1.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
2.Allogeneic leukocyte immunization combined with IVF-ET for treatment of infertility induced by recurrent spontaneous abortion.
Ling-hong KONG ; Zhong LIU ; Hong LI ; Si-mei CHEN ; Fu-qi XING
Journal of Southern Medical University 2008;28(7):1300-1301
OBJECTIVETo investigate the effect of allogeneic leukocyte immunization combined with in vitro fertilization-embryo transfer (IVF-ET) for treatment of infertility induced by habitual abortion.
METHODSAllogeneic leukocyte immunization was performed in 9 patients with infertility induced by habitual abortion, with another 9 patients undergoing IVF-ET without habitual abortion as the control group. All the patients were treated with long GnRH-a protocols. The infertility patients with recurrent spontaneous abortion history were immunized with lymphocytes from the husband for before IVF-ET and after clinical pregnancy.
RESULTSThe fertilization rates of the immunotherapy group and control group were 81.3% and 82.2%, respectively, showing no significant difference (P>0.05). Five patients in each group had clinical pregnancy, and a twin pregnancy occurred in the control group. The embryo implantation rates were also comparable between the two groups (22.7% vs 28.6%, P>0.05). All the fetuses resulted from IVF-ET developed normally and were healthily delivered.
CONCLUSIONAllogeneic leukocyte immunotherapy along with IVF-ET is effective for treatment of infertility resulting from recurrent spontaneous abortion.
Abortion, Habitual ; physiopathology ; Adoptive Transfer ; methods ; Adult ; Embryo Transfer ; Female ; Fertilization in Vitro ; methods ; Humans ; Infertility, Female ; physiopathology ; therapy ; Pregnancy ; Pregnancy Outcome ; Treatment Outcome
3.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
4.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
5.An Alternative Dendritic Cell-Induced Murine Model of Asthma Exhibiting a Robust Th2/Th17-Skewed Response
Sang Chul PARK ; Hongmin KIM ; Yeeun BAK ; Dahee SHIM ; Kee Woong KWON ; Chang Hoon KIM ; Joo Heon YOON ; Sung Jae SHIN
Allergy, Asthma & Immunology Research 2020;12(3):537-555
PURPOSE: Simple and reliable animal models of human diseases contribute to the understanding of disease pathogenesis as well as the development of therapeutic interventions. Although several murine models to mimic human asthma have been established, most of them require anesthesia, resulting in variability among test individuals, and do not mimic asthmatic responses accompanied by T-helper (Th) 17 and neutrophils. As dendritic cells (DCs) are known to play an important role in initiating and maintaining asthmatic inflammation, we developed an asthma model via adoptive transfer of allergen-loaded DCs.METHODS: Ovalbumin (OVA)-loaded bone marrow-derived DCs (BMDCs) (OVA-BMDCs) were injected intravenously 3 times into non-anesthetized C57BL/6 mice after intraperitoneal OVA-sensitization.RESULTS: OVA-BMDC-transferred mice developed severe asthmatic immune responses when compared with mice receiving conventional OVA challenge intranasally. Notably, remarkable increases in systemic immunoglobulin (Ig) E and IgG1 responses, Th2/Th17-associated cytokines (interleukin [IL]-5, IL-13 and IL-17), Th2/Th17-skewed T-cell responses, and cellular components, including eosinophils, neutrophils, and goblet cells, were observed in the lungs of OVA-BMDC-transferred mice. Moreover, the asthmatic immune responses and severity of inflammation were correlated with the number of OVA-BMDCs transferred, indicating that the disease severity and asthma type may be adjusted according to the experimental purpose by this method. Furthermore, this model exhibited less variation among the test individuals than the conventional model. In addition, this DCs-based asthma model was partially resistant to steroid treatment.CONCLUSIONS: A reliable murine model of asthma by intravenous (i.v.) transfer of OVA-BMDCs was successfully established without anesthesia. This model more accurately reflects heterogeneous human asthma, exhibiting a robust Th2/Th17-skewed response and eosinophilic/neutrophilic infiltration with good reproducibility and low variation among individuals. This model will be useful for understanding the pathogenesis of asthma and would serve as an alternative tool for immunological studies on the function of DCs, T-cell responses and new drugs.
