obtained from the patient proliferated in response to HIV gpl60 in culture. The T cell proliferative activity continued to rise for two weeks and was maintained at the same level for over six weeks after the first infusion.
Additionally, T cells obtained from the same patient also exhibited HIV-specific CTL (FIG. 4) . Therefore, the administration of HIV antigen-pulsed DC induced both HIV-specific CD4* and CD8* T cells. Since a delay of clinical symptoms of HIV correlates with a detectable HIV-specific T cell response in an HIV- infected individual, an augmentation of such T cell responses will likely contribute to the control of the spread of HIV, thereby delaying HIV clinical latency, and prolonging of survival of infected individuals. The present invention is not to be limited in scope by the exemplified embodiments, which are intended as illustrations of individual aspects of the invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims . All publications cited herein are incorporated by reference in their entirety.

Claims

WHAT IS CLAIMED IS:
1. A method of activating an immune response m a human patient to an antigen comprising:
(a) isolating human dendritic cells;
(b) pulsing the cells with an antigen; and
(c) administering the cells pulsed with the antigen into the human patient .
2. The method of Claim 1 m which the dendritic cells are isolated from human peripheral blood.
3. The method of Claim 1 m which the immune response is mediated by T cells.
4. The method of Claim 1 in which the immune response is mediated by B cells or their secreted antibodies .
5. The method of Claim 1 which the antigen is produced by tumor cells.
6. The method of Claim 5 in which the antigen is produced by B-cell tumor cells
7. The method of Claim 6 in which the antigen is an immunoglobulin.
8. The method of Claim 1 m which the antigen is a whole microorganism.
9. The method of Claim 1 m which the antigen is a virus
10. The method of Claim 1 in which the antigen is a polypeptide.
5 11. The method of Claim 10 in which the polypeptide is gpl60 encoded by HIV.
12. The method of Claim 1 in which the antigen is a peptide. 0
13. A pharmaceutical composition comprising isolated human dendritic cells which have been pulsed with an antigen in vi tro .
5 14. The pharmaceutical composition of Claim 13 in which the dendritic cells are isolated from human peripheral blood.
15. The pharmaceutical composition of Claim 13 in o which the antigen is produced by tumor cells.
16. The pharmaceutical composition of Claim 15 in which the antigen is produced by B-cell tumor cells. 5
17. The pharmaceutical composition of Claim 16 in which the antigen is an immunoglobulin.
18. The pharmaceutical composition of Claim 13 in 0 which the antigen is a microorganism.
19. The pharmaceutical composition of Claim 13 in which the antigen is a virus.
5 20. The pharmaceutical composition of Claim 13 in which the antigen is a polypeptide.
21. The pharmaceutical composition of Claim 20 in which the polypeptide is gpl60 encoded by HIV.
22. The pharmaceutical composition of Claim 13 in which the antigen is a peptide.
23. A method for treating cancer in a human cancer patient, comprising:
(a) isolating human dendritic cells;
(b) pulsing the cells with an antigen produced by cancer cells; and
(c) administering the cells pulsed with the antigen into the human patient having the cancer cells.
24. The method of claim 23 in which the cancer cells are lymphoma cells.
PCT/US1996/019954 1995-12-20 1996-12-17 Methods for in vivo t cell activation by antigen-pulsed dendritic cells WO1997022349A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU11523/97A AU1152397A (en) 1995-12-20 1996-12-17 Methods for in vivo t cell activation by antigen-pulsed dendritic cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57543295A 1995-12-20 1995-12-20
US08/575,432 1995-12-20

Publications (1)

Publication Number Publication Date
WO1997022349A1 true WO1997022349A1 (en) 1997-06-26

Family

ID=24300304

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/019954 WO1997022349A1 (en) 1995-12-20 1996-12-17 Methods for in vivo t cell activation by antigen-pulsed dendritic cells

Country Status (2)

