• Title/Summary/Keyword: immune cell therapy

Search Result 265, Processing Time 0.025 seconds

Construction of Glomerular Epithelial Cells Expressing Both Immune Tolerance and GFP Genes and Application to Cell Therapy by Cell Transplantation

  • Ohga, Masahiro;Ogura, Mariko;Matsumura, Mastoshi;Wang, Pi-Chao
    • Biotechnology and Bioprocess Engineering:BBE
    • /
    • v.7 no.5
    • /
    • pp.303-310
    • /
    • 2002
  • Cell therapy applied to wound healing or tissue regeneration presents a revolutionary realm to which principles of gene engineering and delivery may be applied. One promising application is the transplantation of cells into the wounded tissue to help the tissue repair. However, when cells are transplanted from in vitro to in vivo, immune rejection occurs due to the immune response triggered by the activation of T-cell, and the transplanted cells are destroyed by the attack of activated T-cell and lose their function. Immune suppressant such as FK506 is commonly used to suppress immune rejection during transplantation. However, such kind of immune suppressants not only suppresses immune rejection in the periphery of transplanted cells but also suppresses whole immune response system against pathogenic infection. In order to solve this problem, we developed a method to protect the desired cells from immune rejection without impairing whole immune system during cell transplantation. Previously, we reported the success of constructing glomerular epithelial cells for removal of immune complex, in which complement receptor of type 1 (CR1) was over-expressed on the membrane of renal glomerular epithelial cells and could bind immune complex of DNA/anti-DNA-antibody to remove immune complex through phagocy-tosis [1]. Attempting to apply the CR1-expressing cells to cell therapy and evade immune rejection during cell transplantation, we constructed three plasmids containing genes encoding a soluble fusion protein of cytolytic T lymphocyte associated antigen-4 (CTLA4Ig) and an enhanced green fluorescent protein (EGFP). The plasmids were transfected to the above-mentioned glomerular epithelial cells to express both genes simultaneously. Using the clone cells for cell transplantation showed that mice with autoimmune disease prolonged their life significantly as compared with the control mice, and two injections of the cells at the beginning of two weeks resulted in remarkable survivability, whereas it requires half a year and 50 administrations of proteins purified from the same amount of cells to achieve the same effect.

Development and Effects of Horticulture Therapy on Quality of Sleep and Immune Function in Patients in Hospice Units (병동형 호스피스 환자를 위한 원예요법의 개발 및 수면의 질과 면역기능에 미치는 효과)

  • Kim, Chung Nam;Song, Mi Ok;Kwon, Yun Hee
    • Journal of Korean Clinical Nursing Research
    • /
    • v.16 no.3
    • /
    • pp.39-50
    • /
    • 2010
  • Purpose: The study was done to examine the development and effects of horticulture therapy on quality of sleep and immune function in patients in hospice units. Methods: The participants in this study were hospice patients in D hospital in D city. Thirty hospice patients were assigned to the experimental group, thirty to the control group. Data were collected from April 29 to July 26, 2009. The horticulture therapy program consisted of indoor and outdoor horticultural activities. The horticulture therapy was conducted for 30 minutes, 6 times a week for 3 weeks (a total 18 times). Measures were quality of sleep, and immune function by serum T-cell, NK-cell count. Data were analyzed using descriptive statistics, chi-square test and t-test with SPSS/WIN 13.0 version. Results: Patients in the experimental group receiving horticulture therapy had a significant difference in changes in the quality of sleep compared to the control group. There were also a significant difference in changes in the immune function (serum T cell and serum NK cell) between the experimental group and control group. Conclusion: The study results indicate that horticulture therapy developed for hospice patients is an effective, palliative intervention program to improve the quality of sleep and immune function of hospice patients.