Adoptive Transfer
;
Anesthesia
;
Animals
;
Asthma
;
Cytokines
;
Dendritic Cells
;
Eosinophils
;
Goblet Cells
;
Humans
;
Immunoglobulin G
;
Immunoglobulins
;
Inflammation
;
Interleukin-13
;
Lung
;
Methods
;
Mice
;
Models, Animal
;
Neutrophils
;
Ovalbumin
;
Ovum
;
T-Lymphocytes
6.In vivo tracing of transferred apoptotic cell labeled using CFSE: a flow cytometry-based assay method.
Yan WANG ; Yi GAO ; Er-wei SUN ; Jin-min XIE ; Hui-ying ZHANG ; Jian-bo CHEN
Journal of Southern Medical University 2006;26(5):599-602
OBJECTIVETo establish an assay method for detecting the migration of transferred apoptotic cells into the recipient using flow cytometry.
METHODSSpleen lymphocytes were isolated and labeled with an intracellular amine dye, carboxyfluorescein diacetate succinimidyl ester (CFSE), to allow discrimination. The labeled cells were induced with dexamethasone to undergo apoptosis and transferred into recipient mice via tail venous transfusion. Flow cytometry and histological examination of different tissues were performed at different time points. The stability of CFSE labeling for apoptotic cells was also tested.
RESULTSThe CFSE-labeled apoptotic cells were highly fluorescent with a positive labeling rate of (98.0+/-1.9)%. The stability of CFSE-labeling was testified, and the CFSE-labeled apoptotic cells entering different tissues at different time points were detected by flow cytometry and verified by histological examination.
CONCLUSIONFlow cytometry using CFSE labeling is reliable, sensitive, precise and convenient for apoptotic cell tracing in vivo and in vitro.
Adoptive Transfer ; methods ; Animals ; Apoptosis ; Dexamethasone ; pharmacology ; Female ; Flow Cytometry ; methods ; Fluoresceins ; chemistry ; pharmacokinetics ; Fluorescent Dyes ; chemistry ; pharmacokinetics ; Lymphocytes ; chemistry ; cytology ; drug effects ; Mice ; Mice, Inbred BALB C ; Reproducibility of Results ; Spleen ; cytology ; Succinimides ; chemistry ; pharmacokinetics
7.Therapeutic effect of autologous cytokine-induced killer cells on patients with liver cirrhosis caused by HBV infection.
Hai-bin SU ; Han-wei LI ; Hong-lan ZHAO ; Ming SHI ; Bing ZHANG ; Zi-rong TANG ; Zhou-yun LEI ; Hui-fen WANG ; Fu-sheng WANG
Chinese Journal of Experimental and Clinical Virology 2007;21(1):64-66
OBJECTIVETo observe the therapeutic effect of autologous cytokine-induced killer cells (CIK) on HBV DNA positive patients with liver cirrhosis.
METHODSHBV DNA positive 33 patients with cirrhosis were treated with CIK. Before and after cultured in vitro and post-treatment, CD3+, CD3+CD4+, CD3+CD8+, CD3+CD56+ cells, mDC and pDC were detected by flow cytometry. The indexes of virus and liver function were compared between pre- and post-treatment.
RESULTSCD3+, CD3+CD8+ cells and CD3+CD56+ cells were higher after cultured in vitro and after transfused back than those before culture (91.5 +/- 10.3, 74.4 +/- 9.9 vs. 67.9 +/- 12.8; 60.9 +/- 15.5, 37.3 +/- 15.1 vs. 27.9 +/- 10.9; 18.4 +/- 11.7, 14.5 +/- 7.5 vs. 10.6 +/- 7.1). The percentages of mDC and pDC also increased after-treatment vs. pre-treatment (0.54 +/- 0.18 vs. 0.70 +/- 0.29; 0.26 +/- 0.13 vs. 0.41 +/- 0.25). HBV DNA became undetectable in 12 patients and decrease exceeded 100 times in 4 patients after treatment. HBeAg became undetectable in 10 of 14 patients who were HBeAg positive pretreatment patients, among them 2 patients had HBeAb sero conversion. The liver function was improved after treatment. All patients tolerated the treatment.
