1.Adoptive Transfer of Colon Cancer Derived Peptide-specific CD8+ T Cells in HHD Mice.
Hun Soon JUNG ; In Sook AHN ; Hyung Ki DO ; Francois A LEMONNIER ; Boaz TIROSH ; Esther TZEHOVAL ; Ezra VADAI ; Lea EISENBACH ; Myoung Sool DO
Immune Network 2004;4(1):31-37
BACKGROUND: 1-8D gene is a member of human 1-8 interferon inducible gene family and is shown to be overexpressed in fresh colon cancer tissues. Three peptides 1-6, 3-5 and 3-7 derived from 1-8D gene were shown to have immunogenicity against colon cancer. METHODS: To study tumor immunotherapy of these peptides we established an adoptive transfer model. D(b-/-)Xbeta2 microglobulin (beta2m) null mice transgenic for a chimeric HLA-A2.1/D(b)-beta2m single chain (HHD mice) were immunized with irradiated peptide-loaded RMA-S/HHD/B7.1 transfectants. Spleens were removed after last immunization, and splenocytes were re-stimulated in vitro. Lymphocytes from vaccinated HHD mice were transferred together with IL-2 to the tumor bearing nude mice that were challenged S.C. with the HCT/HHD/B7 colon carcinoma cell line that was found to grow in these mice. RESULTS: Peptide 3-5 was found to be highly effective in CTL activity. Adoptively transferred anti-peptide 3-5 cytolytic T lymphocytes caused significant retardation in tumor growth. CONCLUSION: This study shows that peptide 3-5 can be the most effective candidate for the vaccine of adoptive immunotherapy against colon cancer.
Adoptive Transfer*
;
Animals
;
Cell Line
;
Colon*
;
Colonic Neoplasms*
;
Humans
;
Immunization
;
Immunotherapy
;
Immunotherapy, Adoptive
;
Interferons
;
Interleukin-2
;
Lymphocytes
;
Mice*
;
Mice, Nude
;
Peptides
;
Spleen
;
T-Lymphocytes*
2.Adoptive cell transfer therapy for hepatocellular carcinoma.
Renyu ZHANG ; Zhao ZHANG ; Zekun LIU ; Ding WEI ; Xiaodong WU ; Huijie BIAN ; Zhinan CHEN
Frontiers of Medicine 2019;13(1):3-11
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. This malignancy is associated with poor prognosis and high mortality. Novel approaches for prolonging the overall survival of patients with advanced HCC are urgently needed. The antitumor activities of adoptive cell transfer therapy (ACT), such as strategies based on tumor-infiltrating lymphocytes and cytokine-induced killer cells, are more effective than those of traditional strategies. Currently, chimeric antigen receptor T-cell (CAR-T) immunotherapy has achieved numerous breakthroughs in the treatment of hematological malignancies, including relapsed or refractory lymphoblastic leukemia and refractory large B-cell lymphoma. Nevertheless, this approach only provides a modest benefit in the treatment of solid tumors. The clinical results of CAR-T immunotherapy for HCC that could be obtained at present are limited. Some published studies have demonstrated that CAR-T could inhibit tumor growth and cause severe side effects. In this review, we summarized the current application of ACT, the challenges encountered by CAR-T technology in HCC treatment, and some possible strategies for the future direction of immunotherapeutic research.
Adoptive Transfer
;
methods
;
Carcinoma, Hepatocellular
;
immunology
;
therapy
;
Humans
;
Immunotherapy, Adoptive
;
methods
;
Liver Neoplasms
;
immunology
;
therapy
;
Lymphocytes, Tumor-Infiltrating
;
cytology
;
Randomized Controlled Trials as Topic
;
Receptors, Chimeric Antigen
;
T-Lymphocytes
;
cytology
3.Present status in studying immunotherapy for acute leukemia and its perspective--Editorial.
