Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
J Immunother Cancer ; 10(12)2022 12.
Article En | MEDLINE | ID: mdl-36521930

Current Food and Drug Administration (FDA)-approved CD19-specific chimeric antigen receptor (CAR) T-cell therapies for B-cell malignancies are constitutively active and while efficacious, can cause morbidity and mortality. Their toxicities might be reduced if CAR T-cell activity was regulatable rather than constitutive. To test this, we compared the efficacies and morbidities of constitutively active (conventional) and regulatable (switchable) CAR (sCAR) T-cells specific for human CD19 (huCD19) in an immune-competent huCD19+ transgenic mouse model.Conventional CAR (CAR19) and sCAR T-cells were generated by retrovirally transducing C57BL/6 (B6) congenic T-cells with constructs encoding antibody-derived single chain Fv (sFv) fragments specific for huCD19 or a peptide neoepitope (PNE), respectively. Transduced T-cells were adoptively transferred into huCD19 transgenic hemizygous (huCD19Tg/0 ) B6 mice; healthy B-cells in these mice expressed huCD19Tg Prior to transfer, recipients were treated with a lymphodepleting dose of cyclophosphamide to enhance T-cell engraftment. In tumor therapy experiments, CAR19 or sCAR T-cells were adoptively transferred into huCD19Tg/0 mice bearing a syngeneic B-cell lymphoma engineered to express huCD19. To regulate sCAR T cell function, a switch protein was generated that contained the sCAR-specific PNE genetically fused to an anti-huCD19 Fab fragment. Recipients of sCAR T-cells were injected with the switch to link sCAR effector with huCD19+ target cells. Mice were monitored for survival, tumor burden (where appropriate), morbidity (as measured by weight loss and clinical scores), and peripheral blood lymphocyte frequency.CAR19 and sCAR T-cells functioned comparably regarding in vivo expansion and B-cell depletion. However, sCAR T-cells were better tolerated as evidenced by the recipients' enhanced survival, reduced weight loss, and improved clinical scores. Discontinuing switch administration allowed healthy B-cell frequencies to return to pretreatment levels.In our mouse model, sCAR T-cells killed huCD19+ healthy and malignant B-cells and were better tolerated than CAR19 cells. Our data suggest sCAR might be clinically superior to the current FDA-approved therapies for B-cell lymphomas due to the reduced acute and chronic morbidities and mortality, lower incidence and severity of side effects, and B-cell reconstitution on cessation of switch administration.


Antigens, CD19 , Lymphoma, B-Cell , United States , Humans , Mice , Animals , Mice, Inbred C57BL , T-Lymphocytes , Lymphoma, B-Cell/therapy , Disease Models, Animal , Mice, Transgenic , Morbidity , Weight Loss
2.
JCI Insight ; 7(17)2022 09 08.
Article En | MEDLINE | ID: mdl-35917188

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) efficacy is complicated by graft-versus-host disease (GVHD), a leading cause of morbidity and mortality. Regulatory T cells (Tregs) have shown efficacy in preventing GVHD. However, high Treg doses are often required, necessitating substantial ex vivo or in vivo expansion that may diminish suppressor function. To enhance in vivo suppressor function, murine Tregs were transduced to express an anti-human CD19 chimeric antigen receptor (hCAR19) and infused into lethally irradiated, hCD19-transgenic recipients for allo-HSCT. Compared with recipients receiving control transduced Tregs, those receiving hCAR19 Tregs had a marked decrease in acute GVHD lethality. Recipient hCD19 B cells and murine hCD19 TBL12-luciferase (TBL12luc) lymphoma cells were both cleared by allogeneic hCAR19 Tregs, which was indicative of graft-versus-tumor (GVT) maintenance and potentiation. Mechanistically, hCAR19 Tregs killed syngeneic hCD19+ but not hCD19- murine TBL12luc cells in vitro in a perforin-dependent, granzyme B-independent manner. Importantly, cyclophosphamide-treated, hCD19-transgenic mice given hCAR19 cytotoxic T lymphocytes without allo-HSCT experienced rapid lethality due to systemic toxicity that has been associated with proinflammatory cytokine release; in contrast, hCAR19 Treg suppressor function enabled avoidance of this severe complication. In conclusion, hCAR19 Tregs are a potentially novel and effective strategy to suppress GVHD without loss of GVT responses.


Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Neoplasms , Receptors, Chimeric Antigen , T-Lymphocytes, Regulatory , Animals , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Mice , Receptors, Antigen, T-Cell/metabolism , Transplantation, Homologous
3.
Front Immunol ; 10: 2488, 2019.
Article En | MEDLINE | ID: mdl-31681338

Immunotherapy is now mainstream. Advertisements are ubiquitous in print and visual media for immune based-therapies for various conditions and diseases. Smaller companies that develop novel immunotherapies are often quickly acquired by larger companies. More and more clinical trials are open for immune-based therapies, particularly for immune checkpoint blockades. As such, immunologists need to engage the public in conversations about the strengths and limitations of immunotherapy, and the necessity of research in propelling the field further. In this article, we discuss approaches we have taken to convey key concepts in immunology and cancer immunotherapy to non-scientists and health care professionals without expertise in immunology. Although the devil is always in the details, basic concepts in immunology and immunotherapy can be readily conveyed using stories and analogies, some of which we present here.


Immunotherapy, Adoptive/methods , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Educational Status , Health Literacy/methods , Health Literacy/standards , Health Literacy/statistics & numerical data , Humans , Medical Informatics/methods , Medical Informatics/standards , Medical Informatics/statistics & numerical data , Pamphlets , Patient Education as Topic/methods , Patient Education as Topic/standards , Patient Education as Topic/statistics & numerical data
4.
Nat Commun ; 10(1): 567, 2019 02 04.
Article En | MEDLINE | ID: mdl-30718505

The immunosuppressive tumor microenvironment limits the success of current immunotherapies. The host retains memory T cells specific for previous infections throughout the entire body that are capable of executing potent and immediate immunostimulatory functions. Here we show that virus-specific memory T cells extend their surveillance to mouse and human tumors. Reactivating these antiviral T cells can arrest growth of checkpoint blockade-resistant and poorly immunogenic tumors in mice after injecting adjuvant-free non-replicating viral peptides into tumors. Peptide mimics a viral reinfection event to memory CD8+ T cells, triggering antigen presentation and cytotoxic pathways within the tumor, activating dendritic cells and natural killer cells, and recruiting the adaptive immune system. Viral peptide treatment of ex vivo human tumors recapitulates immune activation gene expression profiles observed in mice. Lastly, peptide therapy renders resistant mouse tumors susceptible to PD-L1 blockade. Thus, re-stimulating known antiviral immunity may provide a unique therapeutic approach for cancer immunotherapy.


Immunotherapy/methods , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Child , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Middle Aged , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/physiology , Young Adult
5.
Cancers (Basel) ; 11(2)2019 Jan 24.
Article En | MEDLINE | ID: mdl-30682795

Recent advances in immunotherapy have included inhibition of immune checkpoint proteins in the tumor microenvironment and tumor lysate-based vaccination strategies. We combined these approaches in pet dogs with high-grade glioma. Administration of a synthetic peptide targeting the immune checkpoint protein, CD200, enhanced the capacity of antigen-presenting cells to prime T-cells to mediate an anti-glioma response. We found that in canine spontaneous gliomas, local injection of a canine-specific, CD200-directed peptide before subcutaneous delivery of an autologous tumor lysate vaccine prolonged survival relative to a historical control treated with autologous tumor lysate alone (median survivals of 12.7 months and 6.36 months, respectively). Antigen-presenting cells and T-lymphocytes primed with this peptide suppressed their expression of the inhibitory CD200 receptor, thereby enhancing their ability to initiate immune reactions in a glioblastoma microenvironment replete with the immunosuppressive CD200 protein. These results support consideration of a CD200 ligand as a novel glioblastoma immunotherapeutic agent.

