1
|
Ramachandran K. An Immunohistochemical Study on the Role of CD83+ Dendritic Cells (DCs) in Malignant and Benign Lesions of the Human Cervix. Cureus 2024; 16:e71327. [PMID: 39529763 PMCID: PMC11554423 DOI: 10.7759/cureus.71327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background Dendritic cells (DCs) are a group of cells that mainly function as antigen-presenting cells in the human body. Proper knowledge and understanding of such cells in the human cervix would be beneficial for understanding the role of CD83+ cells in benign and malignant lesions of the cervix. Materials and methods This retrospective study was performed on cervical specimens. After processing, the CD83+ cells were counted for every 20 high-power fields. The average count per high power field (HPF) was then calculated. The CD83+ cell distributions in cervicitis, cervical dysplasia, and cervical carcinoma were then analyzed. Results A total of 30 cervical specimens were studied. Of these, 16 were cervicitis and seven were squamous cell carcinoma. Vaginal bleeding was the most common presentation in 21 patients. The mean age was 44.7 years. The mean CD83+ DCs in benign lesions was 1.75 and in malignant tissues was 12.26 per HPF (P<0.001). The area under the curve suggested a 100% sensitivity and specificity of CD83 in distinguishing benign and malignant lesions. The receiver operating characteristic (ROC) curve indicated that the probability of malignancy is higher if the number of CD83+ DCS is more than 179.50/20 HPF. Conclusions Dendritic cells play a major role in the tumoricidal activities of the host cervical tissues. Malignant cervical tissue possesses a higher concentration of CD83+ DCs than benign ones, with 100% sensitivity and specificity. This research work on CD83+ DCs in the cervix would pave the way for further research on the immune functions of the human body.
Collapse
Affiliation(s)
- Kalpana Ramachandran
- Anatomy, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| |
Collapse
|
2
|
Jiang L, Dong R, Ying M, He Q, Cao J, Yang B. Immune cells in the tumour: new routes of retinoids for chemoprevention and chemotherapeutics. Br J Pharmacol 2018; 175:4285-4294. [PMID: 30298911 DOI: 10.1111/bph.14511] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/18/2018] [Accepted: 09/24/2018] [Indexed: 01/27/2023] Open
Abstract
Retinoids, vitamin A and its natural and synthetic analogues have various functions, including being involved in cell proliferation and differentiation and participating in the formation of vertebrate morphology. In addition, they may activate certain tumour suppressor genes that then act as tumour inhibitors. In the past decades, retinoids have been regarded as promising chemotherapeutic and chemopreventive agents; however, their mechanisms are still not fully understood. Immune cells that participate in or are associated with the immune response play vital roles in the initiation and development of many cancers. Interestingly, recent studies have demonstrated that retinoids can also exert various effects on immune cells including macrophages, T cells and dendritic cells in tumour tissues to execute anti-tumour actions, providing new insights into chemoprevention and chemotherapeutics. In this review, we focus on the effects of retinoids on immune cells in the tumour, which may provide new approaches for antineoplastic strategies.
Collapse
Affiliation(s)
- Li Jiang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Rong Dong
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Vo MC, Yang S, Jung SH, Chu TH, Lee HJ, Lakshmi TJ, Park HS, Kim HJ, Lee JJ. Synergistic Antimyeloma Activity of Dendritic Cells and Pomalidomide in a Murine Myeloma Model. Front Immunol 2018; 9:1798. [PMID: 30123221 PMCID: PMC6085413 DOI: 10.3389/fimmu.2018.01798] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 07/20/2018] [Indexed: 01/14/2023] Open
Abstract
We have previously shown that immunization with tumor antigen-loaded dendritic cells (DCs) and the immunomodulating drug, lenalidomide, synergistically potentiates the enhancing antitumor immunity in a myeloma mouse model. In this study, we investigated the immunogenicity of DCs combined with pomalidomide and dexamethasone in a myeloma mouse model. MOPC-315 cells were injected subcutaneously to establish myeloma-bearing mice. Four test groups were used to mimic clinical protocol: (1) PBS control, (2) DCs, (3) pomalidomide + dexamethasone, and (4) DCs + pomalidomide + dexamethasone. The combination of DCs plus pomalidomide and dexamethasone displayed greater inhibition of tumor growth compared to the other groups. This effect was closely related with reduced numbers of immune suppressor cells including myeloid-derived suppressor cells, M2 macrophages, and regulatory T cells, with the induction of immune effector cells such as CD4+ and CD8+ T cells, memory T cells, natural killer (NK) cells, and M1 macrophages, and with the activation of T lymphocytes and NK cells in the spleen. Moreover, the level of the immunosuppressive factor vascular endothelial growth factor was significantly reduced in the tumor microenvironment. The collective findings in the murine myeloma model suggest that tumor antigen-loaded DCs combined with pomalidomide and dexamethasone synergistically enhance antitumor immunity by skewing the immune-suppressive status toward an immune-supportive status.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Seoyun Yang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Tan-Huy Chu
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyun-Ju Lee
- Vaxcell-Bio Therapeutics, Hwasun, South Korea
| | - Thangaraj Jaya Lakshmi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hye-Seong Park
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Vaxcell-Bio Therapeutics, Hwasun, South Korea
| |
Collapse
|
4
|
Vo MC, Jung SH, Chu TH, Lee HJ, Lakshmi TJ, Park HS, Kim HJ, Rhee JH, Lee JJ. Lenalidomide and Programmed Death-1 Blockade Synergistically Enhances the Effects of Dendritic Cell Vaccination in a Model of Murine Myeloma. Front Immunol 2018; 9:1370. [PMID: 29967612 PMCID: PMC6015916 DOI: 10.3389/fimmu.2018.01370] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/01/2018] [Indexed: 01/22/2023] Open
Abstract
The therapeutic efficacy of dendritic cell (DC)-based immunotherapy may be potentiated in combination with other anticancer therapies that enhance DC function by modulating immune responses and the tumor microenvironment. In this study, we investigated the efficacy of DC vaccination in combination with lenalidomide and programmed death (PD)-1 blockade in a model of murine myeloma. MOPC-315 cell lines were injected subcutaneously to establish myeloma-bearing mice and the following five test groups were established: PBS control, DCs, DCs + lenalidomide, DCs + PD-1 blockade, and DCs + lenalidomide + PD-1 blockade. The combination of DCs plus lenalidomide and PD-1 blockade more potently inhibited tumor growth compared to the other groups. This effect was associated with a reduction in immune suppressor cells (such as myeloid-derived suppressor cells, M2 macrophages, and regulatory T cells) and an increase in immune effector cells [such as CD4+ and CD8+ T cells, natural killer (NK) cells, and M1 macrophages] in the spleen. Functional activities of cytotoxic T lymphocytes and NK cells were also enhanced by the triple combination. Levels of immunosuppressive cytokines, such as TGF-β and IL-10, were significantly reduced in the tumor microenvironment. These findings suggest that the combination of DCs plus lenalidomide and PD-1 blockade synergistically establishes a robust anti-myeloma immunity through a two-way mechanism, which inhibits immunosuppressive cells while activating effector cells with superior polarization of the Th1/Th2 balance in favor of the tumor immune response. This result should provide an experimental ground for incorporating check point inhibitors to existing immunotherapeutic modalities against multiple myeloma.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea
| | - Tan-Huy Chu
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyun-Ju Lee
- Research Institute, VaxCell-Bio Therapeutics, Hwasun, South Korea
| | - Thangaraj Jaya Lakshmi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hye-Seong Park
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea
| | - Joon Haeng Rhee
- Research Institute, VaxCell-Bio Therapeutics, Hwasun, South Korea.,Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Hwasun, South Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, South Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital and Chonnam National University Medical School, Hwasun, South Korea.,Research Institute, VaxCell-Bio Therapeutics, Hwasun, South Korea
| |
Collapse
|
5
|
Vo MC, Nguyen-Pham TN, Lee HJ, Jung SH, Choi NR, Hoang MD, Kim HJ, Lee JJ. Chaetocin enhances dendritic cell function via the induction of heat shock protein and cancer testis antigens in myeloma cells. Oncotarget 2018; 8:46047-46056. [PMID: 28512265 PMCID: PMC5542247 DOI: 10.18632/oncotarget.17517] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 03/24/2017] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC)-based vaccines are considered useful in cancer immuno-therapy, and the interactions of DC and dying tumor cells are important and promising for cancer immunotherapy. We investigated whether chaetocin could be used to induce death of myeloma cells, for loading onto DCs can affect DCs function. In this study, we show that the dying myeloma cells treated with chaetocin resulted in the induction of heat shock protein (HSP) 90, which was inhibited by antioxidant N-acetyl cysteine, and showed an increase in the expression of MAGE-A3 and MAGE-C1/CT7. DCs loaded with chaetocin-treated dying myeloma cells produced low levels of IL-10 and enhanced the cross presentation of DCs. Additionally, these DCs most potently inhibited regulatory T cells, induced Th1 polarization and activated myeloma-specific cytotoxic T lymphocytes compared with DCs loaded with UVB-irradiated dying myeloma cells. These results suggest that the pretreatment of myeloma cells with chaetocin can enhance DC function through the up-regulation of HSP90 and cancer testis antigens in dying myeloma cells and can potently induce the Th1 polarization of DCs and myeloma-specific cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Thanh-Nhan Nguyen-Pham
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Nu-Ri Choi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - My-Dung Hoang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| |
Collapse
|
6
|
