Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
2.
Blood Cancer J ; 3: e145, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-24013666

ABSTRACT

The ability to target myeloid leukemia with immunotherapy would represent a significant therapeutic advance. We report here immunological analysis of clinical trials of primary and secondary vaccination with K562/GM-CSF immunotherapy in adult chronic phase chronic myeloid leukemia patients (CML-CP) with suboptimal responses to imatinib mesylate. Using serological analysis of recombinant cDNA expression libraries of K562 with autologous vaccinated patient serum, we have identified 12 novel chronic myeloid leukemia-associated antigens (LAAs). We show that clinical responses following K562/GM-CSF vaccination are associated with induction of high-titer antibody responses to multiple LAAs. We observe markedly discordant patterns of baseline and induced antibody responses in these identically vaccinated patients. No single antigen was recognized in all responses to vaccination. We demonstrate that an additional 'booster' vaccination series can be given safely to those with inadequate responses to initial vaccination, and is associated with more frequent induction of IgG responses to antigens overexpressed in K562 vaccine compared with primary CML-CP. Finally, those with induced immune responses to the same LAAs often shared HLA subtypes and patients with clinical responses following vaccination recognized a partially shared but non-identical spectrum of antigens; both findings have potentially significant implications for cancer vaccine immunotherapy.

4.
Cancer Immunol Immunother ; 60(1): 15-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21080166

ABSTRACT

Many assays to evaluate the nature, breadth, and quality of antigen-specific T cell responses are currently applied in human medicine. In most cases, assay-related protocols are developed on an individual laboratory basis, resulting in a large number of different protocols being applied worldwide. Together with the inherent complexity of cellular assays, this leads to unnecessary limitations in the ability to compare results generated across institutions. Over the past few years a number of critical assay parameters have been identified which influence test performance irrespective of protocol, material, and reagents used. Describing these critical factors as an integral part of any published report will both facilitate the comparison of data generated across institutions and lead to improvements in the assays themselves. To this end, the Minimal Information About T Cell Assays (MIATA) project was initiated. The objective of MIATA is to achieve a broad consensus on which T cell assay parameters should be reported in scientific publications and to propose a mechanism for reporting these in a systematic manner. To add maximum value for the scientific community, a step-wise, open, and field-spanning approach has been taken to achieve technical precision, user-friendliness, adequate incorporation of concerns, and high acceptance among peers. Here, we describe the past, present, and future perspectives of the MIATA project. We suggest that the approach taken can be generically applied to projects in which a broad consensus has to be reached among scientists working in fragmented fields, such as immunology. An additional objective of this undertaking is to engage the broader scientific community to comment on MIATA and to become an active participant in the project.


Subject(s)
Consensus , Neoplasms/immunology , T-Lymphocytes/immunology , Allergy and Immunology/trends , Humans , Immunologic Techniques/standards , Monitoring, Physiologic/standards , Practice Guidelines as Topic , Program Development , Research Design
5.
Blood ; 98(4): 1070-7, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11493453

ABSTRACT

Tumor antigen-specific T-cell tolerance may limit the efficacy of therapeutic cancer vaccines. Direct presentation of antigens by tumor cells incapable of providing adequate costimulation to tumor-specific T cells has been suggested as the basis for this unresponsiveness. Using parent-into-F1 bone marrow (BM) chimeras, this study unambiguously demonstrates that the induction of this tolerant state requires T-cell recognition of tumor antigen presented by BM-derived antigen-presenting cells (APCs), not tumor cells themselves. In the absence of host APC presentation, tumor-specific T cells remained functional, even in the setting of antigen expressed by B-cell lymphomas residing in secondary lymphoid tissues. The intrinsic APC capacity of tumor cells has therefore little influence over T-cell priming versus tolerance, a decision that is regulated at the level of host APCs. (Blood. 2001;98:1070-1077)


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Lymphoma, B-Cell/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/pharmacology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/transplantation , Clone Cells/immunology , Disease Progression , Hemagglutinins, Viral/immunology , Immune Tolerance/drug effects , Lymphocyte Activation/immunology , Lymphoma, B-Cell/pathology , Male , Mice , Mice, SCID , Mice, Transgenic , Models, Biological , Neoplasm Transplantation , Transplantation Chimera , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/transplantation
6.
J Immunol ; 166(7): 4773-9, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11254740