CONCLUSIONCIK treatment can increase immune effector cells and has some antiviral effect and is safe.
Adoptive Transfer ; adverse effects ; methods ; Adult ; Aged ; Cells, Cultured ; Cytokine-Induced Killer Cells ; cytology ; immunology ; transplantation ; Fatigue ; etiology ; Female ; Headache ; etiology ; Hepatitis B ; complications ; virology ; Humans ; Liver Cirrhosis ; etiology ; immunology ; therapy ; Male ; Middle Aged ; Transplantation, Autologous ; Treatment Outcome
8.Efficient amplification of melanoma-specific CD8+ T cells using artificial antigen presenting complex.
Experimental & Molecular Medicine 2006;38(6):591-598
In vitro large amplification of tumor-specific cytotoxic T lymphocytes (CTLs) and adoptive transfer of these cells is one of the most promising approaches to treat malignant diseases in which an effective immune response is not achieved by active immunization. However, generating sufficient numbers of tumor-specific CTLs stimulated with autologous antigen presenting cells (APCs) in vitro is one of the most problematic steps in the adoptive cell transfer (ACT) therapy. To circumvent this problem, we have developed an artificial antigen presenting complex (aAPCs) using MHC class I molecules loaded with a melanoma-specific TRP-2 peptide epitope. Our results show that TRP-2-specific CD8+ T cells elicited by immunization with recombinant adenovirus expressing the mini-gene epitope are efficiently stimulated and amplified in vitro to a greater extent by aAPCs than by natural splenic APCs. These aAPC-induced CTLs recognized endogenously processed antigens present on B16F10 melanoma cells. Efficient stimulation and proliferation of antigen- specific T cells was also confirmed using ovalbumin peptide-loaded aAPCs and OT-I TCR transgenic cells. These results demonstrate that prior in vivo immunization, which increases the precursor frequency, simplifies posterior expansion of tumor- specific CD8+ T cells, and aAPCs is superior to autologous APC for in vitro amplification. This prime and expand regimen can be an alternative method for large amplification of rare tumor-specific CTLs and aAPCs should be a useful tool for ACT immunotherapy.
Substrate Specificity
;
Molecular Sequence Data
;
Mice, Inbred C57BL
;
Mice
;
Melanoma/genetics/*immunology
;
Lymphocyte Count
;
Genetic Vectors/genetics
;
Cell Line, Tumor
;
CD8-Positive T-Lymphocytes/*cytology/*immunology/metabolism
;
Biomimetics/*methods
;
Antigen-Presenting Cells/immunology/metabolism
;
Antigen Presentation/*immunology
;
Animals
;
Amino Acid Sequence
;
Adoptive Transfer/methods
9.Induced tolerance to cardiac allografts with multiple intravenous injection of donor spleen cells.
Hong-Wei GUO ; Qing-Yu WU ; Shu-Sheng XIE ; Qing-Yin ZHANG ; Xiu-Bin YANG ; Meng-Ping SHAO
Chinese Journal of Surgery 2004;42(11):664-667
OBJECTIVETo study the methods and mechanisms of immune tolerance in cardiac transplantation.
METHODSMale DA rat hearts were transplanted to male Lewis rats using Ono's model and randomly divided into five groups: untreated, intravenous injection of 1 x 10(8) DA splenocytes to Lewis rat, intraperitoneal injection of cyclophosphamide (100 mg/kg) to Lewis rat, intravenous injection of 1 x 10(8) DA splenocytes combined with intraperitoneal injection of cyclophosphamide (100 mg/kg) to Lewis rat, multiple injection of DA rat splenocytes with intraperitoneal injection of cyclophosphamide, 11 days later heart transplantation was performed. Mean survival time (MST), histological changes, mixed lymphocyte reaction (MLR), the role of interleukin-2 (IL-2) to MLR and the role of tolerant rat splenocytes to MLR were measured after operation.