Vo MC, Nguyen-Pham TN, Lee HJ, Jaya Lakshmi T, Yang S, Jung SH, Kim HJ, Lee JJ. Combination therapy with dendritic cells and lenalidomide is an effective approach to enhance antitumor immunity in a mouse colon cancer model. Oncotarget 2018; 8:27252-27262. [PMID: 28460478 PMCID: PMC5432332 DOI: 10.18632/oncotarget.15917] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 02/20/2017] [Indexed: 11/25/2022] Open
Abstract
In this study, we investigated efficacy of lenalidomide in combination with tumor antigen-loaded dendritic cells (DCs) in murine colon cancer model. MC-38 cell lines were injected subcutaneously to establish colon cancer-bearing mice. After tumor growth, lenalidomide (50 mg/kg/day) was injected intraperitoneally on 3 consecutive days in combination with tumor antigen-loaded DC vaccination on days 8, 12, 16, and 20. The tumor antigen-loaded DCs plus lenalidomide combination treatment exhibited a significant inhibition of tumor growth compared with the other groups. These effects were associated with a reduction in immune suppressor cells, such as myeloid-derived suppressor cells and regulatory T cells, with the induction of immune effector cells, such as natural killer cells, CD4+ T cells and CD8+ T cells in spleen, and with the activation of cytotoxic T lymphocytes and NK cells. This study suggests that a combination of tumor antigen-loaded DC vaccination and lenalidomide synergistically enhanced antitumor immune response in the murine colon cancer model, by inhibiting the generation of immune suppressive cells and recovery of effector cells, and demonstrated superior polarization of Th1/Th2 balance in favor of Th1 immune response. This combination approach with DCs and lenalidomide may provide a new therapeutic option to improve the treatment of colon cancer.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Thanh-Nhan Nguyen-Pham
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Thangaraj Jaya Lakshmi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Seoyun Yang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| |
Collapse
|
7
|
Cellular Engineering for the Production of New Blood Components. Transfus Med 2016. [DOI: 10.1002/9781119236504.ch18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
8
|
Vo MC, Lee HJ, Kim JS, Hoang MD, Choi NR, Rhee JH, Lakshmanan VK, Shin SJ, Lee JJ. Dendritic cell vaccination with a toll-like receptor agonist derived from mycobacteria enhances anti-tumor immunity. Oncotarget 2016; 6:33781-90. [PMID: 26418952 PMCID: PMC4741802 DOI: 10.18632/oncotarget.5281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/04/2015] [Indexed: 11/30/2022] Open
Abstract
Dendritic cell (DC)-based vaccines are considered useful in cancer immunotherapy, and the interaction of DC and adjuvants is important in the design of the next generation vaccines. In this study, whether DC combined with Rv2299c derived from mycobacteria could improve anti-tumor immune responses in a colon cancer mouse model was evaluated. MC38 cell lines were injected subcutaneously to establish colon-cancer-bearing mice and the following four groups were evaluated: PBS control, tumor antigen (TA) loaded-DC, Rv2299c, and a combination of TA-loaded-DC and Rv2299c. The combination treatment with TA-loaded-DC and Rv2299c exhibited greater inhibition of tumor growth compared to other groups. These effects were associated with the reduction of suppressor cells, such as myeloid-derived suppressor cells and regulatory T cells, and the induction of effector cells, such as CD4+ T cells and CD8+ T cells, in spleen, and with the activation of cytotoxic T Lymphocytes and NK cells. These results suggest that TA-loaded-DC vaccination with Rv2299c derived from mycobacteria enhanced anti-tumor immunity in a mouse colon cancer model by inhibiting the generation of immune-suppressive cells and recovering numbers of effector cells, and demonstrated superior polarization of the Th1/Th2 balance in favor of the Th1 immune response.
Collapse
Affiliation(s)
- Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea
| | - Jong-Seok Kim
- Department of Microbiology and Institute of Immunology and Immunological Diseases, Yonsei University, Seoul, Republic of Korea
| | - My-Dung Hoang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea
| | - Nu-Ri Choi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea
| | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Vinoth-Kumar Lakshmanan
- Department of Biomedical Science, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sung-Jae Shin
- Department of Microbiology and Institute of Immunology and Immunological Diseases, Yonsei University, Seoul, Republic of Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Jeollanamdo, Republic of Korea
| |
Collapse
|
9
|
Qian ZD, Huang LL, Zhu XM. An immunohistochemical study of CD83- and CD1a-positive dendritic cells in the decidua of women with recurrent spontaneous abortion. Eur J Med Res 2015; 20:2. [PMID: 25563385 PMCID: PMC4301856 DOI: 10.1186/s40001-014-0076-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 12/08/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are more and more women with recurrent spontaneous abortion (RSA). The mechanism of RSA is still unclear. Immunological factors have been postulated to play a role in the etiology of RSA. Dendritic cells (DCs) are the most potent antigen-presenting cells in the immune system, and the decidual DCs may take part in the occurrence of RSA. The difference in maturity status of decidual DCs among women with RSA and women with normal pregnancies is worthy of studying for its application to prevention and therapy. METHODS The EnVision two-step immunohistochemical staining technique was used to detect the expression of CD83 and CD1a in the decidua of women with RSA (30 cases) and normal pregnancies (30 cases). The maturity status, distribution and quantity of DCs in the two groups were observed. Observation of the staining and cell counting were done using microscope within 30 randomly selected high-power fields (HPF, 40 × 10). All data analyses were conducted with SPSS 17.0 and the statistical significance was set at P <0.05. RESULTS The decidua from the two groups contained DCs that stained with the anti-CD83 and anti-CD1a antibody. Most of the decidual CD83(+)DCs from two groups were located in the stroma. There were more CD83(+)DCs clustered with other DCs in the stroma from women with RSA than normal pregnancies. Most of the CD1a(+)DCs in the decidua from the two groups are located close to maternal glandular epithelium. No difference in the location of CD1a(+)DCs was found in the decidua between two groups. The number of decidual CD83(+)DCs was statistically significantly higher in RSA women than in normal early pregnant women (14.20 ± 13.34/30 HPF versus 4.77 ± 2.64/30 HPF; t = 3.800, P = 0.001). The number of CD1a(+)DCs in the decidua was statistically significantly lower in RSA women compared with normal early pregnant women (3.97 ± 3.75/30 HPF versus 7.60 ± 6.08/30 HPF; t = 2.786, P = 0.008). CONCLUSIONS These findings suggest that the increase in the number of mature DCs and the decrease in the quantity of immature DCs in the decidua may be related to RSA. The maturation of decidual DCs may play an important role in the pathogenesis of RSA.
Collapse
Affiliation(s)
- Zhi-Da Qian
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Hangzhou, Zhejiang Province, 310006, People's Republic of China.
| | - Li-Li Huang
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Hangzhou, Zhejiang Province, 310006, People's Republic of China.
| | - Xiao-Ming Zhu
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, 1 Xueshi Road, Hangzhou, Zhejiang Province, 310006, People's Republic of China.
- Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, People's Republic of China.
| |
Collapse
|
10
|
Abstract
Dendritic cells (DCs) are the most powerful professional antigen-presenting cells and are unique in their capability to initiate, maintain and regulate the intensity of primary immune responses, including specific antitumor responses. Development of practical procedures to prepare sufficient numbers of functional human DCs in culture from the peripheral blood precursors, paved the way for clinical trials to evaluate various DC-based strategies in patients with malignant diseases. However, no definite conclusions regarding the clinical and even immunological efficacy of DC vaccination can be stated, despite the fact that 12 years have passed since the first clinical trial utilizing DCs in cancer patients. Many unanswered questions hamper the development of DC-based vaccines, including the source of DC preparation and protocols for DC generation, activation and loading with tumor antigens, source of tumor antigens, route of vaccine administration and methods of immunomonitoring. Fortunately, in spite of the many obstacles, DC vaccines continue to hold promise for cancer therapy.
Collapse
Affiliation(s)
- Hua Zhong
- Shanghai Jiao Tong University, Shanghai Chest Hospital, 241 Huaihai Road (w), Shanghai 200030, China.
| | | | | |
Collapse
|
11
|
Nguyen-Pham TN, Lee YK, Lee HJ, Kim MH, Yang DH, Kim HJ, Lee JJ. Cellular immunotherapy using dendritic cells against multiple myeloma. THE KOREAN JOURNAL OF HEMATOLOGY 2012; 47:17-27. [PMID: 22479274 PMCID: PMC3317466 DOI: 10.5045/kjh.2012.47.1.17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 02/09/2012] [Accepted: 03/02/2012] [Indexed: 11/17/2022]
Abstract
Cellular therapy with dendritic cells (DCs) is emerging as a useful immunotherapeutic tool to treat multiple myeloma (MM). DC-based idiotype vaccination was recently suggested to induce idiotype-specific immune responses in MM patients. However, the clinical results so far have been largely disappointing, and the clinical effectiveness of such vaccinations in MM still needs to be demonstrated. DC-based therapies against MM may need to be boosted with other sources of tumor-associated antigens, and potent DCs should be recruited to increase the effectiveness of treatment. DCs with both high migratory capacity and high cytokine production are very important for effective DC-based cancer vaccination in order to induce high numbers of Th1-type CD4+ T cells and CD8+ cytotoxic T lymphocytes. The tumor microenvironment is also important in the regulation of tumor cell growth, proliferation, and the development of therapeutic resistance after treatment. In this review, we discuss how the efficacy of DC vaccination in MM can be improved. In addition, novel treatment strategies that target not only myeloma cells but also the tumor microenvironment are urgently needed to improve treatment outcomes.
