Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Mater ; 34(29): e2109661, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35165959

ABSTRACT

The ability to rapidly assess and monitor patient immune responses is critical for clinical diagnostics, vaccine design, and fundamental investigations into the presence or generation of protective immunity against infectious diseases. Recently, findings on the limits of antibody-based protection provided by B-cells have highlighted the importance of engaging pathogen-specific T-cells for long-lasting and broad protection against viruses and their emergent variants such as in SARS-CoV-2. However, low-cost and point-of-care tools for detecting engagement of T-cell immunity in patients are conspicuously lacking in ongoing efforts to assess and control population-wide disease risk. Currently available tools for human T-cell analysis are time and resource-intensive. Using multichannel silicon-nanowire field-effect transistors compatible with complementary metal-oxide-semiconductor, a device designed for rapid and label-free detection of human T-cell immune responses is developed. The generalizability of this approach is demonstrated by measuring T-cell responses against melanoma antigen MART1, common and seasonal viruses CMV, EBV, flu, as well as emergent pandemic coronavirus, SARS-CoV-2. Further, this device provides a modular and translational platform for optimizing vaccine formulations and combinations, offering quick and quantitative readouts for acquisition and persistence of T-cell immunity against variant-driven pathogens such as flu and pandemic SARS-CoV-2.


Subject(s)
Biosensing Techniques , COVID-19 , Nanowires , Antiviral Agents , COVID-19/diagnosis , Humans , SARS-CoV-2 , T-Lymphocytes
2.
Methods Mol Biol ; 2097: 173-195, 2020.
Article in English | MEDLINE | ID: mdl-31776926

ABSTRACT

Generation of large numbers of dendritic cells (DC) for research or immunotherapeutic purposes typically involves in vitro conversion of murine bone marrow precursors or human blood monocytes to DC via cultivation with supraphysiologic concentrations of cytokines such as GM-CSF and IL-4 for up to 7 days. Alternatively, our group has recently established a new approach, based on the underlying mechanism of action of a widely used cancer immunotherapy termed Extracorporeal Photochemotherapy (ECP). Our method of rapid and cytokine-free production of therapeutically relevant DC populations, leveraging the innate physiologic programs likely responsible for DC differentiation from blood monocytes in vivo, potentially offers a novel, inexpensive, and easily accessible source of DC for clinical and research uses. This approach involves ex vivo physiologic reprogramming of blood monocytes to immunologically tunable dendritic antigen-presenting cells, which we term "phDC," for physiological DC. To facilitate access and utilization of these new DC populations by the research community, in this chapter, we describe the use of a scaled-down version of the clinical ECP leukocyte-treatment device termed the Transimmunization (TI) chamber or plate, suitable for processing both mouse and human samples. We highlight the methodological sequences necessary to isolate mouse or human peripheral blood mononuclear cell (PBMC) from whole blood, and to expose those PBMC to the TI chamber for facilitating monocyte activation and conversion to physiological DC (phDC) through interaction with blood proteins and activated platelets under controlled flow conditions. We then provide sample protocols for potential applications of the generated DC, including their use as vaccinating antigen-presenting cells (APC) in murine in vivo antitumor models, and in human ex vivo T-cell stimulation and antigen cross-presentation assays which mimic clinical vaccination. We additionally highlight the technical aspects of loading mouse or human phDC with tumor-associated antigens (TAA) in the form of peptides or apoptotic tumor cells. We provide a simple and clinically relevant means to reprogram blood monocytes into functional APC, potentially replacing the comparatively expensive and clinically disappointing cytokine-derived DC which have previously dominated the dendritic cell landscape.