ABSTRACT

We tested the hypothesis that APCs genetically engineered to present an Ag and to express Fas ligand (FasL) simultaneously can target and eliminate Ag-specific T cells. Transgenic T cells specific for influenza hemagglutinin (HA) were used as targets. We prepared recombinant vaccinia virus vectors (VVV) to transfer the gene constructs individually or simultaneously into APCs. We prevented unwanted viral replication by attenuating the VVVs with psoralen-UV light treatment. For presentation of the HA Ag, APCs were transduced with cDNA for HA flanked by sequences of the lysosome-associated membrane protein that direct efficient processing and presentation of the Ag by APCs. As a "warhead" for the APCs, we transduced them with the gene for FasL, which induces apoptosis of Fas-expressing activated T cells. To protect the transduced APCs from self-destruction by FasL, we transferred cDNA for a truncated form of Fas-associated death domain, which inhibits Fas-mediated cell death. Our results show that the engineered APCs effectively expressed the genes of interest. APCs transduced with VVV carrying all three gene constructs specifically killed HA-transgenic T cells in culture. Coculture with T cells specific for an unrelated Ag (OVA) had no significant effect. Our in vitro findings show that APCs can be genetically engineered to target and kill Ag-specific T cells and represent a promising novel strategy for the specific treatment of autoimmune diseases.


Subject(s)
Adaptor Proteins, Signal Transducing , Adoptive Transfer/methods , Antigen-Presenting Cells/transplantation , Protein Engineering/methods , Adjuvants, Immunologic/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Carrier Proteins/genetics , Cell Line , Cytotoxicity, Immunologic/genetics , Fas Ligand Protein , Fas-Associated Death Domain Protein , Gene Targeting/methods , Gene Transfer Techniques , Genetic Vectors/chemical synthesis , Genetic Vectors/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Interphase/genetics , Interphase/immunology , Ligands , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/toxicity , Mice , Mice, Inbred BALB C , Mice, Transgenic , Recombination, Genetic/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Vaccinia virus/genetics , Vaccinia virus/immunology , fas Receptor/genetics , fas Receptor/immunology
8.
Cancer Chemother Pharmacol ; 46 Suppl: S67-72, 2000.
Article in English | MEDLINE | ID: mdl-10950151

ABSTRACT

When irradiated and administered intradermally as vaccines, cancer cells engineered to secrete high levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) by gene transfer elicit potent anticancer immune responses in a variety of animal tumor models. Upon vaccination, antigens present in the cancer cells are phagocytosed and processed by skin dendritic cells. These dendritic cells then prime anticancer immune responses by presenting antigenic peptides to T cells. The immune responses generated are capable of eradicating small but lethal cancer cell inocula with minimal toxicity in preclinical animal tumor studies. To develop this vaccination strategy for the treatment of human genitourinary cancers, we have conducted phase I clinical trials using human genitourinary cancer cells as sources of cancer cell antigens. In the first human clinical trial of genetically engineered cancer cell vaccines, a phase I clinical trial of kidney cancer cell vaccines (n = 18), kidney cancer cells were removed at surgery, propagated briefly in vitro, and then genetically modified to secrete high levels of GM-CSF via ex vivo transduction with the retrovirus MFG-GM-CSF. After irradiation, the kidney cancer cells were administered as vaccines to 18 patients with advanced kidney cancers. Vaccine treatment, which caused few side effects, nonetheless appeared to trigger anticancer immune responses manifest as conversion of delayed-type hypersensitivity (DTH) skin responses against irradiated autologous cancer cells after vaccination. Biopsies of vaccine sites yielded findings reminiscent of biopsies from preclinical animal model studies, with evidence of vaccine cell recruitment of dendritic cells, T cells, and eosinophils. One patient with measurable kidney cancer metastases treated at the highest vaccine dose level experienced a partial treatment response. The bioactivity of GM-CSF-secreting autologous cancer cell vaccines was confirmed in a phase I clinical trial for prostate cancer (n = 8). Vaccine cells were prepared from surgically harvested prostate tumors by ex vivo transduction with MFG-GM-CSF in a manner similar to that used for the kidney cancer trial. Vaccine treatment was well tolerated and associated with induction of anticancer immunity as assessed using DTH skin testing. In addition, new antiprostate cancer cell antibodies were detected in serum samples from treated men as a consequence of vaccination. These first clinical trials of GM-CSF-secreting cancer cell vaccines for the treatment of genitourinary cancers have demonstrated both safety and bioactivity, in that very few side effects have been seen and anticancer immune responses have been detected. Future clinical studies will be required to assess vaccine treatment efficacy, refine vaccination dose and schedule, define the appropriate clinical context for the use of such vaccines, and ascertain optimal combinations involving vaccines and other local or systemic anticancer treatments.