Collapse
Affiliation(s)
- Thanh-Nhan Nguyen-Pham
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | | | | | | | | | | | | |
Collapse
|
12
|
Immunotherapy using dendritic cells against multiple myeloma: how to improve? Clin Dev Immunol 2012; 2012:397648. [PMID: 22481968 PMCID: PMC3312256 DOI: 10.1155/2012/397648] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 01/02/2012] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a good target disease in which one can apply cellular immunotherapy, which is based on the graft-versus-myeloma effect. This role of immune effector cells provides the framework for the development of immune-based therapeutic options that use antigen-presenting cells (APCs) with increased potency, such as dendritic cells (DCs), in MM. Current isolated idiotype (Id), myeloma cell lysates, myeloma dying cells, DC-myeloma hybrids, or DC transfected with tumor-derived RNA has been used for immunotherapy with DCs. Immunological inhibitory cytokines, such as TGF-β, IL-10, IL-6 and VEGF, which are produced from myeloma cells, can modulate antitumor host immune response, including the abrogation of DC function, by constitutive activation of STAT3. Therefore, even the immune responses have been observed in clinical trials, the clinical response was rarely improved following DC vaccinations in MM patients. We are going to discuss how to improve the efficacy of DC vaccination in MM.
Collapse
|
13
|
Cellular Engineering for the Production of New Blood Components. Transfus Med 2011. [DOI: 10.1002/9781444398748.ch18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
14
|
Park MH, Yang DH, Kim MH, Jang JH, Jang YY, Lee YK, Jin CJ, Pham TNN, Thi TAN, Lim MS, Lee HJ, Hong CY, Yoon JH, Lee JJ. Alpha-Type 1 Polarized Dendritic Cells Loaded with Apoptotic Allogeneic Breast Cancer Cells Can Induce Potent Cytotoxic T Lymphocytes against Breast Cancer. Cancer Res Treat 2011; 43:56-66. [PMID: 21509164 PMCID: PMC3072536 DOI: 10.4143/crt.2011.43.1.56] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Accepted: 08/05/2010] [Indexed: 01/08/2023] Open
Abstract
Purpose Various tumor antigens can be loaded onto dendritic cells (DCs) to induce a potent cytotoxic T lymphocyte (CTL) response in DC-based immunotherapy against breast cancer. However, in the clinical setting, obtaining a sufficient number of autologous tumor cells as a source of tumor antigens is a laborious process. We therefore investigated the feasibility of immunotherapy using breast-cancer-specific CTLs generated in vitro by use of alpha-type 1 polarized DCs (α DC1s) loaded with ultraviolet B-irradiated cells of the breast cancer cell line MCF-7. Materials and Methods αDC1s were induced by loading allogeneic tumor antigen generated from the MCF-7 UVB-irradiated breast cancer cell line. Antigen-pulsed αDC1s were evaluated by morphological and functional assays, and the breast-cancer-specific CTL response was analyzed by cytotoxic assay. Results The αDC1s significantly increased the expression of several molecules related to DC maturation without differences according to whether the αDC1s were loaded with tumor antigens. The αDC1s showed a high production of interleukin-12 both during maturation and after subsequent stimulation with CD40L, which was not significantly affected by loading with tumor antigens. Breast-cancer-specific CTLs against autologous breast cancer cells were successfully induced by αDC1s loaded with apoptotic MCF-7 cells. Conclusion Autologous DCs loaded with an allogeneic breast cancer cell line can generate potent breast-cancer-specific CTL responses. This may be a practical method for cellular immunotherapy in patients with breast cancer.
Collapse
Affiliation(s)
- Min-Ho Park
- Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University School of Medicine, Hwasun, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Huang FP, Chen YX, To CKW. Guiding the “misguided” - functional conditioning of dendritic cells for the DC-based immunotherapy against tumours. Eur J Immunol 2010; 41:18-25. [DOI: 10.1002/eji.201040543] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 10/06/2010] [Accepted: 11/15/2010] [Indexed: 12/15/2022]
|
16
|
Marchi LHL, Paschoalin T, Travassos LR, Rodrigues EG. Gene therapy with interleukin-10 receptor and interleukin-12 induces a protective interferon-γ-dependent response against B16F10-Nex2 melanoma. Cancer Gene Ther 2010; 18:110-22. [DOI: 10.1038/cgt.2010.58] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Yu X, Xia W, Zhang T, Wang H, Xie Y, Yang J, Miao J. Enhanced cytotoxicity of IL-24 gene-modified dendritic cells co-cultured with cytokine-induced killer cells to hepatocellular carcinoma cells. Int J Hematol 2010; 92:276-82. [PMID: 20697855 DOI: 10.1007/s12185-010-0654-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 07/21/2010] [Indexed: 12/29/2022]
Abstract
As a novel human cytokine found recently, IL-24 could selectively kill tumor cells by multiple ways. Dendritic cells (DCs) are the major antigen-presenting cells. Recent studies have revealed that IL-24 can promote the antigen-presenting function of DCs. In this study, we evaluated the antitumor effect and mechanism of co-cultured cytokine-induced killer (CIK) cells and autologous DCs modified with IL-24 gene on hepatocellular carcinoma (HCC) cells. DCs and CIK cells were prepared routinely from human peripheral blood mononuclear cells. Recombinant adenovirus AdVGFP/IL-24 (Ad-IL24) was constructed expressing IL-24. IL-24 gene was transduced into DCs via Ad-IL24, the cells obtained were named DC-IL-24. We demonstrated that the expression rates of CD80, CD83, CD1a, HLA-DR, CD40, CXCR4 on DC-IL-24 were significantly increased compared with those of the control group. DC-IL-24 produced markedly higher levels of IL-24, IL-12 and TNF-alpha as compared with those of DCs. On comparison with non-transfected DCs co-cultured with CIK cells, transfected DCs co-cultured with CIK cells had a significant higher lytic activity against SMMC7721 cells, a HCC cell line.
Collapse
Affiliation(s)
- Xin Yu
- Blood Transfusion Research Institute, Wuxi Red Cross Blood Center, Wuxi, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Enhanced cytotoxicity of IL-24 gene-modified dendritic cells co-cultured with cytokine-induced killer cells to hepatocellular carcinoma cells. Int J Hematol 2010. [PMID: 20697855 DOI: 10.1007/s12185-010-0654-1.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
As a novel human cytokine found recently, IL-24 could selectively kill tumor cells by multiple ways. Dendritic cells (DCs) are the major antigen-presenting cells. Recent studies have revealed that IL-24 can promote the antigen-presenting function of DCs. In this study, we evaluated the antitumor effect and mechanism of co-cultured cytokine-induced killer (CIK) cells and autologous DCs modified with IL-24 gene on hepatocellular carcinoma (HCC) cells. DCs and CIK cells were prepared routinely from human peripheral blood mononuclear cells. Recombinant adenovirus AdVGFP/IL-24 (Ad-IL24) was constructed expressing IL-24. IL-24 gene was transduced into DCs via Ad-IL24, the cells obtained were named DC-IL-24. We demonstrated that the expression rates of CD80, CD83, CD1a, HLA-DR, CD40, CXCR4 on DC-IL-24 were significantly increased compared with those of the control group. DC-IL-24 produced markedly higher levels of IL-24, IL-12 and TNF-alpha as compared with those of DCs. On comparison with non-transfected DCs co-cultured with CIK cells, transfected DCs co-cultured with CIK cells had a significant higher lytic activity against SMMC7721 cells, a HCC cell line.
Collapse
|
19
|
Um SJ, Choi YJ, Shin HJ, Son CH, Park YS, Roh MS, Kim YS, Kim YD, Lee SK, Jung MH, Lee MK, Son C, Choi PJ, Chung J, Kang CD, Lee EY. Phase I study of autologous dendritic cell tumor vaccine in patients with non-small cell lung cancer. Lung Cancer 2010; 70:188-94. [PMID: 20223553 DOI: 10.1016/j.lungcan.2010.02.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 02/08/2010] [Accepted: 02/09/2010] [Indexed: 11/27/2022]
Abstract
BACKGROUND A dendritic cell vaccine has been developed as a novel strategy for generating antitumor immunity in the treatment of cancer. The purpose of this study was to assess the maximal tolerated dose, safety, and immunologic response of a new dendritic cell vaccine (DC-Vac) into which tumor lysate was loaded by electroporation and pulse in patients with advanced non-small cell lung cancer (NSCLC). PATIENTS AND METHODS Fifteen patients with inoperable stage III or IV NSCLC were assigned to cohorts that received 3, 6, or 12 × 10(6) DC-Vac intradermally 3 times at 2 week intervals. We also evaluated immunologic and tumor responses. RESULTS The maximum dose of DC-Vac (12 × 10(6)) was shown to be safe. In 5 of 9 patients, the vaccine resulted in increased interferon (IFN)-γ production by CD8+ cells after exposure to tumor lysate. Additionally, there were mixed responses which do fulfill progressive disease definition but demonstrate some clinical benefit in two patients. CONCLUSION The administration of tumor lysate-loaded autologous dendritic cells by electroporation and pulse was non-toxic and induced immunologic responses to tumor antigens. The two mixed tumor responses which were achieved may represent a potential benefit of this new DC-Vac.