Country Link
AU (1) AU1152397A (en)
WO (1) WO1997022349A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999050392A1 (en) 1998-03-31 1999-10-07 Geron Corporation Methods and compositions for eliciting an immune response to a telomerase antigen
WO1999053949A2 (en) * 1998-04-23 1999-10-28 Arch Development Corporation Combination of antigen pulsed apcs and interleukin 12 for tumor and viral therapy
US6500641B1 (en) 1999-05-06 2002-12-31 Wake Forest University School Of Medicine Compositions and methods for identifying antigens which elicit an immune response
US6586243B2 (en) * 1997-02-07 2003-07-01 Cem Cezayirli Directed maturation of CD34 negative stem cells to programmable antigen presenting cells
US6716422B1 (en) 1998-04-23 2004-04-06 Arch Development Corporation Vaccine adjuvants for immunotherapy of melanoma
WO2005009463A2 (en) * 2003-07-24 2005-02-03 Cerus Corporation Antigen-presenting cell vaccines and methods of use thereof
EP1305402B1 (en) * 2000-07-31 2006-03-29 Bio Merieux Lipo-viro-particle complexes, preparation method and uses
US7402307B2 (en) 1998-03-31 2008-07-22 Geron Corporation Method for identifying and killing cancer cells
US7563880B2 (en) 1999-11-30 2009-07-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US7598226B2 (en) 1998-12-28 2009-10-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US7691393B2 (en) 2003-02-06 2010-04-06 Anza Therapeutics, Inc. Listeria attenuated for entry into non-phagocytic cells, vaccines comprising the Listeria, and methods of use thereof
US7833775B2 (en) 2003-02-06 2010-11-16 Aduro Biotech Modified free-living microbes, vaccine compositions and methods of use thereof
US7842289B2 (en) 2003-12-24 2010-11-30 Aduro Biotech Recombinant nucleic acid molecules, expression cassettes, and bacteria, and methods of use thereof
US7855271B2 (en) 1998-12-28 2010-12-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US20110027244A1 (en) * 2009-07-28 2011-02-03 Promising Future, Llc Compositions and methods of preparing alloreactive cytotoxic T cells
US7977095B2 (en) * 2003-05-08 2011-07-12 Life Technologies Corporation Generation and isolation of antigen-specific T cells
US7981673B2 (en) * 2001-08-08 2011-07-19 Biociones (Propietary) Limited Process for the maturation of dendritic cells and a vaccine
US8414899B2 (en) 2006-04-11 2013-04-09 Yeda Research And Development Co. Ltd. Vaccines comprising multimeric HSP60 peptide carriers
WO2017181163A2 (en) 2016-04-16 2017-10-19 Oncocyte Corporation Methods and compositions for detection and diagnosis of breast cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991013632A1 (en) * 1990-03-14 1991-09-19 The Immune Response Corporation Idiotypic vaccination against b cell lymphoma
WO1995034638A1 (en) * 1994-06-14 1995-12-21 The Board Of Trustees Of Leland Stanford Junior University Methods for in vivo t cell activation by antigen-pulsed dendritic cells

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991013632A1 (en) * 1990-03-14 1991-09-19 The Immune Response Corporation Idiotypic vaccination against b cell lymphoma
WO1995034638A1 (en) * 1994-06-14 1995-12-21 The Board Of Trustees Of Leland Stanford Junior University Methods for in vivo t cell activation by antigen-pulsed dendritic cells

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
IMMUNOLOGY LETTERS, 1991, Vol. 30, No. 2, KNIGHT et al., "Stimulatory and Suppressive Effects of Infection of Dendritic Cells With HIV-1", pages 213-218. *
IMMUNOLOGY, 1987, Vol. 62, AUSTYN, "Lymphoid Dendritic Cells", pages 161-170. *
IMMUNOLOGY, 1989, Vol. 67, MACATONIA et al., "Suppression of Immune Responses by Dendritic Cells Infected With HIV", pages 285-289. *
IMMUNOLOGY, 1990, Vol. 71, MACATONIA et al., "Dendritic Cell Infection, Depletion and Dysfunction in HIV-infected Individuals", pages 38-45. *
IMMUNOLOGY, 1991, Vol. 74, MACATONIA et al., "Primary Proliferative and Cytotoxic T-Cell Responses to HIV Induced in Vitro by Human Dendritic Cells", pages 399-406. *
IMMUNOLOGY, 1992, Vol. 76, PANCHOLI et al., "Dendritic Cells Efficiently Immunoselect Mycobacterial-reactive T Cells in Human Blood, Including Clonable Antigen-reactive Precursors", pages 217-224. *
INTERN. REV. IMMUNOL., 1990, Vol. 6, INABA et al., "Dendritic Cells as Antigen Presenting Cells in Vivo", pages 197-206. *
INTERNATIONAL IMMUNOLOGY, 1993, Vol. 5, No. 8, TAKAHASHI et al., "Induction of CD8+ cytotoxic T Lymphocytes by Immunization With Syngeneic Irradiated HIV-1 Envelope Derived Peptide-pulsed Dendritic Cells", pages 849-857. *
J. CLIN. INVESTIGATION, March 1990, Vol. 85, MARKOWICZ et al., "Granulocyte-macrophage Colony-stimulating Factor Promotes Differentiation and Survival of Human Peripheral Blood Dendritic Cells in Vitro", pages 955-961. *
JOURNAL OF EXPERIMENTAL MEDICINE, April 1989, Vol. 169, MACATONIA et al., "Primary Stimulation by Dendritic Cells Induces Antiviral Proliferative and Cytotoxic T Cell Responses in Vitro", pages 1255-1264. *
JOURNAL OF EXPERIMENTAL MEDICINE, August 1990, Vol. 172, INABA et al., "Dendritic Cells Pulsed With Protein Antigens in Vitro Can Prime Antigen-specific, MHC-restricted T Cells in Situ", pages 631-640. *
JOURNAL OF EXPERIMENTAL MEDICINE, January 1992, Vol. 175, BHARDWAJ et al., "Dendritic Cells are Potent Antigen-presenting Cells for Microbial Superantigens", pages 267-273. *
JOURNAL OF EXPERIMENTAL MEDICINE, January 1992, Vol. 175, SORNASSE et al., "Antigen-pulsed Dendritic Cells Can Efficiently Induce an Antibody Response in Vivo", pages 15-21. *
JOURNAL OF IMMUNOLOGY, March 1985, Vol. 134, No. 3, KAYE et al., "Nonphagocytic Dendritic Cells are Effective Accessory Cells for Anti-mycobacterial Responses in Vitro", pages 1930-1934. *
NATURE MEDICINE, January 1996, Vol. 2, No. 1, HSU et al., "Vaccination of Patients With B-Cell Lymphoma Using Autologous Antigen-pulsed Dendritic Cells", pages 52-58. *
NEW ENGLAND JOURNAL OF MEDICINE, 22 October 1992, Vol. 327, No. 17, KWAK et al., "Induction of Immune Responses in Patients With B-cell Lymphoma Against the Surface-immunoglobulin Idiotype Expressed by Their Tumors", pages 1209-1215. *