6.
Mol Ther ; 26(6): 1423-1434, 2018 06 06.
Article En | MEDLINE | ID: mdl-29735365

The clinical success of chimeric antigen receptor (CAR) T cell therapy for CD19+ B cell malignancies can be limited by acute toxicities and immunoglobulin replacement needs due to B cell aplasia from persistent CAR T cells. Life-threatening complications include cytokine release syndrome and neurologic adverse events, the exact etiologies of which are unclear. To elucidate the underlying toxicity mechanisms and test potentially safer CAR T cells, we developed a mouse model in which human CD19 (hCD19)-specific mouse CAR T cells were adoptively transferred into mice whose normal B cells express a hCD19 transgene at hemizygous levels. Compared to homozygous hCD19 transgenic mice that have ∼75% fewer circulating B cells, hemizygous mice had hCD19 frequencies and antigen density more closely simulating human B cells. Hemizygous mice given a lethal dose of hCD19 transgene-expressing lymphoma cells and treated with CAR T cells had undetectable tumor levels. Recipients experienced B cell aplasia and antigen- and dose-dependent acute toxicities mirroring patient complications. Interleukin-6 (IL-6), interferon γ (IFN-γ), and inflammatory pathway transcripts were enriched in affected tissues. As in patients, antibody-mediated neutralization of IL-6 (and IFN-γ) blunted toxicity. Apparent behavioral abnormalities associated with decreased microglial cells point to CAR-T-cell-induced neurotoxicity. This model will prove useful in testing strategies designed to improve hCD19-specific CAR T cell safety.


Antigens, CD19/metabolism , B-Lymphocytes/metabolism , Animals , Female , Humans , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Transgenic
7.
J Immunother Cancer ; 4: 11, 2016.
Article En | MEDLINE | ID: mdl-26885373

BACKGROUND: Annexin A2 (ANXA2) is a pleiotropic, calcium-dependent, phospholipid-binding protein with a broad tissue distribution. It can be intracellular, membrane-bound, or secreted, and it exists as a monomer or heterotetramer. The secreted ANXA2 heterotetramer signals human and murine macrophages to produce IL-1, IL-6, and TNF-α through TLR4/MyD88- and TRIF-dependent pathways. METHODS: GL261 glioma cells were cultured in 5 % or 20 % O2. Monomeric ANXA2 (ANXA2m) was identified as a TLR2-binding protein enriched in 5 % O2 by mass spectrometry. Purified ANXA2m and ANXA2-derived peptides were added to TLR2-expressing reporter cells and immature dendritic cells (DCs) cells in vitro. ANXA2m was then mixed with chicken ovalbumin (OVA) for vaccination of TLR2 (+/+) and TLR2 (-/-) mice for subsequent quantification of antigen-specific CD8(+) T cell responses. The TLR2-binding region of ANXA2m was determined using various peptides derived from the ANXA2 amino terminus on TLR2 reporter cells and in vaccinated mice. RESULTS: ANXA2m is overexpressed by murine glioblastoma GL261 cells grown under 5 % O2, but not atmospheric 20 % O2, and acts as an adjuvant by inducing murine and human DC maturation through TLR2. ANXA2m upregulates CD80 and CD86 expression, enhances antigen cross-presentation, and induces the secretion of IL-12p70, TNF-α, and IFN-γ. The amino-terminal 15 amino acids of ANXA2m are necessary and sufficient for TLR2 binding and DC activation. CONCLUSION: This novel finding adds to the known functions of ANXA2 and suggests ways to exploit it as a vaccine adjuvant. ANXA2-antigen fusion peptides could be developed for patients as "off-the-shelf" agents containing common tumor antigens. Alternatively, they could be "personalized" and synthesized after tumor sequencing to identify immunogenic tumor-specific neo-antigens. As the amino terminal 15 amino acids of ANXA2 are required to stimulate TLR2 activity, a fusion peptide could be as short as 30 amino acids if one or two CD8 T cell epitopes are fused to the ANXA2 amino terminal portion. Future work will address the efficacy of ANXA2 peptide fusions alone and in combination with established TLR agonists to induce synergy in preclinical models of glioma as observed in other vaccines.

8.
Crit Rev Immunol ; 35(2): 153-72, 2015.
Article En | MEDLINE | ID: mdl-26351148

Despite the growing number of preclinical and clinical trials focused on immunotherapy for the treatment of malignant gliomas, the prognosis for this disease remains grim. Cancer immunotherapy seeks to recruit an effective immune response to eliminate tumor cells. To date, cancer vaccines have shown only limited effectiveness because of our incomplete understanding of the necessary effector cells and mechanisms that yield efficient tumor clearance. CD8+ T cell cytotoxic activity has long been proposed as the primary effector function necessary for tumor regression. However, there is increasing evidence that indicates that components of the immune system other than CD8+ T cells play important roles in tumor eradication and control. The following review should provide an understanding of the mechanisms involved in an effective antitumor response to guide future therapeutic designs. The information provided suggests an alternate means of effective tumor clearance in malignant glioma to the canonical CD8+ cytotoxic T cell mechanism.