Subject(s)
Cancer Vaccines/immunology , Carcinoma, Renal Cell/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Kidney Neoplasms/immunology , Prostatic Neoplasms/immunology , Adult , Aged , Antigens, Neoplasm/blood , Antigens, Neoplasm/immunology , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Dose-Response Relationship, Immunologic , Female , Gene Transfer Techniques , Genetic Engineering , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Hypersensitivity, Delayed/immunology , Kidney Neoplasms/therapy , Male , Middle Aged , Prostatic Neoplasms/therapy , Retroviridae/genetics
10.
Blood ; 95(10): 3011-9, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10807763

ABSTRACT

For many cancers, autologous bone marrow transplantation (BMT) achieves a minimal residual disease state, yet relapse rates remain high. Using a syngeneic murine bone marrow transplant model, we demonstrate that vaccination with irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing autologous tumor cells is effective in the post-BMT period and actually results in a greater tumor-free survival than vaccination in the nontransplant setting. Employing T cells specific for a model tumor-antigen, we find that transplantation of the tumor-bearing host results in a massive expansion and activation of tumor-specific T cells in the early posttransplant period, but this response rapidly declines in association with tumor progression. Immunization with irradiated GM-CSF tumor cells during the period of immune reconstitution results in the sustained amplification and activation of this response that closely correlates with freedom from relapse. These results demonstrate the feasibility of integrating GM-CSF vaccines in the postautologous BMT setting and suggest mechanisms that may contribute to the observed efficacy of immunization during the critical period of immune reconstitution.


Subject(s)
Bone Marrow Transplantation , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Graft vs Tumor Effect , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunization , Mice , Mice, Inbred BALB C , Secondary Prevention , T-Lymphocytes/immunology
11.
J Exp Med ; 191(3): 541-50, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-10662799

ABSTRACT

In many cases, induction of CD8(+) CTL responses requires CD4(+) T cell help. Recently, it has been shown that a dominant pathway of CD4(+) help is via antigen-presenting cell (APC) activation through engagement of CD40 by CD40 ligand on CD4(+) T cells. To further study this three cell interaction, we established an in vitro system using dendritic cells (DCs) as APCs and influenza hemagglutinin (HA) class I and II peptide-specific T cell antigen receptor transgenic T cells as cytotoxic T lymphocyte precursors and CD4(+) T helper cells, respectively. We found that CD4(+) T cells can provide potent help for DCs to activate CD8(+) T cells when antigen is provided in the form of either cell lysate, recombinant protein, or synthetic peptides. Surprisingly, this help is completely independent of CD40. Moreover, CD40-independent CD4(+) help can be documented in vivo. Finally, we show that CD40-independent T cell help is delivered through both sensitization of DCs and direct CD4(+)-CD8(+) T cell communication via lymphokines. Therefore, we conclude that CD4(+) help comprises at least three components: CD40-dependent DC sensitization, CD40-independent DC sensitization, and direct lymphokine-dependent CD4(+)-CD8(+) T cell communication.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Animals , Antigens, Differentiation, T-Lymphocyte , Cell Communication/drug effects , Dendritic Cells/drug effects , Lymphocyte Activation , Lymphokines/pharmacology , Mice , Mice, Inbred BALB C , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology
12.
Cancer Res ; 59(20): 5160-8, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10537292