A literature study on oncological Immune therapy (암(癌)의 면역치료(免疫治療)에 대(對)한 고찰(考察))

  • Park, Jong-Hak;Son, Chang-Gyu;Cho, Chong-kwan
    • Journal of Haehwa Medicine
    • /
    • v.9 no.2
    • /
    • pp.211-221
    • /
    • 2001
  • A literature study on oncological immune therapy was done, and the results were as follows. 1. Oncological immune therapy is classified as specific non specific therapy or active inactive therapy, and in tumor immune response, cellular immunity operates mainly, so activity of T lymphocytes and macrophages are closely related with growth, progress, metastasis and prospect of tumor. Recently, Immune therapies of gene which use cytokines and HLA-B7 are carrying out. 2. In oriental medicine, development of disease is closely related to up and down of healthy qi, so healthy qi operates as a immune factor and resistance factor. 3. On the base of theory "Increasing healthy qi reduces mass(養正則積自除)", strengthening body resistance is emphasized in cancer therapy. Also strengthening body resistance activates cellular immune response and promote killing tumor facility of T-cell. 4. In clinical view, using immune therapy after operation, radiation, and chemotheraphy is more effective than immune therapy itself, so it is expected that east-west cooperation will be effective in cancer therapy. 5. The study of oncological immunity is progressed on emphasizing T-cell and it is related to oriental medical theory "strengthening healthy qi to eliminate pathogen(扶定祛邪)" and advanced study is expected in future.

  • PDF

Ex vivo Boosted Immune Cell Therapy for Canine Hepatic Disease

  • Bae, Seulgi;Oh, Taeho
    • Journal of Veterinary Clinics
    • /
    • v.38 no.4
    • /
    • pp.179-183
    • /
    • 2021
  • A 12-year-old male American Cocker Spaniel was diagnosed with a type of chronic hepatits (CH) called cholangioheaptits. Routine supportive medication was administered to the patient, and ex vivo boosted immune cell (EBI-C) therapy was used for the treatment. A histopathologic examination of the liver 19 months later revealed that the cholangiohepatitis had progressed to cholangiocarcinoma. The medication and immune cell therapy was maintained. Two months after the new diagnosis, the patient's state worsened, and the dog died 635 days after the first visit. EBI-C therapy is a type of immunotherapy, where immune cells are isolated from the patient's peripheral blood mononuclear cells, expanded ex vivo, and then infused into the patient intravenously every two weeks. EBI-Cs (mean: 2.78 × 108 cells) were obtained 38 times and infused every two weeks. Most EBI-C were T-lymphocytes (99.24% of total EBI cells). T-lymphocytes produce large interferon (IFN)-γ, and IFN-γ inhibits liver fibrosis in dogs with CH. Moreover, in bile duct cancer, an increase in T-lymphocytes correlates with decreasing tumor invasion and metastasis. Thus, we propose that EBI-C therapy is applicable as a new supportive therapy for canine liver disease if other treatments like drug medication, surgery, or radiation are unavailable.

Immunocell Therapy for Lung Cancer: Dendritic Cell Based Adjuvant Therapy in Mouse Lung Cancer Model (폐암의 면역세포 치료: 동물 모델에서 수지상 세포를 이용한 Adjuvant Therapy 가능성 연구)

  • Lee, Seog-Jae;Kim, Myung-Joo;In, So-Hee;Baek, So-Young;Lee, Hyun-Ah
    • IMMUNE NETWORK
    • /
    • v.5 no.1
    • /
    • pp.36-44
    • /
    • 2005
  • Background: The anti-tumor therapeutic effect of autologous tumor cell lysate pulseddendritic cells (DCs) was studied for non-immunogenic and immune suppressive lung cancer model. To test the possibility as an adjuvant therapy, minimal residual disease model was considered in mouse in vivo experiments. Methods: Syngeneic 3LL lung cancer cells were inoculated intravenously into the C57BL/6 mouse. Autologous tumor cell (3LL) or allogeneic leukemia cell (WEHI-3) lysate pulsed-DCs were injected twice in two weeks. Intraperitoneal DC injection was started one day (MRD model) after tumor cell inoculation. Two weeks after the final DC injection, tumor formation in the lung and the tumor-specific systemic immunity were observed. Tumor-specific lymphocyte proliferation and the IFN-${\gamma}$ secretion were analyzed for the immune monitoring. Therapeutic DCs were cultured from the bone marrow myeloid lineage cells with GM-CSF and IL-4 for 7 days and pulsed with tumor cell lysate for 18 hrs. Results: Compared to the saline treated group, tumor formation was suppressed in 3LL tumor cell lysate pulsed-DC treated group, while 3LL-specific immune stimulation was minimum. WEHI-3-specific immune stimulation occurred in WEHI-3 lysate-pulsed DC treated group, which had no correlation with tumor regression. Conclusion: The data suggest the possible anti-tumor effect of cultured DCs as an adjuvant therapy for minimal residual disease state of lung cancer. The significance of immune modulation in DC therapy including the possible involvement of NK cell as well as antigen-specific cytotoxic T cell activity induction was discussed.