Collapse
Affiliation(s)
- Soo-Jung Um
- Department of Internal Medicine, Dong-A University College of Medicine, 3-Ga Dongdaeshin-dong, Seo-gu, Busan 602-715, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Jin CJ, Hong CY, Takei M, Chung SY, Park JS, Pham TNN, Choi SJN, Nam JH, Chung IJ, Kim HJ, Lee JJ. All-trans retinoic acid inhibits the differentiation, maturation, and function of human monocyte-derived dendritic cells. Leuk Res 2009; 34:513-20. [PMID: 19883939 DOI: 10.1016/j.leukres.2009.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Revised: 09/10/2009] [Accepted: 10/04/2009] [Indexed: 11/16/2022]
Abstract
All-trans retinoic acid (ATRA) affects on the function of antigen presenting cells with somewhat controversies. We investigated the effects of ATRA on differentiation, maturation and function of human monocyte-derived dendritic cells (DCs). Low dose (10(-14)M) or high dose (10(-6)M) of ATRA was added either when monocytes were differentiated into immature DCs (imDCs) or mature DCs (mDCs) were induced. Apoptotic cell populations were dramatically increased in imDCs or mDCs with increasing concentration of ATRA. The productions of IL-12p40 and IL-12p70 were significantly suppressed in imDCs or mDCs induced by the addition of ATRA in the dose-dependent manner, whereas IL-10 was increased. DCs cultured with ATRA induced the differentiation of naïve T cells towards a helper T cell type 2 (Th2) response and expansion of CD4(+)CD25(+)Foxp3(+) regulatory T cells. Allostimulatory capacity of DCs was suppressed with increasing concentration of ATRA. These findings suggest that ATRA inhibits the effects on the differentiation, maturation and function of human monocyte-derived DCs in vitro and also enhance the differentiation of naïve T cell toward the Th2 type.
Collapse
Affiliation(s)
- Chun-Ji Jin
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Jeonnam, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kaminski ER, Goddard RV, Prentice AG. Dendritic Cells and their Potential Therapeutic Role in Haematological Malignancy. Leuk Lymphoma 2009; 44:1657-66. [PMID: 14692516 DOI: 10.1080/1042819031000090219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The generation of an effective immune response is dependent on the efficient capture and presentation of antigen by antigen-presenting cells. The most potent antigen-presenting cells are dendritic cells (DC). These cells have the capability of activating naive helper and cytotoxic T cells. In recent years it has been demonstrated that in vivo responses to a number of solid tumours can be generated by DC pulsed with either purified tumour antigen or whole tumour cell lysate. In addition, a number of in vivo studies using DC have also been attempted in solid tumours, with some encouraging results. In haematological malignancies, there is now strong evidence that previous T cell anergy can be reversed and significant anti-tumour immune responses generated, in vitro, against the majority of leukaemias. As far as in vivo studies in haematological malignancies are concerned, although T cell responses have been demonstrated in the majority of cases and some dramatic early clinical responses reported, overall results appear disappointing. However, considering the fact that many of these studies were performed in patients with advanced disease and that such therapeutic strategies are still in their infancy, the overall results are actually quite encouraging. Although there is a real potential for DC immunotherapy in the future, it is important to be realistic about the limitations and obstacles to its development. It is highly unlikely that any form of immunotherapy is going to be effective in advanced disease due to the physical bulk of tumour, the immunosuppressive effects of tumours themselves and to any secondary immunosuppression following standard cancer therapy. The potential for immunotherapy is likely to lie either in adjunctive therapy or for treating minimal residual disease. Even in those situations, one of the major obstacles to be overcome is the state of immunological anergy or tolerance that many tumours seem able to induce. Indeed, there is evidence that, under certain circumstances, DC themselves can present antigen in such a way as to produce this state of anergy. Although, in vitro manipulation of DC and T cells can generate tumour-specific T cells from previously "anergic" cells, once reintroduced in vivo, these cells will be re-exposed to the tumour environment with the risk of being rendered anergic again.
Collapse
Affiliation(s)
- E R Kaminski
- Plymouth Post-graduate Medical School, Derriford Combined Laboratories, Derriford Hospital, Plymouth PL6 8DH, UK.
| | | | | |
Collapse
|
22
|
Ritchie D, Hermans I, Yang J, Walton J, Matthews K, Carter J, Findlay M, Dady P, Rawson P, Ronchese F. Autologous dendritic cells pulsed with eluted peptide as immunotherapy for advanced B-cell malignancies. Leuk Lymphoma 2009; 47:675-82. [PMID: 16690526 DOI: 10.1080/10428190500376365] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We have studied the feasibility, safety and efficacy of vaccination with autologous dendritic cells pulsed with eluted peptide in patients with advanced low-grade B-cell malignancies. This study demonstrates that autologous dendritic cell vaccines can be successfully produced from patients with advanced disease and be delivered without significant toxicity. Furthermore, we have demonstrated immunological and clinical responses in two of ten patients treated. These results provide further evidence for the use of immunotherapy in the management of B-cell malignancies, but also suggest that sustained responses may only be possible in patients with low bulk disease early in the disease course.
Collapse
Affiliation(s)
- David Ritchie
- Malaghan Institute of Medical Research, Wellington, New Zealand.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Induction of myeloma-specific cytotoxic T lymphocytes ex vivo by CD40-activated B cells loaded with myeloma tumor antigens. Ann Hematol 2009; 88:1113-23. [DOI: 10.1007/s00277-009-0721-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2008] [Accepted: 02/25/2009] [Indexed: 11/25/2022]
|
24
|
Chen YX, Man K, Ling GS, Chen Y, Sun BS, Cheng Q, Wong OH, Lo CK, Ng IOL, Chan LC, Lau GK, Lin CLS, Huang F, Huang FP. A crucial role for dendritic cell (DC) IL-10 in inhibiting successful DC-based immunotherapy: superior antitumor immunity against hepatocellular carcinoma evoked by DC devoid of IL-10. THE JOURNAL OF IMMUNOLOGY 2007; 179:6009-15. [PMID: 17947674 DOI: 10.4049/jimmunol.179.9.6009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The dendritic cell (DC)-based tumor immunotherapy has been a new promise of cure for cancer patients, but animal studies and clinical trials have thus far only shown limited success, especially in treating established tumors. Certain immunosuppressive mechanisms triggered by tumor cells or the derivatives are believed to be a major obstacle. We studied the role of DC-derived IL-10 and its negative impact on vaccine efficacy in mouse models. Liver tumor cells were injected via the portal vein, giving rise to disseminated intrahepatic tumors, or s.c. to form solid but extrahepatic tumors. Bone marrow-derived DCs were generated from normal or IL-10-deficient mice and used as the vector to deliver tumor Ags. We demonstrate here that DCs devoid of IL-10, a potent immunosuppressive cytokine, are superior over conventional DCs in triggering antitumor immunity. The IL-10(-/-)DCs were highly immunogenic, expressed enhanced levels of surface MHC class II molecules, and secreted increased amounts of Th1-related cytokines. By inducing tumor-specific killing and through the establishment of immunological memory, the vaccines delivered by IL-10(-/-)DCs could evoke strong therapeutic and protective immunity against hepatocellular carcinoma in the mouse models. These findings will have great clinical impact once being translated into the treatment of malignant, and potentially infectious, diseases in humans.
Collapse
Affiliation(s)
- Yu-Xiao Chen
- Department of Pathology, University of Honk Kong, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Nguyen Pham TN, Choi BH, Kang HK, Jin CC, Minh NHT, Kim SK, Nam JH, Yang DH, Kim YK, Kim HJ, Chung IJ, Lee JJ. Optimization and limitation of calcium ionophore to generate DCs from acute myeloid leukemic cells. Cancer Res Treat 2007; 39:175-80. [PMID: 19746185 DOI: 10.4143/crt.2007.39.4.175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Accepted: 11/12/2007] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Calcium ionophore (CI) is used to generate dendritic cells (DCs) from progenitor cells, monocytes, or leukemic cells. The aim of this study was to determine the optimal dose of CI and the appropriate length of cell culture required for acute myeloid leukemia (AML) cells and to evaluate the limitations associated with CI. MATERIALS AND METHODS To generate leukemic DCs, leukemic cells (4x10(6) cells) from six AML patients were cultured with various concentrations of CI and/or IL-4 for 1, 2 or 3 days. RESULTS Potent leukemic DCs were successfully generated from all AML patients, with an average number of 1.2x10(6) cells produced in the presence of CI (270 ng/ml) for 2 days. Several surface molecules were clearly upregulated in AML cells supplemented with CI and IL-4, but not CD11c. Leukemic DCs cultured with CI had a higher allogeneic T cell stimulatory capacity than untreated AML cells, but the addition of IL-4 did not augment the MLR activity of these cells. AML cells cultured with CI in the presence or absence of IL-4 showed increased levels of apoptosis in comparison to primary cultures of AML cells. CONCLUSION Although CI appears to be advantageous in terms of time and cost effectiveness, the results of the present study suggest that the marked induction of apoptosis by CI limits its application to the generation of DCs from AML cells.