CD8-Positive T-Lymphocytes/immunology , Glioblastoma/immunology , Glioblastoma/therapy , Immunotherapy , Animals , Cancer Vaccines/therapeutic use , Humans , Immunotherapy/methods
9.
J Immunother Cancer ; 2(1): 46, 2014.
Article En | MEDLINE | ID: mdl-25598973

BACKGROUND: Immunological quiescence in the central nervous system (CNS) is a potential barrier to immune mediated anti-tumor response. One suppressive mechanism results from the interaction of parenchyma-derived CD200 and its receptor on myeloid cells. We suggest that CD200/CD200R interactions on myeloid cells expand the myeloid-derived suppressor cell (MDSC) population and that blocking tumor-derived CD200 will enhance the efficacy of immunotherapy. METHODS: CD200 mRNA expression levels in human brain tumor tissue samples were measured by microarray. The amount of circulating CD200 protein in the sera of patients with brain tumors was determined by ELISA and, when corresponding peripheral blood samples were available, was correlated quantitatively with MDSCs. CD200-derived peptides were used as competitive inhibitors in a mouse model of glioblastoma immunotherapy. RESULTS: CD200 mRNA levels were measured in human brain tumors, with different expression levels being noted among the sub groups of glioblastoma, medulloblastoma and ependymoma. Serum CD200 concentrations were highest in patients with glioblastoma and correlated significantly with MDSC expansion. Similarly, in vitro studies determined that GL261 cells significantly expanded a MDSC population. Interestingly, a CD200R antagonist inhibited the expansion of murine MDSCs in vitro and in vivo. Moreover, inclusion of CD200R antagonist peptide in glioma tumor lysate-derived vaccines slowed tumor growth and significantly enhanced survival. CONCLUSION: These data suggest that CNS-derived tumors can evade immune surveillance by engaging CD200. Because of the homology between mouse and human CD200, our data also suggest that blockade of CD200 binding to its receptor will enhance the efficacy of immune mediated anti-tumor strategies for brain tumors.

10.
Mol Ther ; 21(8): 1526-35, 2013 Aug.
Article En | MEDLINE | ID: mdl-23689601

Clinical trials reveal that plasmid DNA (pDNA)-based gene delivery must be improved to realize its potential to treat human disease. Current pDNA platforms suffer from brief transgene expression, primarily due to the spread of transcriptionally repressive chromatin initially deposited on plasmid bacterial backbone sequences. Minicircle (MC) DNA lacks plasmid backbone sequences and correspondingly confers higher levels of sustained transgene expression upon delivery, accounting for its success in preclinical gene therapy models. In this study, we show for the first time that MC DNA also functions as a vaccine platform. We used a luciferase reporter transgene to demonstrate that intradermal delivery of MC DNA, relative to pDNA, resulted in significantly higher and persistent levels of luciferase expression in mouse skin. Next, we immunized mice intradermally with DNA encoding a peptide that, when presented by the appropriate major histocompatibility complex class I molecule, was recognized by endogenous CD8(+) T cells. Finally, immunization with peptide-encoding MC DNA, but not the corresponding full-length (FL) pDNA, conferred significant protection in mice challenged with Listeria monocytogenes expressing the model peptide. Together, our results suggest intradermal delivery of MC DNA may prove more efficacious for prophylaxis than traditional pDNA vaccines.


CD8-Positive T-Lymphocytes/immunology , DNA, Circular/immunology , Epitopes, T-Lymphocyte/immunology , Plasmids/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Cell Line , DNA, Circular/genetics , Epitopes, T-Lymphocyte/genetics , Female , Gene Expression , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Humans , Listeriosis/immunology , Listeriosis/prevention & control , Mice , Plasmids/genetics , Skin/metabolism , Transgenes/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
11.
J Immunol ; 190(2): 613-20, 2013 Jan 15.
Article En | MEDLINE | ID: mdl-23248259

Malignant gliomas are lethal brain tumors for which novel therapies are urgently needed. In animal models, vaccination with tumor-associated Ags efficiently primes T cells to clear gliomas. In clinical trials, cancer vaccines have been less effective at priming T cells and extending survival. Generalized immune suppression in the tumor draining lymph nodes has been documented in multiple cancers. However, a systematic analysis of how vaccination at various distances from the tumor (closest to farthest) has not been reported. We investigated how the injection site chosen for vaccination dictates CD8 T cell priming and survival in an OVA-transfected murine glioma model. Glioma-bearing mice were vaccinated with Poly:ICLC plus OVA protein in the neck, hind leg, or foreleg for drainage into the cervical, inguinal, or axillary lymph nodes, respectively. OVA-specific CD8 T cell number, TCR affinity, effector function, and infiltration into the brain decreased as the vaccination site approached the tumor. These effects were dependent on the presence of the tumor, because injection site did not appreciably affect CD8 T cell priming in tumor-free mice. Our data suggest the site of vaccination can greatly impact the effectiveness of cancer vaccines. Considering that previous and ongoing clinical trials have used a variety of injection sites, vaccination site is potentially a critical aspect of study design that is being overlooked.


Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Glioma/immunology , Animals , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Disease Models, Animal , Female , Glioma/mortality , Glioma/therapy , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
12.
J Virol ; 85(11): 5331-7, 2011 Jun.
Article En | MEDLINE | ID: mdl-21411533

How viruses evolve to select their receptor proteins for host cell entry is puzzling. We recently determined the crystal structures of NL63 coronavirus (NL63-CoV) and SARS coronavirus (SARS-CoV) receptor-binding domains (RBDs), each complexed with their common receptor, human angiotensin-converting enzyme 2 (hACE2), and proposed the existence of a virus-binding hot spot on hACE2. Here we investigated the function of this hypothetical hot spot using structure-guided biochemical and functional assays. The hot spot consists of a salt bridge surrounded by hydrophobic tunnel walls. Mutations that disturb the hot spot structure have significant effects on virus/receptor interactions, revealing critical energy contributions from the hot spot structure. The tunnel structure at the NL63-CoV/hACE2 interface is more compact than that at the SARS-CoV/hACE2 interface, and hence RBD/hACE2 binding affinities are decreased either by NL63-CoV mutations decreasing the tunnel space or by SARS-CoV mutations increasing the tunnel space. Furthermore, NL63-CoV RBD inhibits hACE2-dependent transduction by SARS-CoV spike protein, a successful application of the hot spot theory that has the potential to become a new antiviral strategy against SARS-CoV infections. These results suggest that the structural features of the hot spot on hACE2 were among the driving forces for the convergent evolution of NL63-CoV and SARS-CoV.


Coronavirus NL63, Human/physiology , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus/metabolism , Severe acute respiratory syndrome-related coronavirus/physiology , Virus Attachment , Angiotensin-Converting Enzyme 2 , DNA Mutational Analysis , Humans , Peptidyl-Dipeptidase A/genetics , Protein Binding , Receptors, Virus/genetics , Transduction, Genetic
13.
Protein Eng Des Sel ; 19(1): 37-45, 2006 Jan.
Article En | MEDLINE | ID: mdl-16243897

Immunotoxins are targeted therapeutics designed to kill cancer cells. The targeting moiety of an immunotoxin selectively binds to a tumor cell and targets it for death via an attached toxin. Because the toxins are typically of plant or bacterial origin, their clinical use is limited by immunogenicity and nonspecific toxicity. To circumvent these problems, we have begun to engineer immunotoxins containing human pancreatic ribonuclease. Here we describe the generation of ribonuclease mutants designed to evade a ubiquitous cytosolic inhibitor that would otherwise block cytotoxicity. Two mutants retained catalytic activity and were relatively resistant to the inhibitor. To deliver them to human T leukemic cells, these ribonuclease variants were fused to a single chain Fv fragment specific for CD 7. The ribonuclease-sFv fusion proteins bound CD 7(+) T cells and were internalized yet were not cytotoxic. Transfection of the proteins directly into the cytosol reduced cell viability, suggesting that the failure of the immunotoxins to kill cells when added externally resulted from the inability of the ribonuclease moiety to access the cytosol efficiently. Our results indicate appropriate intracellular routing, as well as resistance to inhibition, is critical to the cytotoxicity of human ribonuclease-based immunotoxins.