Journal of Experimental Hematology 2005;13(2):169-173
One of the important approaches for further prolonging remission duration and eradicating minimal residual disease in acute leukemia is immunotherapy. Four kinds of immunotherapy for acute leukemia are under investigation: (1) monoclonal antibodies, among them, Mylotarg (cytotoxic antibiotic calicheamicin linked to CD33 Mab) is given for the treatment of refractory or relapsed acute myeloid leukemia and molecular relapse in acute promyelocytic leukemia with good results, Campath-1H (antiCD52 Mab) is administered in the treatment of prolymphocytic leukemia and Rituximab (anti-CD20 Mab) in B-PLL with high complete remission rates. Other Mabs under preclinical and clinical trials include anti-IL-2 receptor Mab for the treatment of acute T lymphocytic leukemia, anti-220 kD Mab-6G7 for acute leukemias, recombinant immune toxin BL22 (anti-CD22) for hairy cell leukemia and Mabs labeled with radio-isotopes for different types of acute leukemias; (2) adoptive cellular immunotherapy using cytokine-induced killer cell, alloreactive NK cells, allogeneic or autologous leukemic-specific CD8(+) cytotoxic T lymphocytes, and other immune effector cells; (3) cytokines and other immune modulators comprising IL-2, IL-12, GM-CSF, CD40L, FLT-3L and thalidomide and its derivatives; (4) leukemia vaccines of several different formulations including antigen-specific, leukemia cell-based, leukemia antigen-pulsed dendritic cell (DC) and leukemia-derived DC vaccines, the latter two formulations are more attractive. In conclusion, up to now, the most effective example of immunotherapy in acute leukemia is provided by the administration of Mabs, and the majority of other approaches in immunotherapy for acute leukemia although promising, need further studies.
Acute Disease
;
Adoptive Transfer
;
methods
;
Antibodies, Monoclonal
;
immunology
;
therapeutic use
;
Cancer Vaccines
;
therapeutic use
;
Humans
;
Immunotherapy
;
methods
;
trends
;
Leukemia
;
immunology
;
therapy
4.Development of Auto Antigen-specific Regulatory T Cells for Diabetes Immunotherapy.
Immune Network 2016;16(5):281-285
CD4⁺ regulatory T cells (Tregs) are essential for normal immune surveillance, and their dysfunction can lead to the development of autoimmune diseases, such as type-1 diabetes (T1D). T1D is a T cell-mediated autoimmune disease characterized by islet β cell destruction, hypoinsulinemia, and severely altered glucose homeostasis. Tregs play a critical role in the development of T1D and participate in peripheral tolerance. Pluripotent stem cells (PSCs) can be utilized to obtain a renewable source of healthy Tregs to treat T1D as they have the ability to produce almost all cell types in the body, including Tregs. However, the right conditions for the development of antigen (Ag)-specific Tregs from PSCs (i.e., PSC-Tregs) remain undefined, especially molecular mechanisms that direct differentiation of such Tregs. Auto Ag-specific PSC-Tregs can be programmed to be tissue-associated and infiltrate to local inflamed tissue (e.g., islets) to suppress autoimmune responses after adoptive transfer, thereby avoiding potential overall immunosuppression from non-specific Tregs. Developing auto Ag-specific PSC-Tregs can reduce overall immunosuppression after adoptive transfer by accumulating inflamed islets, which drives forward the use of therapeutic PSC-Tregs for cell-based therapies in T1D.
Adoptive Transfer
;
Autoimmune Diseases
;
Autoimmunity
;
Glucose
;
Homeostasis
;
Immunosuppression
;
Immunotherapy*
;
Peripheral Tolerance
;
Pluripotent Stem Cells
;
Stem Cells
;
T-Lymphocytes
;
T-Lymphocytes, Regulatory*
5.Development of Auto Antigen-specific Regulatory T Cells for Diabetes Immunotherapy.
Immune Network 2016;16(5):281-285
CD4⁺ regulatory T cells (Tregs) are essential for normal immune surveillance, and their dysfunction can lead to the development of autoimmune diseases, such as type-1 diabetes (T1D). T1D is a T cell-mediated autoimmune disease characterized by islet β cell destruction, hypoinsulinemia, and severely altered glucose homeostasis. Tregs play a critical role in the development of T1D and participate in peripheral tolerance. Pluripotent stem cells (PSCs) can be utilized to obtain a renewable source of healthy Tregs to treat T1D as they have the ability to produce almost all cell types in the body, including Tregs. However, the right conditions for the development of antigen (Ag)-specific Tregs from PSCs (i.e., PSC-Tregs) remain undefined, especially molecular mechanisms that direct differentiation of such Tregs. Auto Ag-specific PSC-Tregs can be programmed to be tissue-associated and infiltrate to local inflamed tissue (e.g., islets) to suppress autoimmune responses after adoptive transfer, thereby avoiding potential overall immunosuppression from non-specific Tregs. Developing auto Ag-specific PSC-Tregs can reduce overall immunosuppression after adoptive transfer by accumulating inflamed islets, which drives forward the use of therapeutic PSC-Tregs for cell-based therapies in T1D.