Subject(s)
Dendritic Cells/cytology , Immunotherapy/methods , Animals , Anticoagulants/pharmacology , Antigens, Neoplasm/metabolism , Blood Donors , Cells, Cultured , Humans , Male , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Peptides/metabolism , Photochemotherapy
3.
J Vis Exp ; (147)2019 05 17.
Article in English | MEDLINE | ID: mdl-31157760

ABSTRACT

Extracorporeal photochemotherapy (ECP) is a widely used cancer immunotherapy for cutaneous T cell lymphoma (CTCL), operative in over 350 university centers worldwide. While ECP's clinical efficacy and exemplary safety profile have driven its widespread use, elucidation of the underlying mechanisms has remained a challenge, partly owing to lack of a laboratory ECP model. To overcome this obstacle and create a simple, user-friendly platform for ECP research, we developed a scaled-down version of the clinical ECP leukocyte-processing device, suitable for work with both mouse models, and small human blood samples. This device is termed the Transimmunization (TI) chamber, or plate. In a series of landmark experiments, the miniaturized device was used to produce a cellular vaccine that regularly initiated therapeutic anti-cancer immunity in several syngeneic mouse tumor models. By removing individual factors from the experimental system and ascertaining their contribution to the in vivo anti-tumor response, we then elucidated key mechanistic drivers of ECP immunizing potential. Collectively, our results revealed that anti-tumor effects of ECP are initiated by dendritic cells (DC), physiologically generated through blood monocyte interaction with platelets in the TI plate, and loaded with antigens from tumor cells whose apoptotic cell death is finely titrated by exposure to the photoactivatable DNA cross-linking agent 8-methoxypsoralen and UVA light (8-MOPA). When returned to the mouse, this cellular vaccine leads to specific and transferable anti-tumor T cell immunity. We verified that the TI chamber is also suitable for human blood processing, producing human DCs fully comparable in activation state and profile to those derived from the clinical ECP chamber. The protocols presented here are intended for ECP studies in mouse and man, controlled generation of apoptotic tumor cells with 8-MOPA, and rapid production of physiologic human and mouse monocyte-derived DCs for a variety of applications.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/physiology , Animals , Apoptosis , Blood Platelets/immunology , Cell Communication , Humans , Immunization , Melanoma/pathology , Mice, Inbred C57BL , Monocytes/immunology , Photopheresis , Skin Neoplasms
4.
Cancer Res ; 78(14): 4045-4058, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29764863

ABSTRACT

Extracorporeal photochemotherapy (ECP) is a cancer immunotherapy for cutaneous T-cell lymphoma (CTCL) operative in more than 350 centers worldwide. Although its efficacy and favorable safety profile have driven its widespread use, elucidation of its underlying mechanism has been difficult. In this study, we identify the principal contributors to the anticancer immunotherapeutic effects of ECP, with the goal of enhancing potency and broadening applicability to additional malignancies. First, we scaled down the clinical ECP leukocyte-processing device to mouse size. Second, we used that miniaturized device to produce a cellular vaccine that regularly initiated therapeutic antimelanoma immunity. Third, we individually subtracted key factors from either the immunizing inoculum or the treated animal to ascertain their contribution to the in vivo antimelanoma response. Platelet-signaled monocyte-to-dendritic cell (DC) differentiation followed by sorting/processing/presentation of tumor antigens derived from internalized apoptotic tumor cells were absolute requirements. As in clinical ECP, immunogenic cell death of tumor cells was finely titrated by DNA cross-linkage mediated by photoactivated 8-methoxypsoralen (8-MOPA). ECP-induced tumor-loaded DC were effective immunotherapeutic agents only if they were spared exposure to 8-MOPA, indicating that healthy DC are required for ECP. Infusion of responder T cells into naïve tumor-challenged mice established the protective role of stimulated T-cell antitumor immunity. Collectively, these results reveal that selective antitumor effects of ECP are initiated by tumor antigen-loaded, ECP-induced DC, which promote potent collaboration between CD4 and CD8 tumor-specific T cells. These mechanistic insights suggest potential therapeutic applicability of ECP to solid tumors in addition to CTCL.Significance: These findings identify principal cellular contributors to the anticancer immunotherapeutic impact of ECP and suggest this treatment may be applicable to a broad spectrum of immunogenic malignancies. Cancer Res; 78(14); 4045-58. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/drug effects , Lymphoma, T-Cell, Cutaneous/drug therapy , Monocytes/drug effects , Animals , Antigens, Neoplasm/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Line, Tumor , Dendritic Cells/metabolism , Immunotherapy/methods , Lymphoma, T-Cell, Cutaneous/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Photochemotherapy/methods
5.
Proc Natl Acad Sci U S A ; 111(23): 8571-6, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24799704