Collapse
|
26
|
Svane IM, Pedersen AE, Johansen JS, Johnsen HE, Nielsen D, Kamby C, Ottesen S, Balslev E, Gaarsdal E, Nikolajsen K, Claesson MH. Vaccination with p53 peptide-pulsed dendritic cells is associated with disease stabilization in patients with p53 expressing advanced breast cancer; monitoring of serum YKL-40 and IL-6 as response biomarkers. Cancer Immunol Immunother 2007; 56:1485-99. [PMID: 17285289 PMCID: PMC11030002 DOI: 10.1007/s00262-007-0293-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Accepted: 01/13/2007] [Indexed: 10/23/2022]
Abstract
p53 Mutations are found in up to 30% of breast cancers and peptides derived from over-expressed p53 protein are presented by class I HLA molecules and may act as tumor-associated epitopes in cancer vaccines. A dendritic cell (DC) based p53 targeting vaccine was analyzed in HLA-A2+ patients with progressive advanced breast cancer. DCs were loaded with 3 wild-type and 3 P2 anchor modified HLA-A2 binding p53 peptides. Patients received up to 10 sc vaccinations with 5 x 10(6) p53-peptide loaded DC with 1-2 weeks interval. Concomitantly, 6 MIU/m(2) interleukine-2 was administered sc. Results from a phase II trial including 26 patients with verified progressive breast cancer are presented. Seven patients discontinued treatment after only 2-3 vaccination weeks due to rapid disease progression or death. Nineteen patients were available for first evaluation after 6 vaccinations; 8/19 evaluable patients attained stable disease (SD) or minor regression while 11/19 patients had progressive disease (PD), indicating an effect of p53-specific immune therapy. This was supported by: (1) a positive correlation between p53 expression of tumor and observed SD, (2) therapy induced p53 specific T cells in 4/7 patients with SD but only in 2/9 patients with PD, and (3) significant response associated changes in serum YKL-40 and IL-6 levels identifying these biomarkers as possible candidates for monitoring of response in connection with DC based cancer immunotherapy. In conclusion, a significant fraction of breast cancer patients obtained SD during p53-targeting DC therapy. Data encourage initiation of a randomized trial in p53 positive patients evaluating the impact on progression free survival.
Collapse
Affiliation(s)
- Inge Marie Svane
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kang HK, Park JS, Kim SK, Choi BH, Pham TNN, Zhu XW, Cho D, Nam JH, Kim YJ, Rhee JH, Chung IJ, Kim HJ, Lee JJ. Down-regulation of cellular vascular endothelial growth factor levels induces differentiation of leukemic cells to functional leukemic-dendritic cells in acute myeloid leukemia. Leuk Lymphoma 2007; 47:2224-33. [PMID: 17071499 DOI: 10.1080/10428190600732236] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We examined the effect of cellular vascular endothelial growth factor (VEGF) levels on the generation of leukemic dendritic cells (DCs). Leukemic DCs were successfully generated in vitro from bone marrow cells of 16 of 21 acute myeloid leukemia (AML) patients, and the cellular VEGF concentrations in the leukemic cells and the neutralization of VEGF with anti-VEGF antibody were determined. AML cells that failed to generate leukemic DCs showed significantly higher cellular VEGF levels compared with generated leukemic DCs, and down-regulation of cellular VEGF levels induced the generation of leukemic DCs from AML cells. Inhibition of cellular VEGF levels increased interleukin (IL)-12 production and the allostimulatory capacity of leukemic DCs. These results suggest that the generation of leukemic DCs from AML cells is inversely related to the VEGF production of the cells and that the down-regulation of cellular VEGF levels can induce potential differentiation of leukemic cells to functional leukemic DCs in patients with AML.
Collapse
Affiliation(s)
- Hyun-Kyu Kang
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hayati AR, Zulkarnaen M. An immunohistochemical study of CD1a and CD83-positive infiltrating dendritic cell density in cervical neoplasia. Int J Gynecol Pathol 2007; 26:83-8. [PMID: 17197902 DOI: 10.1097/01.pgp.0000225850.90115.bc] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cervical carcinoma is the second leading cancer in women in Malaysia, after breast cancer. Human papillomavirus (HPV) has been implicated in the development of dysplasia or cervical intraepithelial neoplasia and progression to squamous cell carcinoma. Because of the confinement of the human papillomavirus infection within the epithelial layer, the presence of dentritic cells or Langerhans cells in epithelial layer of the ectocervix is paramount in producing immune response. The mature dentritic cells express CD83 and high CD40/80/86, whereas the immature cells express CD1a and low CD40/80/86. By identifying CD1a and CD83, theoretically, both immature and mature dentritic cell populations can be studied. In view of the facts, we investigated the infiltrating cell density of mature and immature dentritic cells in cervical neoplasia.
Collapse
Affiliation(s)
- Abdul Rahman Hayati
- Department of Pathology, Faculty of Medicine, Hospital Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | |
Collapse
|
29
|
Choi BH, Kang HK, Park JS, Kim SK, Pham TNN, Zhu XW, Cho D, Nam JH, Chung IJ, Kim YJ, Rhee JH, Kim HJ, Lee JJ. Optimization of the concentration of autologous serum for generation of leukemic dendritic cells from acute myeloid leukemic cells for clinical immunotherapy. J Clin Apher 2006; 21:233-40. [PMID: 17120232 DOI: 10.1002/jca.20105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Clinical application of immunotherapy for acute myeloid leukemia (AML) requires the efficient induction of dendritic cells (DCs) from AML blast cells using in vitro culture. We examined the effect of autologous serum on the properties of leukemic DCs derived from leukemic cells of AML patients by culture in AIM-V medium with GM-CSF, IL-4, TNF-alpha, and 0, 2, 5, or 10% human autologous serum. The expressions of CD80, CD83, CD86, and HLA-DR were upregulated under all culture conditions; however, 10% autologous serum induced the highest expression levels of several molecules. The capacity of leukemic DCs to stimulate allogeneic T cells increased with increasing serum concentration. Stimulation of autologous CD3(+) T cells with leukemic DCs grown in the presence of various concentrations of autologous serum resulted in induction of more IFN-gamma-secreting cells than was the case for unprimed CD3(+) T cells. Leukemic DCs cultured with 10% autologous serum induced the highest numbers of IFN-gamma-secreting cells and CD8(+)CD56(+) T cells from autologous T cells. These results suggest that culture of AML blast cells in the presence of autologous serum could be used to generate leukemic DCs for immunotherapy against AML. The highest serum concentration appeared optimal for generating the most potent leukemic DCs.
Collapse
Affiliation(s)
- Bo-Hwa Choi
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lee JJ, Park MS, Park JS, Kang HK, Kim SK, Nguyen Pham TN, Zhu XW, Cho D, Nam JH, Kim YJ, Rhee JH, Chung IJ, Kim HJ. Induction of leukemic-cell-specific cytotoxic T lymphocytes by autologous monocyte-derived dendritic cells presenting leukemic cell antigens. J Clin Apher 2006; 21:188-94. [PMID: 16570260 DOI: 10.1002/jca.20085] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Leukemic-dendritic cells (leukemic-DCs) have certain limitations, which include difficult generation in 30-40% of patients, and low levels of expression of several key molecules. Therefore, an alternative approach using monocyte-derived DCs pulsed with tumor antigens is required. We investigated the possibility of immunotherapy for AML using leukemic-cell-specific cytotoxic T lymphocytes that were stimulated in vitro by autologous DCs pulsed with tumor antigens. To generate DCs, CD14(+) cells were isolated from peripheral blood mononuclear cells using magnetic-activated cell sorting, and cultured in the presence of GM-CSF and IL-4. On day 6, maturation of DCs was induced by addition of cytokine cocktail (TNF-alpha, IL-1beta, IL-6, and prostaglandin E(2)) for 2 days, and then the mature DCs were pulsed with whole leukemic cell lysates or apoptotic leukemic cells. There were no differences in the phenotypic expressions of mature DCs generated by pulsing with or without leukemic antigens. The mature DCs pulsed with tumor cell lysates or apoptotic leukemic cells showed a higher allostimulatory capacity for allogeneic CD3(+) T cells as compared with mature non-pulsed DCs. Autologous CD3(+) T cells stimulated by the mature pulsed DCs showed more potent cytotoxic activities against autologous leukemic cells than those stimulated by mature non-pulsed DCs. These results suggest that use of DCs pulsed with leukemic cell lysates or apoptotic leukemic cells is a feasible alternative immunotherapeutic approach to overcome the limitations of leukemic-DCs for the treatment of AML patients.