Adoptive Transfer
;
Autoimmune Diseases
;
Autoimmunity
;
Glucose
;
Homeostasis
;
Immunosuppression
;
Immunotherapy*
;
Peripheral Tolerance
;
Pluripotent Stem Cells
;
Stem Cells
;
T-Lymphocytes
;
T-Lymphocytes, Regulatory*
6.Vaccinations with Dendritic Cells Can Elicit Preventive Immunity in a Squamous Cell Carcinoma Animal Model.
Young Ik SON ; Ye Jeung KO ; Seung Won CHUNG ; Chi Kyou LEE
Korean Journal of Otolaryngology - Head and Neck Surgery 2004;47(12):1289-1296
BACKGROUND AND OBJECTIVES: Dendritic cells (DC) can effectively mediate the prevention and regression of a variety of solid malignancies when they are properly primed with tumor antigens. However, little has been determined about the efficacy of DCbased vaccine for the prevention of squamous cell carcinoma (SCC). SUBJECTS AND METHOD: Bone marrow derived DC of C3H mouse were pulsed with ultraviolet B-irradiated apoptotic SCCVII cells, which are known as a poorly immunogenic SCC cell line. After vaccinations with those DC, a tumorigenic dosage of healthy SCCVII cells were challenged into the flank of animals and the tumor growth was assessed in a blinded and coded fashion. RESULTS: DC pulsed with apoptotic SCCVII cells effectively prevented the formation of solid tumor in 81% of vaccinated animals and mediated significant retardation of tumor growth (p=0.0029) in 19% of vaccinated animals when compared to control groups. SCCVII cell-specific T-cell response (IFN-gamma production) was observed in the immunized mice. CONCLUSION: Adoptive transfer of DC primed with apoptotic SCCVII cells can serve as an effective preventive vaccine in the poorly immunogenic SCC animal model.
Adoptive Transfer
;
Animals*
;
Antigens, Neoplasm
;
Bone Marrow
;
Carcinoma, Squamous Cell*
;
Cell Line
;
Dendritic Cells*
;
Immunotherapy
;
Mice
;
Mice, Inbred C3H
;
Models, Animal*
;
T-Lymphocytes
;
Vaccination*
;
Vaccines
7.Methods of trasferral of tissues and cells on slices.
Chinese Journal of Pathology 2008;37(5):348-349
Adoptive Transfer
;
methods
;
trends
;
Cells
;
pathology
;
Cells, Cultured
;
pathology
;
Humans
8.CD43 Expression Regulated by IL-12 Signaling Is Associated with Survival of CD8 T Cells.
Immune Network 2010;10(5):153-163
BACKGROUND: In addition to TCR and costimulatory signals, cytokine signals are required for the differentiation of activated CD8 T cells into memory T cells and their survival. Previously, we have shown that IL-12 priming during initial antigenic stimulation significantly enhanced the survival of activated CD8 T cells and increased the memory cell population. In the present study, we analyzed the mechanisms by which IL-12 priming contributes to activation and survival of CD8 T cells. METHODS: We observed dramatically decreased expression of CD43 in activated CD8 T cells by IL-12 priming. We purified CD43(lo) and CD43(hi) cells after IL-12 priming and analyzed the function and survival of each population both in vivo and in vitro. RESULTS: Compared to CD43(hi) effector cells, CD43(lo) effector CD8 T cells exhibited reduced cytolytic activity and lower granzyme B expression but showed increased survival. CD43(lo) effector CD8 T cells also showed increased in vivo expansion after adoptive transfer and antigen challenge. The enhanced survival of CD43(lo) CD8 T cells was also partly associated with CD62L expression. CONCLUSION: We suggest that CD43 expression regulated by IL-12 priming plays an important role in differentiation and survival of CD8 T cells.
Adoptive Transfer
;
Granzymes
;
Interleukin-12
;
Memory
;
T-Lymphocytes
9.The human application of gene therapy to re-program T-cell specificity using chimeric antigen receptors.