ABSTRACT

Vaccination with irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting gene-transduced cancer vaccines induces tumoricidal immune responses. In a Phase I human gene therapy trial, eight immunocompetent prostate cancer (PCA) patients were treated with autologous, GM-CSF-secreting, irradiated tumor vaccines prepared from ex vivo retroviral transduction of surgically harvested cells. Expansion of primary cultures of autologous vaccine cells was successful to meet trial specifications in 8 of 11 cases (73%); the yields of the primary culture cell limited the number of courses of vaccination. Side effects were pruritus, erythema, and swelling at vaccination sites. Vaccine site biopsies manifested infiltrates of dendritic cells and macrophages among prostate tumor vaccine cells. Vaccination activated new T-cell and B-cell immune responses against PCA antigens. T-cell responses, evaluated by assessing delayed-type hypersensitivity (DTH) reactions against untransduced autologous tumor cells, were evident in two of eight patients before vaccination and in seven of eight patients after treatment. Reactive DTH site biopsies manifested infiltrates of effector cells consisting of CD45RO+ T-cells, and degranulating eosinophils consistent with activation of both Th1 and Th2 T-cell responses. A distinctive eosinophilic vasculitis was evident near autologous tumor cells at vaccine sites, and at DTH sites. B-cell responses were also induced. Sera from three of eight vaccinated men contained new antibodies recognizing polypeptides of 26, 31, and 150 kDa in protein extracts from prostate cells. The 150-kDa polypeptide was expressed by LNCaP and PC-3 PCA cells, as well as by normal prostate epithelial cells, but not by prostate stromal cells. No antibodies against prostate-specific antigen were detected. These data suggest that both T-cell and B-cell immune responses to human PCA can be generated by treatment with irradiated, GM-CSF gene-transduced PCA vaccines.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Prostatic Neoplasms/therapy , Vaccines, Synthetic/immunology , B-Lymphocytes/immunology , Gene Transfer Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Hypersensitivity, Delayed/etiology , Male , Middle Aged , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , T-Lymphocytes/immunology , Vaccination
13.
Proc Natl Acad Sci U S A ; 96(20): 11476-81, 1999 Sep 28.
Article in English | MEDLINE | ID: mdl-10500201

ABSTRACT

The efficacy of therapeutic vaccination for the treatment of cancer is limited by peripheral tolerance to tumor antigens. In vivo blockade of CTLA-4, a negative regulator of T cell function, can induce the regression of established tumors and can augment the tumor rejection achieved through therapeutic vaccination. These outcomes may reflect enhanced tumor-specific T cell priming and/or interference with the development of tolerance to tumor antigens. We examined the effect of CTLA-4 blockade on the fate and function of T cells specific for a model tumor antigen in the tumor-bearing host. We found that while CTLA-4 blockade enhanced the priming of responsive T cells, it did not prevent the induction of tolerance to tumor antigens. These results demonstrate that there is a critical window in which the combination of CTLA-4 blockade and vaccination achieves an optimal response, and they point to mechanisms other than CTLA-4 engagement in mediating peripheral T cell tolerance to tumor antigens.


Subject(s)
Antigens, Differentiation/physiology , Antigens, Neoplasm/immunology , Immune Tolerance , Immunoconjugates , T-Lymphocytes/immunology , Abatacept , Animals , Antigens, CD/physiology , B7-1 Antigen/physiology , B7-2 Antigen , CTLA-4 Antigen , Male , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Vaccination
14.
Hum Gene Ther ; 10(12): 1983-91, 1999 Aug 10.
Article in English | MEDLINE | ID: mdl-10466632

ABSTRACT

Irradiated tumor cells transduced with the gene encoding the cytokine GM-CSF have been extensively studied as a vaccine formulation capable of priming systemic antitumor immune responses in the tumor-bearing host. In spite of the therapeutic promise of this vaccine strategy demonstrated in both animal models and early-phase clinical trials, clinical development has been limited by difficulties pertaining to the need to establish in culture the tumor of each patient and to perform individualized gene transfer. To circumvent these issues, we generated an HLA-negative human cell line producing large quantities of human GM-CSF for use as a universal bystander cell to be mixed with unmodified autologous tumor cells in the formulation of a vaccine. This line is easily propagated as a suspension culture in defined, serum-free medium. In a mouse model, we find that vaccination with a mixture of autologous tumor cells and an MHC-negative allogeneic GM-CSF-producing bystander cell primes antitumor immune responses that are equivalent or better than those achieved using autologous tumor cells directly transduced to secrete GM-CSF. This strategy greatly simplifies further clinical development of autologous tumor cell-based vaccines.


Subject(s)
Cancer Vaccines , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Tumor Cells, Cultured , Animals , Cell Line , Cell Transplantation , Disease Models, Animal , Gene Transfer Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Transduction, Genetic
15.
Nat Med ; 5(7): 780-7, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10395323

ABSTRACT

Tumor antigen-specific T-cell tolerance limits the efficacy of therapeutic cancer vaccines. Antigen-presenting cells mediate the induction of T-cell tolerance to self-antigens. We therefore assessed the fate of tumor-specific CD4+ T cells in tumor-bearing recipients after in vivo activation of antigen-presenting cells with antibodies against CD40. Such treatment not only preserved the responsiveness of this population, but resulted in their endogenous activation. Established tumors regressed in vaccinated mice treated with antibody against CD40 at a time when no response was achieved with vaccination alone. These results indicate that modulation of antigen-presenting cells may be a useful strategy for enhancing responsiveness to immunization.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Cancer Vaccines , Carcinoma, Renal Cell/prevention & control , Kidney Neoplasms/prevention & control , Lung Neoplasms/secondary , Membrane Glycoproteins/immunology , Adoptive Transfer , Animals , CD40 Ligand , Carcinoma, Renal Cell/immunology , Immune Tolerance , Kidney Neoplasms/immunology , Lung Neoplasms/prevention & control , Lymphocyte Transfusion , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Tumor Cells, Cultured
16.
Biol Reprod ; 59(5): 1108-15, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9780316