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6586243B2 (en) * 1997-02-07 2003-07-01 Cem Cezayirli Directed maturation of CD34 negative stem cells to programmable antigen presenting cells
JP2007137889A (en) * 1998-03-31 2007-06-07 Geron Corp Method and composition for inducing immunological response to telomerase antigen
EP1068296A1 (en) * 1998-03-31 2001-01-17 Geron Corporation Methods and compositions for eliciting an immune response to a telomerase antigen
US6440735B1 (en) 1998-03-31 2002-08-27 Geron Corporation Dendritic cell vaccine containing telomerase reverse transcriptase for the treament of cancer
WO1999050392A1 (en) 1998-03-31 1999-10-07 Geron Corporation Methods and compositions for eliciting an immune response to a telomerase antigen
EP1068296A4 (en) * 1998-03-31 2003-06-04 Geron Corp Methods and compositions for eliciting an immune response to a telomerase antigen
US7824849B2 (en) 1998-03-31 2010-11-02 Geron Corporation Cellular telomerase vaccine and its use for treating cancer
US7402307B2 (en) 1998-03-31 2008-07-22 Geron Corporation Method for identifying and killing cancer cells
WO1999053949A2 (en) * 1998-04-23 1999-10-28 Arch Development Corporation Combination of antigen pulsed apcs and interleukin 12 for tumor and viral therapy
WO1999053949A3 (en) * 1998-04-23 2000-03-09 Arch Dev Corp Combination of antigen pulsed apcs and interleukin 12 for tumor and viral therapy
AU767208B2 (en) * 1998-04-23 2003-11-06 Arch Development Corporation Combination of antigen pulsed APCS and interleukin 12 for tumor and viral therapy
US6716422B1 (en) 1998-04-23 2004-04-06 Arch Development Corporation Vaccine adjuvants for immunotherapy of melanoma
US7855271B2 (en) 1998-12-28 2010-12-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US8182823B2 (en) 1998-12-28 2012-05-22 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US7598226B2 (en) 1998-12-28 2009-10-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US6716623B2 (en) 1999-05-06 2004-04-06 Wake Forest University School Of Medicine Compositions and methods for identifying antigens which elicit an immune response
US6500641B1 (en) 1999-05-06 2002-12-31 Wake Forest University School Of Medicine Compositions and methods for identifying antigens which elicit an immune response
US7749752B2 (en) 1999-05-06 2010-07-06 Wake Forest University Health Sciences Compositions and methods for identifying antigens which elicit an immune response
US7563880B2 (en) 1999-11-30 2009-07-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
EP1305402B1 (en) * 2000-07-31 2006-03-29 Bio Merieux Lipo-viro-particle complexes, preparation method and uses
US7981673B2 (en) * 2001-08-08 2011-07-19 Biociones (Propietary) Limited Process for the maturation of dendritic cells and a vaccine
US7691393B2 (en) 2003-02-06 2010-04-06 Anza Therapeutics, Inc. Listeria attenuated for entry into non-phagocytic cells, vaccines comprising the Listeria, and methods of use thereof
US7833775B2 (en) 2003-02-06 2010-11-16 Aduro Biotech Modified free-living microbes, vaccine compositions and methods of use thereof
US7695725B2 (en) 2003-02-06 2010-04-13 Aduro Biotech Modified free-living microbes, vaccine compositions and methods of use thereof
US7927606B2 (en) 2003-02-06 2011-04-19 Aduro Biotech Modified free-living microbes, vaccine compositions and methods of use thereof
US7977095B2 (en) * 2003-05-08 2011-07-12 Life Technologies Corporation Generation and isolation of antigen-specific T cells
WO2005009463A2 (en) * 2003-07-24 2005-02-03 Cerus Corporation Antigen-presenting cell vaccines and methods of use thereof
WO2005009463A3 (en) * 2003-07-24 2005-06-02 Cerus Corp Antigen-presenting cell vaccines and methods of use thereof
US7842289B2 (en) 2003-12-24 2010-11-30 Aduro Biotech Recombinant nucleic acid molecules, expression cassettes, and bacteria, and methods of use thereof
US8414899B2 (en) 2006-04-11 2013-04-09 Yeda Research And Development Co. Ltd. Vaccines comprising multimeric HSP60 peptide carriers
US20110027244A1 (en) * 2009-07-28 2011-02-03 Promising Future, Llc Compositions and methods of preparing alloreactive cytotoxic T cells
US8586359B2 (en) * 2009-07-28 2013-11-19 Promising Furture, LLC Compositions and methods of preparing alloreactive cytotoxic T cells
WO2017181163A2 (en) 2016-04-16 2017-10-19 Oncocyte Corporation Methods and compositions for detection and diagnosis of breast cancer