Alan D GUERRERO ; Judy S MOYES ; Laurence J N COOPER
Chinese Journal of Cancer 2014;33(9):421-433
The adoptive transfer of T cells is a promising approach to treat cancers. Primary human T cells can be modified using viral and non-viral vectors to promote the specific targeting of cancer cells via the introduction of exogenous T-cell receptors (TCRs) or chimeric antigen receptors (CARs). This gene transfer displays the potential to increase the specificity and potency of the anticancer response while decreasing the systemic adverse effects that arise from conventional treatments that target both cancerous and healthy cells. This review highlights the generation of clinical-grade T cells expressing CARs for immunotherapy, the use of these cells to target B-cell malignancies and, particularly, the first clinical trials deploying the Sleeping Beauty gene transfer system, which engineers T cells to target CD19+ leukemia and non-Hodgkin's lymphoma.
B-Lymphocytes
;
Gene Transfer Techniques
;
Genetic Therapy
;
Genetic Vectors
;
Humans
;
Immunotherapy, Adoptive
;
Leukemia
;
Lymphoma, B-Cell
;
therapy
;
Lymphoma, Non-Hodgkin
;
therapy
;
Neoplasms
;
Receptors, Antigen
;
Receptors, Antigen, T-Cell
;
Receptors, CCR1
;
T-Cell Antigen Receptor Specificity
;
T-Lymphocytes
10.Targeting the epitope spreader Pep19 by naïve human CD45RA⁺ regulatory T cells dictates a distinct suppressive T cell fate in a novel form of immunotherapy.
Hyun Joo KIM ; Gil Sun CHA ; Ji Young JOO ; Juyoun LEE ; Sung Jo KIM ; Jeongae LEE ; So Youn PARK ; Jeomil CHOI
Journal of Periodontal & Implant Science 2017;47(5):292-311
PURPOSE: Beyond the limited scope of non-specific polyclonal regulatory T cell (Treg)-based immunotherapy, which depends largely on serendipity, the present study explored a target Treg subset appropriate for the delivery of a novel epitope spreader Pep19 antigen as part of a sophisticated form of immunotherapy with defined antigen specificity that induces immune tolerance. METHODS: Human polyclonal CD4⁺CD25⁺CD127(lo−) Tregs (127-Tregs) and naïve CD4⁺CD25⁺CD45RA⁺ Tregs (45RA-Tregs) were isolated and were stimulated with target peptide 19 (Pep19)-pulsed dendritic cells in a tolerogenic milieu followed by ex vivo expansion. Low-dose interleukin-2 (IL-2) and rapamycin were added to selectively exclude the outgrowth of contaminating effector T cells (Teffs). The following parameters were investigated in the expanded antigen-specific Tregs: the distinct expression of the immunosuppressive Treg marker Foxp3, epigenetic stability (demethylation in the Treg-specific demethylated region), the suppression of Teffs, expression of the homing receptors CD62L/CCR7, and CD95L-mediated apoptosis. The expanded Tregs were adoptively transferred into an NOD/scid/IL-2Rγ(−/−) mouse model of collagen-induced arthritis. RESULTS: Epitope-spreader Pep19 targeting by 45RA-Tregs led to an outstanding in vitro suppressive T cell fate characterized by robust ex vivo expansion, the salient expression of Foxp3, high epigenetic stability, enhanced T cell suppression, modest expression of CD62L/CCR7, and higher resistance to CD95L-mediated apoptosis. After adoptive transfer, the distinct fate of these T cells demonstrated a potent in vivo immunotherapeutic capability, as indicated by the complete elimination of footpad swelling, prolonged survival, minimal histopathological changes, and preferential localization of CD4⁺CD25⁺ Tregs at the articular joints in a mechanistic and orchestrated way. CONCLUSIONS: We propose human naïve CD4⁺CD25⁺CD45RA⁺ Tregs and the epitope spreader Pep19 as cellular and molecular targets for a novel antigen-specific Treg-based vaccination against collagen-induced arthritis.
Adoptive Transfer
;
Animals
;
Apoptosis
;
Arthritis, Experimental
;
Arthritis, Rheumatoid
;
Autoimmune Diseases
;
Dendritic Cells
;
Epigenomics
;
Eragrostis
;
Heat-Shock Proteins
;
Humans*
;
Immune Tolerance
;
Immunotherapy*
;
In Vitro Techniques
;
Interleukin-2
;
Joints
;
Mice
;
Sensitivity and Specificity
;
Sirolimus
;
T-Lymphocytes
;
T-Lymphocytes, Regulatory*
;
Vaccination