Post-transcriptional Regulation of NK Cell Activation

  • Kim, Tae-Don;Park, Ju-Yeong;Choi, In-Pyo
    • IMMUNE NETWORK
    • /
    • v.9 no.4
    • /
    • pp.115-121
    • /
    • 2009
  • Natural killer (NK) cells play key roles in innate and adaptive immune defenses. NK cell responses are mediated by two major mechanisms: the direct cytolysis of target cells, and immune regulation by production of various cytokines. Many previous reports show that the complex NK cell activation process requires de novo gene expression regulated at both transcriptional and post-transcriptional levels. Specialized un-translated regions (UTR) of mRNAs are the main mechanisms of post-transcriptional regulation. Analysis of posttranscriptional regulation is needed to clearly understand NK cell biology and, furthermore, harness the power of NK cells for therapeutic aims. This review summarizes the current understanding of mRNA metabolism during NK cell activation, focusing primarily on post-transcriptional regulation.

Effects of Laughter Therapy on Depression, Quality of Life, Resilience and Immune Responses in Breast Cancer Survivors (웃음요법이 유방암 생존자의 우울, 삶의 질, 극복력, 면역반응에 미치는 효과)

  • Cho, Eun-A;Oh, Hyun-Ei
    • Journal of Korean Academy of Nursing
    • /
    • v.41 no.3
    • /
    • pp.285-293
    • /
    • 2011
  • Purpose: In this study, the effects of laughter therapy on levels of depression, quality of life, resilience and immune responses in breast cancer survivors were examined. Methods: A quasi-experimental nonequivalent control group, pretest-posttest design was used. Participants (n=37) included breast cancer survivors who finished chemotheraphy and radiation therapy: 16 in the experiment group and 21 in the control group. Data were collected from August to November 2009. The experimental group participated in laughter therapy eight times, twice a week for 60 min per session. Questionnaires were used to me-asure pretest and posttest levels of depression, quality of life and resilience. A blood test was used to analyze changes in Total T cell, T helper, T suppressor, Th/Ts ratio, Total B cell, T cell/B cell ratio and NK cell for immune responses. Results: The results showed that laughter therapy was effective in increasing the quality of life and resilience in breast cancer survivors. but depression and immune responses did not differ significantly between the groups. Conclusion: The results of the study indicate that laughter therapy may be an effective nursing intervention to improve quality of life and resilience in breast cancer survivors.

Impact of Cellular Immune Function on Prognosis of Lung Cancer Patients after Cytokine-induced Killer Cell Therapy