Collapse
Affiliation(s)
- J-J Lee
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Iinuma H, Okinaga K, Fukushima R, Inaba T, Iwasaki K, Okinaga A, Takahashi I, Kaneko M. Superior protective and therapeutic effects of IL-12 and IL-18 gene-transduced dendritic neuroblastoma fusion cells on liver metastasis of murine neuroblastoma. THE JOURNAL OF IMMUNOLOGY 2006; 176:3461-9. [PMID: 16517714 DOI: 10.4049/jimmunol.176.6.3461] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fusion vaccine of dendritic cells (DCs) and tumor cells has the advantage of inducing an immune response against multiple tumor Ags, including unknown tumor Ags. Using the liver metastasis model of C1300 neuroblastoma cells, we assessed the protective and therapeutic effects of fusion cells transduced with the IL-12 gene and/or the IL-18 gene. Improving the fusion method by combining polyethylene glycol and electroporation increased loading efficiency. In the A/J mice vaccinated with fusion cells modified with the LacZ gene (fusion/LacZ), IFN-gamma production and CTL activity increased significantly compared with that of DCs/LacZ, C1300/LacZ, or a mixture of the two (mixture/LacZ). With the transduction of IL-12 and IL-18 genes into the fusion cells (fusion/IL-12/IL-18), the level of IFN-gamma increased more than five times that of other fusion groups. In addition, NK cell activity and CTL activity increased significantly compared with that of mixture/LacZ, fusion/LacZ, DC/LacZ, or C1300/LacZ. In the protective and therapeutic studies of fusion cell vaccine, mice vaccinated with fusion/LacZ, fusion/IL-12, fusion/IL-18, or fusion/IL-12/IL-18 showed a significant decrease in liver metastasis and a significant increase in survival compared with mice given a mixture/LacZ, DCs/LacZ, or C1300/LacZ. In particular, the mice receiving fusion/IL-12/IL-18 vaccine showed a complete protective effect and the highest therapeutic effects. The present study investigates the improved loading efficiency of fusion cells and suggests that the introduction of IL-12 and IL-18 genes can induce extremely strong protective and therapeutic effects on liver metastasis of neuroblastoma.
Collapse
Affiliation(s)
- Hisae Iinuma
- Department of Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-0003.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Robin M, Schlageter MH, Chomienne C, Padua RA. Targeted immunotherapy in acute myeloblastic leukemia: from animals to humans. Cancer Immunol Immunother 2005; 54:933-43. [PMID: 15889256 PMCID: PMC11034191 DOI: 10.1007/s00262-005-0678-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Accepted: 01/14/2005] [Indexed: 11/29/2022]
Abstract
Immunity against acute myeloid leukemia (AML) is demonstrated in humans by the graft-versus-leukemia effect in allogeneic hematopoietic stem cell transplantation. Specific leukemic antigens have progressively been discovered and circulating specific T lymphocytes against Wilms tumor antigen, proteinase peptide or fusion-proteins produced from aberrant oncogenic chromosomal translocations have been detected in leukemic patients. However, due to the fact that leukemic blasts develop various escape mechanisms, antileukemic specific immunity is not able to control leukemic cell proliferation. The aim of immunotherapy is to overcome tolerance and boost immunity to elicit an efficient immune response against leukemia. We review different immunotherapy strategies tested in preclinical animal models of AML and the human trials that spurred from encouraging results obtained in animal models, demonstrate the feasibility of immunotherapy in AML patients.
Collapse
Affiliation(s)
- Marie Robin
- LBCH INSERM U718, Hôpital Saint Louis APHP, Institut Universitaire d'Hématologie, 1 Avenue Claude Vellefaux, 75010, Paris, France
| | | | | | | |
Collapse
|
33
|
Panoskaltsis N. Dendritic cells in MDS and AML--cause, effect or solution to the immune pathogenesis of disease? Leukemia 2005; 19:354-7. [PMID: 15674424 DOI: 10.1038/sj.leu.2403634] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- N Panoskaltsis
- Department of Haematology, Imperial College London, London, UK.
| |
Collapse
|
34
|
Do TH, Johnsen HE, Kjaersgaard E, Taaning E, Svane IM. Impaired circulating myeloid DCs from myeloma patients. Cytotherapy 2005; 6:196-203. [PMID: 15203976 DOI: 10.1080/14653240410006004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND In clinical trials, cancer patients have received immunotherapy based on DCs generated from leukapheresed blood. It would therefore be an advantage to be able to measure blood levels and estimate the phenotype of DC before leukapheresis, to estimate the yield required for preparation of vaccines, or ex vivo stimulation of T cells for adoptive immunotherapy. METHODS Recently, circulating lineage negative (Lin-) myeloid DC cells and their precursors have been identified by flow cytometry. We apply this strategy to the screening of blood samples from patients with multiple myeloma, in an attempt to characterize and quantitate the subset. By a direct flow cytometry approach, the blood levels of circulating lineage (CD3, CD19, CD14) negative, CD33++, HLA-DR+ cells were estimated before and following ex vivo cell differentiation, and phenotyped by MAbs with specificity against HLA-DR, HLA-ABC, CD1a, CD11c, CD33, CD40, CD49d, CD49e, CD54, CD80, CD83, and CD86. RESULTS This study demonstrated that multiple myeloma patients have a 50% reduced blood level of Lin-, CD33++, HLA-DR+ myeloid DC, but a DC-precursor level within normal range. Furthermore, GM-CSF and IL-4 ex vivo stimulated DCs demonstrated an impaired up-regulation of the co-stimulatory molecule CD80 and the adhesion molecule CD54. DISCUSSION These results may have clinical implications as a predictor for yield and functionality of the harvested DCs to be used in vaccination of myeloma patients.
Collapse
Affiliation(s)
- T H Do
- Department of Hematology, Herlev Hospital, University of Copenhagen Herlev, Denmark
| | | | | | | | | |
Collapse
|
35
|
Lee JJ, Kook H, Park MS, Nam JH, Choi BH, Song WH, Park KS, Lee IK, Chung IJ, Hwang TJ, Kim HJ. Immunotherapy using autologous monocyte-derived dendritic cells pulsed with leukemic cell lysates for acute myeloid leukemia relapse after autologous peripheral blood stem cell transplantation. J Clin Apher 2005; 19:66-70. [PMID: 15274198 DOI: 10.1002/jca.10080] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although a second stem cell transplantation (SCT) can be used as salvage therapy in patients with relapsing leukemia after SCT, most of these patients have a poor outcome. We tried clinical vaccination using monocyte-derived dendritic cells (DCs) pulsed with leukemic lysates to treat relapsing acute myeloid leukemia (AML) after autologous SCT. To generate DCs, CD14+ cells isolated from peripheral blood stem cell products were cultured in AIM-V in the presence of GM-CSF and IL-4. Adding TNF-alpha on day 6 induced maturation of the DCs, which were harvested on day 8 or 9. The DCs were incubated with tumor lysate and KLH for 2 hr at 37 degrees C. After certifying the absence of microorganisms and endotoxins, the patients received four DC vaccinations at two- to three-week intervals. Two patients received four DC vaccinations with means of 7.8 x 10(6) and 9 x 10(6) DCs at two- to three-week intervals. The DC vaccinations were well tolerated with no apparent side effects. After the vaccinations, the patients showed immunological responses with positive delayed-type hypersensitivity skin reaction and increasing autologous T cells stimulatory capacity to the DCs; however, the BM blast percentage of the patients did not improve. The results suggest that DCs are a feasible cellular therapy for relapsing AML after autologous SCT.
Collapse
Affiliation(s)
- Je-Jung Lee
- Blood & Marrow Transplant Program, Chonnam National University Medical School, Gwangju, Korea.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lee JJ, Choi BH, Nam JH, Park MS, Song WH, Yang DH, Kim YK, Cho SH, Chung IJ, Park KS, Lee IK, Kim HJ. The generation of leukemic dendritic cells from acute myeloid leukemia cells is potentiated by the addition of CD40L at the terminal maturation stage. J Clin Apher 2004; 19:130-6. [PMID: 15493054 DOI: 10.1002/jca.20015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Leukemic dendritic cells (DCs) that are derived from acute myeloid leukemia (AML) cells display low-level expression of several key molecules. We investigated the optimal combination of cytokines needed to generate potent leukemic DCs from AML cells in vitro. AML cells were cultured in the presence of the following combinations of cytokines: Group A, granulocyte-macrophage colony-stimulating factor (GM-CSF) + interleukin-4 (IL-4) + tumor necrosis factor-alpha (TNF-alpha); Group B, GM-CSF + IL-4 + CD40L; and Group C, CD40L addition at the terminal maturation point of cells that were grown as for Group A. The AML cells showed clear upregulation of CD80, CD83, CD86, CD40, and HLA-DR expression under all culture conditions, without significant differences between these groups. However, the addition of CD40L (as in Group C) showed a slight upregulation in the expression of CD83 and CD86 on leukemic DCs. The leukemic DCs in Groups A and B had higher allogeneic T-cell stimulatory capacities than untreated AML cells, and the addition of CD40L (Group C) enhanced this effect. The function of the cytotoxicity-stimulating autologous T cells was also augmented by the addition of CD40L (Group C). These results suggest that AML cells may be used to generate leukemic DCs using various cytokine combinations, and that the most potent, mature leukemic DCs are generated by the addition of CD40L to terminal-stage AML cultures that are grown in the presence of conventional cytokine combinations.