ABSTRACT

To determine whether chronic oxytocin pretreatment inhibits adenylyl cyclase, we compared adenylyl cyclase activity in membranes prepared from cultured, immortalized rat myometrial cells that were untreated or pretreated for 24 h with oxytocin. Chronic oxytocin pretreatment (1 x 10(-5) M for 24 h) attenuated basal, guanosine triphosphate (1 x 10(-5) M)-, isoproterenol (1 x 10(-4) M)-, forskolin (1 x 10(-5) M)-, MnCl2 (20 mM)- or NaF (1 x 10(-2) M)-stimulated adenylyl cyclase activity by 27 +/- 5% to 39 +/- 11% (n = 6, p < 0.05). Oxytocin pretreatment for 2 h (n = 5) did not produce a significant effect. To understand the mechanism by which oxytocin pretreatment decreased activity of the adenylyl cyclase pathway, we compared effects of pretreatment with either oxytocin or phenylephrine on adenylyl cyclase activity and determined the effects of Gi inhibition and protein kinase C (PKC) depletion. Chronic (24 h) phenylephrine pretreatment (1 x 10(-4) M) had effects similar to those of oxytocin pretreatment (1 x 10(-5) M). PKC depletion with phorbol 12-myristate 13-acetate (1 x 10(-6) M, 41 h) prevented attenuation of adenylyl cyclase activity by oxytocin pretreatment (1 x 10(-5) M for 24 h). Inhibition of Gi by pertussis toxin pretreatment (1.25 microg/ml, 41 h) had no significant effect. These findings suggest that chronic oxytocin pretreatment desensitizes the adenylyl cyclase pathway by a cross-regulatory mechanism that involves activation of Gq and PKC.


Subject(s)
Adenylyl Cyclase Inhibitors , Enzyme Inhibitors/pharmacology , Myometrium/enzymology , Oxytocin/pharmacology , Adenylate Cyclase Toxin , Animals , Cells, Cultured , Chlorides/pharmacology , Colforsin/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , Guanosine Triphosphate/pharmacology , Isoproterenol/pharmacology , Manganese Compounds/pharmacology , Pertussis Toxin , Phenylephrine/pharmacology , Pregnancy , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Sodium Fluoride/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Virulence Factors, Bordetella/pharmacology
17.
J Immunol ; 160(2): 643-51, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9551898

ABSTRACT

To determine how self-tolerance can alter the ability of the immune system to respond against tumor-associated Ags that are also expressed by normal tissue, we designed experiments in which the same protein was expressed both as a tumor Ag and as a transgene product. Unlike conventional BALB/c mice that rejected renal carcinoma cells transfected with the influenza virus hemagglutinin (Renca-HA), transgenic mice that are tolerant of HA due to its expression as a self-Ag on pancreatic islet beta cells, (Ins-HA mice) supported progressive growth of these tumor cells. However, when Ins-HA mice were immunized with a recombinant strain of vaccinia virus expressing the dominant H-2Kd peptide epitope of HA before receiving Renca-HA cells, they too were able to reject the tumor cells. Rejection of Renca-HA cells by immunized Ins-HA mice was found to be associated with the generation of CTL having much lower avidity for target cells presenting the KdHA epitope than CTL from immunized conventional BALB/c mice. Significantly, we show that self-tolerance to the HA Ag is quantitative rather then absolute, and that vaccination of Ins-HA mice can activate low avidity KdHA-specific CD8+ T cells that are able to reject tumor cells expressing high levels of HA, yet these mice remain tolerant of pancreatic islet beta cells expressing HA.