  • Jin, Congguo;Li, Jia;Wang, Yeying;Chen, Xiaoqun;Che, Yanhua;Liu, Xin;Wang, Xicai;Sriplung, Hutcha
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.15
    • /
    • pp.6009-6014
    • /
    • 2014
  • Aims: To investigate changes in cellular immune function of patients with lung cancer before and after cytokine-induced killer (CIK) cell therapy and to identify variation effects on overall survival (OS) and progression-free survival (PFS). Materials and Methods:A total of 943 lung cancer patients with immune dysfunction were recruited from January 2002 to January 2010, 532 being allocated to conventional therapy and 411 to CIK therapy after a standard treatment according to the NCCN Clinical Practice Guidelines. All the patients were investigated for cellular immune function before and after therapy every three months. and clinical prognostic outcomes were analyzed. Results: After six courses of treatment, immune function was much improved in patients receiving CIK cells therapy as compared to controls. The percentages of recurrence and/or metastases for patients undergoing CIK cell therapy was 56.2% and 49.1% respectively but 78.6% and 70.3% among controls (p<0.001). The median OS times for CIK cell therapy and control groups were 48 and 36 months respectively. The OS rates at 12, 36, 60, 84 months in CIK treated patients were 97.8%, 66.9%, 27.7%, and 4.1% while they were 92.3%, 44.5%, 9.2%, and 1.5% in controls. OS and PFS were significantly different by log rank test between the two groups and across the three immune improvement classes. Conclusions: The immune function of lung cancer patients was improved by CIK cell therapy, associated with an increase in the OS rate and extension of the time to recurrence and/or metastasis.

Peripheral Blood Immune Cell-based Biomarkers in Anti-PD-1/PD-L1 Therapy

  • Kyung Hwan Kim;Chang Gon Kim;Eui-Cheol Shin
    • IMMUNE NETWORK
    • /
    • v.20 no.1
    • /
    • pp.8.1-8.15
    • /
    • 2020
  • Immune checkpoint blockade targeting PD-1 and PD-L1 has resulted in unprecedented clinical benefit for cancer patients. Anti-PD-1/PD-L1 therapy has become the standard treatment for diverse cancer types as monotherapy or in combination with other anticancer therapies, and its indications are expanding. However, many patients do not benefit from anti-PD-1/PD-L1 therapy due to primary and/or acquired resistance, which is a major obstacle to broadening the clinical applicability of anti-PD-1/PD-L1 therapy. In addition, hyperprogressive disease, an acceleration of tumor growth following anti-PD-1/PD-L1 therapy, has been proposed as a new response pattern associated with deleterious prognosis. Anti-PD-1/PD-L1 therapy can also cause a unique pattern of adverse events termed immune-related adverse events, sometimes leading to treatment discontinuation and fatal outcomes. Investigations have been carried out to predict and monitor treatment outcomes using peripheral blood as an alternative to tissue biopsy. This review summarizes recent studies utilizing peripheral blood immune cells to predict various outcomes in cancer patients treated with anti-PD-1/PD-L1 therapy.

Immune cell-derived small extracellular vesicles in cancer treatment

  • Choi, Sung-Jin;Cho, Hanchae;Yea, Kyungmoo;Baek, Moon-Chang
    • BMB Reports
    • /
    • v.55 no.1
    • /
    • pp.48-56
    • /
    • 2022
  • Small extracellular vesicles (sEVs) secreted by most cells carry bioactive macromolecules including proteins, lipids, and nucleic acids for intercellular communication. Given that some immune cell-derived sEVs exhibit anti-cancer properties, these sEVs have received scientific attention for the development of novel anti-cancer immunotherapeutic agents. In this paper, we reviewed the latest advances concerning the biological roles of immune cell-derived sEVs for cancer therapy. sEVs derived from immune cells including dendritic cells (DCs), T cells, natural-killer (NK) cells, and macrophages are good candidates for sEV-based cancer therapy. Besides their role of cancer vaccines, DC-shed sEVs activated cytotoxic lymphocytes and killed tumor cells. sEVs isolated from NK cells and chimeric antigen receptor (CAR) T cells exhibited cytotoxicity against cancer cells. sEVs derived from CD8+ T and CD4+ T cells inhibited cancer-associated cells in tumor microenvironment (TME) and activated B cells, respectively. M1-macrophage-derived sEVs induced M2 to M1 repolarization and also created a pro-inflammatory environment. Hence, these sEVs, via mono or combination therapy, could be considered in the treatment of cancer patients in the future. In addition, sEVs derived from cytokine-stimulated immune cells or sEV engineering could improve their anti-tumor potency.