Collapse
Affiliation(s)
- Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Medical School, Gwangju, Korea.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lee JJ, Nam CE, Nam JH, Lee HC, Chung IJ, Park MS, Choi BH, Song WH, Lee IK, Park KS, Kook H, Hwang TJ, Takei M, Takaue Y, Kim HJ. Generation of cytotoxic donor CD8+ T cells against relapsing leukemic cells following allogeneic transplantation by stimulation with leukemic cell- or leukemic lysate pulsed donor cell-derived dendritic cells. Leuk Res 2004; 28:517-24. [PMID: 15068905 DOI: 10.1016/j.leukres.2003.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2002] [Accepted: 08/13/2003] [Indexed: 11/24/2022]
Abstract
To treat leukemia relapse after allogeneic hematopoietic stem cell transplantation (HSCT), we investigated the possibility of immunotherapy using donor CD8+ T cells that were generated by stimulating leukemic cell-derived dendritic cells (leukemic-DCs) or leukemic cell lysate pulsed donor cell-derived DCs (donor-DCs). Leukemic- and donor-DCs were generated from mononuclear cells of patients and CD14+ cells of HLA-matched donors, respectively. The expression of CD80, CD83, CD86, CD1a, and CD40 on leukemic-DCs was significantly lower than that on donor-DCs. Donor-DCs exhibited a higher capacity to stimulate allogeneic T cells compared with leukemic-DCs. Donor CD8+ T cells stimulated by leukemic- or donor-DCs were more cytotoxic than unprimed CD8+ T cells, and slightly higher cytotoxicity was observed with donor-DCs compared to leukemic-DCs. This study indicates that leukemic- or donor-DCs pulsed with leukemic cell lysates can effectively prime donor cytotoxic T cells in vitro, and that they may be used as a potential alternative tool for treating leukemic patients who relapse after allogeneic HSCT.
Collapse
Affiliation(s)
- Je-Jung Lee
- Genome Research Center for Hematopoietic Diseases, Chonnam National University Hospital, Gwangju, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Palena C, Zhu M, Schlom J, Tsang KY. Human B cells that hyperexpress a triad of costimulatory molecules via avipox-vector infection: an alternative source of efficient antigen-presenting cells. Blood 2004; 104:192-9. [PMID: 15010371 DOI: 10.1182/blood-2003-09-3211] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Dendritic cells (DCs) are the most potent of the antigen-presenting cells (APCs). Preparation of sufficient numbers of mature DCs, however, is both costly and time-consuming. We have examined here the possibility of using an alternative source of APCs that would be easier to obtain, would not require extensive culture, and thus would be more applicable to human immunotherapy protocols. We show here that freshly isolated human B cells can be efficiently infected by a replication-defective fowlpox recombinant vector, designated rF-TRICOM (TRIad of COstimulatory Molecules), to markedly increase surface expression of the human costimulatory molecule B7-1 and moderately increase expression of intercellular adhesion molecule-1 (ICAM-1) and leukocyte function-associated antigen-3 (LFA-3). Peptide-pulsed rF-TRICOM-infected B cells were highly efficient in activating antigen-specific human T cells and shown to be superior to the use of CD40L in enhancing APC potency. Moreover, when infection of freshly isolated B cells with rF-TRICOM was combined with CD40L, a still further marked enhancement of the antigen-presenting potency was observed. Ex vivo-generated antigen-specific T cells activated in this manner might be applied to experimental protocols or used for adoptive transfer in immunotherapy protocols.
Collapse
Affiliation(s)
- Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
39
|
Tam YK, Martinson JA, Doligosa K, Klingemann HG. Ex vivo expansion of the highly cytotoxic human natural killer-92 cell-line under current good manufacturing practice conditions for clinical adoptive cellular immunotherapy. Cytotherapy 2004; 5:259-72. [PMID: 12850795 DOI: 10.1080/14653240310001523] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Adoptive transfer of ex vivo expanded cytotoxic immune cells has become a viable strategy for treatment of malignant disease. Natural killer (NK)-92, a highly cytotoxic, IL2-dependent human NK cell-line, is an excellent candidate as an immunotherapeutic agent, being active for prolonged periods following irradiation and IL2 deprivation, non-toxic and non-immunogenic, and easily expanded. A number of clinical trials using NK-92 for different indications are currently underway. The aim of this study was to develop current good manufacturing practice (cGMP)-compliant expansion methodology for NK-92. METHODS The ability to expand NK-92 ex vivo was evaluated. Serum-free culture media, as well as media supplements (IL2, serum/plasma/albumin), culture containers and feeding regimens were compared for their ability to support expansion, viability and cytotoxicity of NK-92 cells. RESULTS NK-92 cells can be expanded in X-Vivo 10 serum-free media with 500 U/mL of rhIL2 (Proleukin), and 2.5% human serum/plasma to achieve concentrations sufficient to treat patients with >5210(10) cells. The protocol involves cultures initiated at 2.5210(5) cells/mL in 25 mL in 1 L Vuelife culture bags, with addition of fresh media plus IL2 every 3 days to maintain an optimal density of NK-92 cells for expansion. Daily disruption of cell aggregates enhances NK-92 cells expansion and viability during the culture period. Final yields of approximately 1.1-1.3210(6) cells/mL in a 1.2 L volume (1.36-1.56210(9) cells; 218-250 fold expansion) over 15-17 days is achievable under cGMP-compliant conditions with >85% viability. The feasibility of this approach has been shown in ongoing clinical trial with NK-92. DISCUSSION We describe a protocol that allows for >200-fold expansion of NK-92 cells within a 2-2.5 week period under GMP standards, in quality and quantity suitable for clinical adoptive immunotherapy.
Collapse
Affiliation(s)
- Y K Tam
- Section of Bone Marrow Transplant and Cell Therapy, Rush Presbyterian-St Luke's Medical Center, Rush Medical School, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
40
|
Irie M, Homma S, Komita H, Zeniya M, Kufe D, Ohno T, Toda G. Inhibition of spontaneous development of liver tumors by inoculation with dendritic cells loaded with hepatocellular carcinoma cells in C3H/HeNCRJ mice. Int J Cancer 2004; 111:238-45. [PMID: 15197777 DOI: 10.1002/ijc.20247] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We attempted to prevent spontaneous development of liver tumors by s.c. inoculation with DCs loaded with syngeneic HCC cells in C3H/HeNCrj mice. A new cell line, MIH-2, was established from an HCC that had developed spontaneously in a C3H/HeNCrj mouse. Bone marrow-derived DCs were loaded with irradiated MIH-2 cells by treatment with PEG. Fluorescence microscopy and flow-cytometric analysis showed that about 45% of PEG-treated DCs and MIH-2 cells (DC/MIH-2) were DCs loaded with MIH-2 cells. Thirteen-month-old mice received inoculations of DC/MIH-2 (9 x 10(5)/mouse) 4 times at 6-day intervals and were killed at 16 months of age to assess liver tumors. The incidence of liver tumors in these mice was significantly lower than that in mice not receiving inoculations (p < 0.05) but similar to that in 13-month-old mice (the age at which inoculation started), indicating that inoculation inhibited the development of new tumors. Splenocytes from inoculated mice, but not those from uninoculated mice, showed cytotoxic activity against MIH-2 cells. Cytotoxic activity was not elicited by CD4(+) T cells, CD8(+) T cells, or DX5(+) cells isolated from splenocytes but was elicited by adherent cells, identified as CD11b(+) macrophages. CD4(+) T cells, but not CD8(+) T cells, from inoculated mice produced IFN-gamma by incubation with DC/MIH-2. Cytotoxicity by splenocytes was attenuated by anti-IFN-gamma antibody. Immunization with DCs loaded with syngeneic HCC cells induces CD4(+) T cells that produce IFN-gamma by response to antigen of HCC, which would lead to macrophage activation to kill liver tumor cells at an early stage.
Collapse
Affiliation(s)
- Masaki Irie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Mu LJ, Gaudernack G, Saebøe-Larssen S, Hammerstad H, Tierens A, Kvalheim G. A protocol for generation of clinical grade mRNA-transfected monocyte-derived dendritic cells for cancer vaccines. Scand J Immunol 2003; 58:578-86. [PMID: 14629630 DOI: 10.1046/j.1365-3083.2003.01333.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
With the aim of producing large quantities of mRNA-transfected monocyte-derived dendritic cells (DCs) to be used as cancer vaccines, a new clinical grade procedure has been developed. Peripheral blood mononuclear cells (PBMCs) obtained by leukapheresis were enriched for monocytes by immunomagnetic depletion of CD19+ B cells and CD2+ T cells employing the ISOLEX 300i device. After 5 days of culture of enriched monocytes in gas permeable Teflon bags, using serum-free medium supplemented with granulocyte/macrophage-colony stimulating factor and interleukin-4 (IL-4), immature DCs were generated. Following transfection with mRNA from three human prostate cancer cell lines (DU145, LNCaP and PC-3), employing a newly developed square wave electroporation procedure, the immature DCs were immediately transferred to Teflon bags and matured for 48 h, using serum-free medium supplemented with IL-1alpha, IL-6, tumour necrosis factor-alpha and PGE2. The electroporation procedure efficiently transferred mRNA into the DCs with minor effect on the viability of the cells. The generated matured transfected DCs show high expression of the antigens CD83, CD80, CD86 and human leucocyte antigen-DR. Freezing and thawing of the transfected matured DCs had minor effect on cell viability and the phenotype. From 4 x 109 PBMCs, about 1 x 108 transfected matured DCs are produced. The thawed transfected DCs were able to elicit primary T-cell responses in vitro against antigens encoded by the prostate cancer mRNA as shown by enzyme-linked immunospot assay using mock-transfected DCs as control. Based on these results, clinical trials in cancer patients have been initiated.