Also Published As

Publication number Publication date
AU1152397A (en) 1997-07-14

Similar Documents

Publication Publication Date Title
EP0765386B1 (en) Methods for in vivo t cell activation by antigen-pulsed dendritic cells
US6399054B1 (en) Method for the production of activated marked tumor-specific T cells and use thereof in treatment of tumors
WO1997022349A1 (en) Methods for in vivo t cell activation by antigen-pulsed dendritic cells
EP0726941B1 (en) Methods for (ex vivo) therapy using peptide-loaded antigen presenting cells for the activation of ctl
US6821778B1 (en) Methods for using dendritic cells to activate gamma/delta-T cell receptor-positive T cells
JP2022173331A (en) Use of CART19 to deplete normal B cells to induce tolerance
US11903967B2 (en) Method of preparing T cells with increased activity
JP6230208B2 (en) Stimulation of anti-tumor immunity using dendritic cell / tumor cell fusions and anti-CD3 / CD28
WO1994002156A1 (en) Methods for using dendritic cells to activate t cells
JP5184732B2 (en) Compositions and methods of use of monoclonal and polyclonal antibodies specific for T cell subpopulations
JP2011504101A5 (en)
Anderson et al. Identification of MAGE-C1 (CT-7) epitopes for T-cell therapy of multiple myeloma
AU2020213119A1 (en) Compositions and methods for targeting mutant RAS
MXPA03007503A (en) A cell therapy method for the treatment of tumors.
Ellenhorn et al. Mechanism of tumor rejection in anti-CD3 monoclonal antibody-treated mice.
Toujas et al. Human monocyte‐derived macrophages and dendritic cells are comparably effective in vitro in presenting HLA class I‐restricted exogenous peptides
Khalaf et al. In vitro generation of cytotoxic T cells with potential for adoptive tumor immunotherapy of multiple myeloma
AU2017240584A1 (en) Dendritic cell- extracellular vesicle fusions and methods of using same
EP3277292A1 (en) Compositions and methods of treating multiple myeloma
JP5054875B2 (en) Cytotoxic T lymphocytes activated by dendritic cell hybrids
US7405077B2 (en) Sperm protein 17 for the diagnosis and treatment of cancer
AU6551799A (en) Methods for in vivo cell activation by antigen-pulsed dendritic cells
JP2006187215A (en) Method for creating antibody using gene-modified type dendritic cell given by vector
CN116769719A (en) TCR-T cell, preparation method thereof, pharmaceutical composition and application
Patterson Graft-versus-myeloid leukemia responses in murine bone marrow transplantations

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE FI GE HU IL IS JP KG KP KR KZ LC LK LR LS LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK TJ TM TR TT UA UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97522931

Format of ref document f/p: F

122 Ep: pct application non-entry in european phase