Cytosol/drug effects , Drug Resistance/drug effects , Immunotoxins/pharmacology , Protein Synthesis Inhibitors/pharmacology , Ribonucleases/immunology , Catalysis , Cytosol/metabolism , Cytotoxicity, Immunologic , Humans , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Leukemia, T-Cell/pathology , Mutation , Neoplasms/drug therapy , Neoplasms/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Ribonucleases/genetics , Ribonucleases/metabolism , Transfection , Tumor Cells, Cultured
14.
Vet Immunol Immunopathol ; 91(3-4): 205-15, 2003 Feb 10.
Article En | MEDLINE | ID: mdl-12586483

Dendritic cells (DC) are potent inducers of acquired immunity due to their ability to present antigens in the context of a costimulatory environment and consequently serve an essential role in vaccine efficacy. Strategies to enhance their function, such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-4 treatment to induce DC differentiation from peripheral blood monocytes, may therefore be useful as vaccine adjuvants. We now have evaluated the effect of recombinant GM-CSF on the differentiation of DC in swine. GM-CSF mRNA was readily detected in porcine splenocytes, with increased levels following treatment of the cells with ConA and LPS. Porcine GM-CSF was cloned and expressed in the methylotrophic yeast, Pichia pastoris, as a glycosylated protein that induced proliferation of porcine bone marrow cells. P. pastoris-derived GM-CSF induced expression of antigen presenting (MHC class II) and costimulatory (CD80-CD86) molecules and enhanced antigen presenting cell (APC) function consistent with the induction of functional DC. Thus, recombinant GM-CSF produced by P. pastoris may be a potent adjuvant for swine vaccines.


Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Pichia/genetics , Swine/immunology , Animals , Antigen Presentation/drug effects , Antigens, Surface/analysis , Antigens, Surface/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Division/drug effects , Dendritic Cells/immunology , Dose-Response Relationship, Immunologic , Gene Expression , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Immunophenotyping , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins
15.
J Immunol ; 169(10): 5622-9, 2002 Nov 15.
Article En | MEDLINE | ID: mdl-12421941

Heat shock protein 70 (hsp70) is a potent adjuvant that links innate and adaptive immune responses. To study how hsp70 activates naive CD8(+) T cells in vivo, we tracked Ag-specific CD8(+) T cells in mice immunized with a fusion protein containing chicken OVA linked to hsp70 derived from Mycobacterium tuberculosis (OVA.TBhsp70). On a molar basis, OVA.TBhsp70 was several hundred times more effective than OVA peptide plus CFA in eliciting specific CD8(+) T cell responses. Immunization with OVA.TBhsp70 activated >90% of detectable OVA-specific CD8(+) T cells within 3 days and led to the persistence of cytotoxic effectors for at least 17 days. These studies demonstrate that the potent adjuvant effect of M. tuberculosis hsp70 results from the relatively complete, rapid, and durable activation of Ag-specific CD8(+) T cells.


Adjuvants, Immunologic/physiology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HSP70 Heat-Shock Proteins/physiology , Lymphocyte Activation/immunology , Mycobacterium tuberculosis/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Adoptive Transfer , Animals , Antigens, Differentiation/biosynthesis , Bacterial Proteins , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/transplantation , Cell Division/genetics , Cell Division/immunology , Cell Line/transplantation , Cells, Cultured , Cytotoxicity Tests, Immunologic , Egg Proteins/administration & dosage , Egg Proteins/genetics , Egg Proteins/immunology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Freund's Adjuvant/administration & dosage , Genetic Vectors/administration & dosage , Genetic Vectors/analysis , Genetic Vectors/immunology , HSP70 Heat-Shock Proteins/administration & dosage , HSP70 Heat-Shock Proteins/genetics , Lipopolysaccharides/analysis , Lipopolysaccharides/pharmacology , Lymphocyte Activation/genetics , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mycobacterium tuberculosis/genetics , Ovalbumin/administration & dosage , Ovalbumin/genetics , Ovalbumin/immunology , Peptide Fragments , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , Tumor Cells, Cultured
16.
Immunol Res ; 25(2): 177-91, 2002.
Article En | MEDLINE | ID: mdl-11999171

Immunotoxins are therapeutic agents with a high degree of specificity and unique mechanism of action. An immunotoxin is a chimeric protein consisting of a targeting moiety linked to a toxin. The targeting moiety selectively binds to a tumor cell and targets it for death via the attached toxin. Generally, immunotoxins are specifically potent against cancer cells in vitro and in animal models of human malignancies. However, immunotoxins can be limited clinically by immunogenicity, toxicity, and instability. In this review, we offer ways to overcome these limitations to create "ideal immunotoxins" for cancer therapy. These include producing single chain targeting/toxin fusion proteins of fully human origin that are extracellularly stable but once internalized, can be cleaved by intracellular proteases to free the toxin and facilitate its translocation to the cytosol.


Immunotoxins/therapeutic use , Neoplasms/drug therapy , Animals , Humans , Immunotoxins/toxicity , Neoplasms/immunology
...