RESULTSThe survival time of heart allografts in the group of multiple injection of DA rat splenocytes with intraperitoneal injection of cyclophosphamide [MST: (85.3 +/- 7.5) d, t = 0, P < 0.01] was significantly longer than in the groups of untreated [MST: (7.3 +/- 1.0) d], intravenous injection of 1 x 10(8) DA splenocytes to Lewis rat [MST: (7.9 +/- 0.9) d], intraperitoneal injection of cyclophosphamide (100 mg/kg) to Lewis rat [MST: (8.1 +/- 1.2) d], intravenous injection of 1 x 10(8) DA splenocytes combined with intraperitoneal injection of cyclophosphamide (100 mg/kg) to Lewis rat [MST: (25.8 +/- 3.5) d]. Only a few inflammatory cells infiltrated in cardiac allografts in the group of multiple injection of DA rat splenocytes with intraperitoneal injection of cyclophosphamide. MLR in the group of multiple injection of DA rat splenocytes with intraperitoneal injection of cyclophosphamide were significantly decreased compared with those of normal control (t = 0, P < 0.01). IL-2 could partly reversed the hyporesponsiveness of MLR in tolerant rats, the tolerance could be transferred in vitro.
CONCLUSIONSMultiple injection of donor splenocytes combined with intraperitoneal injection of cyclophosphamide to recipients could induce immune tolerance to cardiac allografts.
Adoptive Transfer ; Animals ; Cell Transplantation ; Cyclophosphamide ; pharmacology ; Graft Enhancement, Immunologic ; methods ; Graft Survival ; Heart Transplantation ; immunology ; Injections, Intravenous ; Isoantigens ; administration & dosage ; immunology ; Male ; Rats ; Rats, Inbred BN ; Rats, Inbred Lew ; Rats, Inbred Strains ; Rats, Wistar ; Spleen ; cytology ; Transplantation Tolerance ; drug effects ; Transplantation, Heterologous ; immunology
10.Mesenchymal stem cell therapy for acute respiratory distress syndrome: from basic to clinics.
Protein & Cell 2020;11(10):707-722
The 2019 novel coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has occurred in China and around the world. SARS-CoV-2-infected patients with severe pneumonia rapidly develop acute respiratory distress syndrome (ARDS) and die of multiple organ failure. Despite advances in supportive care approaches, ARDS is still associated with high mortality and morbidity. Mesenchymal stem cell (MSC)-based therapy may be an potential alternative strategy for treating ARDS by targeting the various pathophysiological events of ARDS. By releasing a variety of paracrine factors and extracellular vesicles, MSC can exert anti-inflammatory, anti-apoptotic, anti-microbial, and pro-angiogenic effects, promote bacterial and alveolar fluid clearance, disrupt the pulmonary endothelial and epithelial cell damage, eventually avoiding the lung and distal organ injuries to rescue patients with ARDS. An increasing number of experimental animal studies and early clinical studies verify the safety and efficacy of MSC therapy in ARDS. Since low cell engraftment and survival in lung limit MSC therapeutic potentials, several strategies have been developed to enhance their engraftment in the lung and their intrinsic, therapeutic properties. Here, we provide a comprehensive review of the mechanisms and optimization of MSC therapy in ARDS and highlighted the potentials and possible barriers of MSC therapy for COVID-19 patients with ARDS.
Adoptive Transfer
;
Alveolar Epithelial Cells
;
pathology
;
Animals
;
Apoptosis
;
Betacoronavirus
;
Body Fluids
;
metabolism
;
CD4-Positive T-Lymphocytes
;
immunology
;
Clinical Trials as Topic
;
Coinfection
;
prevention & control
;
therapy
;
Coronavirus Infections
;
complications
;
immunology
;
Disease Models, Animal
;
Endothelial Cells
;
pathology
;
Extracorporeal Membrane Oxygenation
;
Genetic Therapy
;
methods
;
Genetic Vectors
;
administration & dosage
;
therapeutic use
;
Humans
;
Immunity, Innate
;
Inflammation Mediators
;
metabolism
;
Lung
;
pathology
;
physiopathology
;
Mesenchymal Stem Cell Transplantation
;
methods
;
Mesenchymal Stem Cells
;
physiology
;
Multiple Organ Failure
;
etiology
;
prevention & control
;
Pandemics
;
Pneumonia, Viral
;
complications
;
immunology
;
Respiratory Distress Syndrome, Adult
;
immunology
;
pathology
;
therapy
;
Translational Medical Research