Subject(s)
Autoimmunity , Epitopes, T-Lymphocyte/immunology , Graft Rejection , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/immunology , Animals , Autoimmunity/genetics , Cell Division/genetics , Cell Division/immunology , Epitopes, T-Lymphocyte/genetics , Graft Rejection/genetics , H-2 Antigens/genetics , H-2 Antigens/metabolism , Hemagglutinins/genetics , Hemagglutinins/immunology , Hemagglutinins/metabolism , Influenza A virus/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/prevention & control , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Transplantation , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured
18.
Int J Oncol ; 12(1): 161-70, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9454900

ABSTRACT

Cancer cells genetically modified to secrete immunoregulatory cytokines offer great promise for human cancer treatment as tumor vaccines. However, in preclinical animal studies, large established cancer burdens have appeared difficult to eradicate with such vaccines. For example, lethally-irradiated GM-CSF-secreting CT26 colon carcinoma cell vaccine therapy tends to cure only animals bearing 1 x 10(5) wild-type CT26 cells or less. For many human cancers, antineoplastic chemotherapy can often significantly reduce systemic cancer burdens. Unfortunately, for most advanced metastatic solid organ cancers, such as cancers of the breast, colon, and prostate, antineoplastic drug treatments generally fail to effect cancer cures. Treatment regimens combining genetically-modified cancer cell vaccine therapy and antineoplastic chemotherapy have the potential to increase advanced cancer cure rates if antineoplastic drugs and drug combinations that do not inhibit vaccine-induced immune responses can be identified. To assess the potential immunomodulatory properties of commonly-used antineoplastic drugs that might be used in combination with cancer vaccine treatments, we studied the effects of the drugs on antitumor immune responses manifest by animals receiving lethally-irradiated GM-CSF-secreting CT26 cell vaccines. Immunomodulatory properties of the antineoplastic drugs were evaluated i) by monitoring drug effects on the generation of tumor-specific CD8+ cytotoxic T-lymphocytes (CTLs) in response to GM-CSF-secreting CT26 vaccine administration, ii) by determining drug effects on the resistance of vaccinated animals to subsequent challenge with lethal inoculac of CT26 cells, and iii) by evaluating combination drug and vaccine treatment efficacy against established CT26 tumors. Using this approach, doxorubicin was found to possess apparent immunostimulatory activities, depending on the dose and schedule of administration, while cyclophosphamide appeared immunosuppressive. The different immunomodulatory properties of doxorubicin and cyclophosphamide may be clinically relevant: combination doxorubicin and vaccine treatment of established CT26 cancers increased cure rates over that achieved with either agent alone, while combination cyclophosphamide and vaccine treatment of animals carrying CT26 tumors was no better in curing the animals than drug treatment alone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines/therapeutic use , Colonic Neoplasms/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neoplasms, Experimental/therapy , Adjuvants, Immunologic/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Humans , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/analysis , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
20.
Cancer Res ; 57(8): 1537-46, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9108457

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transduced, irradiated tumor vaccines induce potent, T-cell-mediated antitumor immune responses in preclinical models. We report the initial results of a Phase I trial evaluating this strategy for safety and the induction of immune responses in patients with metastatic renal cell carcinoma (RCC). Patients were treated in a randomized, double-blind dose-escalation study with equivalent doses of autologous, irradiated RCC vaccine cells with or without ex vivo human GM-CSF gene transfer. The replication-defective retroviral vector MFG was used for GM-CSF gene transfer. No dose-limiting toxicities were encountered in 16 fully evaluable patients. GM-CSF gene-transduced vaccines were equivalent in toxicity to nontransduced vaccines up to the feasible limits of autologous tumor vaccine yield. No evidence of autoimmune disease was observed. Biopsies of intradermal sites of injection with GM-CSF gene-transduced vaccines contained distinctive macrophage, dendritic cell, eosinophil, neutrophil, and T-cell infiltrates similar to those observed in preclinical models of efficacy. Histological analysis of delayed-type hypersensitivity responses in patients vaccinated with GM-CSF-transduced vaccines demonstrated an intense eosinophil infiltrate that was not observed in patients who received nontransduced vaccines. An objective partial response was observed in a patient treated with GM-CSF gene-transduced vaccine who displayed the largest delayed-type hypersensitivity conversion. No replication-competent retrovirus was detected in vaccinated patients. This Phase I study demonstrated the feasibility, safety, and bioactivity of an autologous GM-CSF gene-transduced tumor vaccine for RCC patients.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Gene Transfer Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Kidney Neoplasms/therapy , Adult , Aged , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cancer Vaccines/radiation effects , Carcinoma, Renal Cell/immunology , Defective Viruses/genetics , Double-Blind Method , Drug Eruptions/etiology , Drug Eruptions/immunology , Drug Eruptions/pathology , Female , Genetic Vectors/genetics , Humans , Hypersensitivity, Delayed/pathology , Kidney Neoplasms/immunology , Male , Middle Aged , Vaccination/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...