ABSTRACT

Nonhuman proteins have valuable therapeutic properties, but their efficacy is limited by neutralizing antibodies. Recombinant immunotoxins (RITs) are potent anticancer agents that have produced many complete remissions in leukemia, but immunogenicity limits the number of doses that can be given to patients with normal immune systems. Using human cells, we identified eight helper T-cell epitopes in PE38, a portion of the bacterial protein Pseudomonas exotoxin A which consists of the toxin moiety of the RIT, and used this information to make LMB-T18 in which three epitopes were deleted and five others diminished by point mutations in key residues. LMB-T18 has high cytotoxic and antitumor activity and is very resistant to thermal denaturation. The new immunotoxin has a 93% decrease in T-cell epitopes and should have improved efficacy in patients because more treatment cycles can be given. Furthermore, the deimmunized toxin can be used to make RITs targeting other antigens, and the approach we describe can be used to deimmunize other therapeutically useful nonhuman proteins.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunotoxins/immunology , Neoplasms/immunology , Recombinant Fusion Proteins/immunology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Amino Acids/genetics , Amino Acids/immunology , Animals , Antibody Formation/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Electrophoresis, Polyacrylamide Gel , Epitope Mapping , Exotoxins/genetics , Exotoxins/immunology , Female , Humans , Immunotherapy/methods , Immunotoxins/genetics , Immunotoxins/therapeutic use , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, SCID , Models, Molecular , Neoplasms/pathology , Neoplasms/therapy , Peptides/genetics , Peptides/immunology , Point Mutation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Virulence Factors/genetics , Virulence Factors/immunology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
6.
Proc Natl Acad Sci U S A ; 109(51): E3597-603, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23213206

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins that are being developed for cancer treatment. We have produced RITs that contain PE38, a portion of the bacterial protein Pseudomonas exotoxin A. Because the toxin is bacterial, it often induces neutralizing antibodies, which limit the number of treatment cycles and the effectiveness of the therapy. Because T cells are essential for antibody responses to proteins, we adopted an assay to map the CD4(+) T-cell epitopes in PE38. We incubated peripheral blood mononuclear cells with an immunotoxin to stimulate T-cell expansion, followed by exposure to overlapping peptide fragments of PE38 and an IL-2 ELISpot assay to measure responses. Our observation of T-cell responses in 50 of 50 individuals correlates with the frequency of antibody formation in patients with normal immune systems. We found a single, highly immunodominant epitope in 46% (23/50) of the donors. The immunodominant epitope is DRB1-restricted and was observed in subjects with different HLA alleles, indicating promiscuity. We identified two amino acids that, when deleted or mutated to alanine, eliminated the immunodominant epitope, and we used this information to construct mutant RITs that are highly cytotoxic and do not stimulate T-cell responses in many donors.


Subject(s)
ADP Ribose Transferases/chemistry , Bacterial Toxins/chemistry , Epitopes, T-Lymphocyte/chemistry , Exotoxins/chemistry , Protein Engineering/methods , Virulence Factors/chemistry , Antibodies/chemistry , CD4-Positive T-Lymphocytes/cytology , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/chemistry , Epitopes, T-Lymphocyte/immunology , Gene Deletion , Genetic Variation , Humans , Immune System , Interleukin-2/metabolism , Leukocytes, Mononuclear/cytology , Molecular Conformation , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL
...