Collapse
Affiliation(s)
- L J Mu
- Laboratory for Cellular Therapy, The Norwegian Radium Hospital, University of Oslo, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
42
|
Panoskaltsis N, Reid CDL, Knight SC. Quantification and cytokine production of circulating lymphoid and myeloid cells in acute myelogenous leukaemia. Leukemia 2003; 17:716-30. [PMID: 12682629 DOI: 10.1038/sj.leu.2402835] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A simple assay was developed to assess the potential of patients with acute myelogenous leukaemia (AML) to respond to immunotherapy. Lymphocytes, monocytes and leukaemic blasts with their corresponding intracellular cytokine profiles were evaluated by four-colour flow cytometry. In 50 microl samples of whole blood, surface labelling for CD45, CD8 and CD3 was used for cell identification prior to intracellular staining for interleukin (IL)-4, IL-10, IL-12 and interferon (IFN)-gamma. Absolute numbers of CD8(+) and CD8(-) (putative CD4(+)) T-cells, NK cells (CD8(+)/CD3(-)) and monocytes were determined by reference to a fixed number of added fluorescent beads. The absolute numbers of CD8(-) and CD8(+) T-cells in the blood of patients with AML were similar to those of normal controls. More of the lymphocytes in the blood of leukaemic patients spontaneously produced cytokines compared with those of controls. Furthermore, primary AML blasts secreted predominantly IFN-gamma. After recovery from chemotherapy, lymphocyte counts tended to be lower than in normals and reduction of NK cells reached significance after the second chemotherapy (P=0.01). A prominent CD8(lo)/CD3(lo-int) lymphocyte subset appeared after recovery in some patients. This laboratory application of the study of cell subsets and intracellular cytokines in patients undergoing treatment may be helpful in monitoring immunological responses in AML.
Collapse
Affiliation(s)
- N Panoskaltsis
- Antigen Presentation Research Geroup, Faculty of Medicine, Imperial College London and Department of Haematology, Northwick Park Hospital, Harrow, UK
| | | | | |
Collapse
|
43
|
Vuillier F, Maloum K, Thomas EK, Magnac C, Dumas G, Payelle-Brogard B, Oppezzo P, Dighiero G, Scott-Algara D. Idiotype-pulsed dendritic cells are able to induce antitumoral cytotoxic CD8 cells in chronic lymphocytic leukaemia. Br J Haematol 2003; 120:243-50. [PMID: 12542481 DOI: 10.1046/j.1365-2141.2003.04075.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Idiotypic structures of immunoglobulins from malignant B cells constitute tumour-specific antigens, though the function of immunoglobulin-specific CD8+ T cells in disease control and rejection remains unclear. We have studied five cases of B chronic lymphocytic leukaemia patients affected with indolent (three patients) or aggressive (two patients) disease. We showed that CD8+ T cells with major histocompatibility complex class I-restricted cytotoxicity against autologous tumour B cells could be generated following repeated stimulations with idiotype-pulsed dendritic cells in vitro. CD8+ T-cell lines were able to upregulate CD69 expression and to release interferon (IFN)-gamma upon contact with the autologous B cells, though cytolytic activity was only substantiated for patients with indolent disease. The failure of cytolytic activity in patients with aggressive disease may be explained by a skewed maturation of memory CD8 cells.
Collapse
MESH Headings
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- B-Lymphocytes/immunology
- CD8 Antigens/immunology
- Dendritic Cells/immunology
- Histocompatibility Antigens Class I
- Humans
- Immunoglobulin M/administration & dosage
- Immunoglobulin M/immunology
- Immunologic Memory
- Immunotherapy, Adoptive/methods
- Interferon-gamma/immunology
- Lectins, C-Type
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Lymphocyte Activation
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Françoise Vuillier
- Unité d'Immuno-Hématologie et d'Immunopathologie, Institut Pasteur, Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Goddard RV, Prentice AG, Copplestone JA, Kaminski ER. In vitro dendritic cell-induced T cell responses to B cell chronic lymphocytic leukaemia enhanced by IL-15 and dendritic cell-B-CLL electrofusion hybrids. Clin Exp Immunol 2003; 131:82-9. [PMID: 12519390 PMCID: PMC1808593 DOI: 10.1046/j.1365-2249.2003.02047.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HLA class II-restricted proliferative and cytotoxic T cell (CTL) responses to B cell chronic lymphocytic leukaemia (B-CLL) can be generated using autologous dendritic cells (DCs) pulsed with tumour cell lysate. In this study a number of different approaches were used to optimize further the in vitro system. First, the effects of a variety of maturation agents were studied. The addition of TNF-alpha, polyriboinosinic polyribocytidylic acid (Poly(I:C)) and LPS to autologous DCs resulted in the emergence of only a small percentage of CD83+ DCs, IFN-alpha having no demonstrable effect. Only the addition of Poly(I:C) to DCs resulted in modestly increased specific cytotoxicity to B-CLL targets, IFN-alpha and LPS having no effect. Secondly, T cells were pretreated with IL-15, prior to culturing with lysate-pulsed autologous DCs. A significant increase in T cell activation (P = 0.038), IFN-gamma secretion (P = 0.030) and specific cytotoxicity to B-CLL targets (P = 0.006) was demonstrated compared to untreated T cells. Thirdly, monocyte derived DCs electrofused with B-CLL B cells were compared with lysate-pulsed DCs. T cells stimulated by fused DCs generated higher levels of specific cytotoxicity to autologous B-CLL B cell targets than those stimulated by lysate pulsed DCs (P = 0.013). Blocking studies demonstrated inhibition of this cytotoxicity by both anti-CD4 (P = 0.062) and anti-CD8 monoclonal antibodies (P = 0.018), suggesting the generation of both HLA class I- and HLA class II-restricted CTL responses. In summary, in vitro B-CLL-specific T cell responses can be enhanced further by preincubating T cells with IL-15 and using autologous fused DC-B-CLL hybrids instead of autologous lysate-pulsed DCs. These preliminary data require confirmation with larger numbers of patients. Such an approach, however, may eventually provide effective immunotherapy for treatment of B-CLL.
Collapse
Affiliation(s)
- R V Goddard
- Plymouth Postgraduate Medical School, Derriford Combined Laboratory, Derriford Hospital, Plymouth, Devon, UK
| | | | | | | |
Collapse
|
45
|
Knight SC, Burke F, Bedford PA. Dendritic cells, antigen distribution and the initiation of primary immune responses to self and non-self antigens. Semin Cancer Biol 2002; 12:301-8. [PMID: 12147204 DOI: 10.1016/s1044-579x(02)00016-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunity or tolerance are determined through the bone marrow-derived, antigen-presenting cells, dendritic cells (DC). Stimulation of lymphocytes by different types of DC, DC at different stages of maturity and DC producing and responding to different growth factors modulate immune responses. Innate receptors for foreign or self antigens provide scope in DC for discrimination between different antigenic stimuli. DC also transfer processed antigens to other DC. We propose that DC do not stimulate responses to antigens in their own environment but only to antigens acquired from other DC, providing a mechanism for discriminating between environmental and non-environmental antigens.
Collapse
Affiliation(s)
- Stella C Knight
- Antigen Presentation Research Group, Northwick Park Institute for Medical Research, Imperial College Faculty of Medicine, Watford Rd., Harrow HA1 3UJ, UK.
| | | | | |
Collapse
|
46
|
Hinterberger-Fischer M, Hinterberger W. Blood stem cell transplantation for breast cancer: new approaches using pre- peri- post-transplant immunotherapy. Expert Opin Biol Ther 2001; 1:1029-48. [PMID: 11728234 DOI: 10.1517/14712598.1.6.1029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Autologous peripheral blood stem cell transplantation (auto-PBSCT) after high dose chemotherapy is usually offered to breast cancer patients carrying a high risk of relapse or having chemosensitive metastatic disease. Whether progression free and overall survival of such patients is improved after auto-PBSCT compared to conventional chemotherapy is a matter of debate. Currently available results of randomised trials could not uniformly prove or disprove auto-PBSCT being advantageous. Yet such studies have not employed any manipulation of the stem cell graft or any post-transplant immunomodulation exploiting the unique immunological environment for tumour eradication which exists only after auto-PBSCT. Preliminary data have discussed the ex vivo and in vivo generation of cytotoxic effector cells employing IL-2 and/or IFN-alpha/gamma in the auto-PBSCT setting. Other cytokines such as IL-12, IL-15 and prolactin have likewise been considered. Several anticancer vaccine protocols after auto-PBSCT are ongoing using monovalent vaccines or anti-idiotypic antibodies. Polyvalent anticancer vaccines, cytokine secreting tumour cells, tumour pulsed or hybridised dendritic cells (DC) enhanced with cytokines are studied. Monoclonal antibodies (mAb) could assist: unlabelled for pretransplant exvivo purging, post-transplant for enhancing antibody-dependent cell mediated cytotoxicity (ADCC) or radioimmunoconjugated as an additive cytotoxic part of the conditioning regimen. Autologous graft versus host induction and allogeneic stem cell transplantation (probably with non-myeloablative conditioning followed by donor lymphocyte infusions) are other approaches. Evaluation of successful combinations, optimal dosages and appropriate timing schedules is the subject of future investigations. Since breast cancer patients belong to countless subgroups, a large number of protocols need to be addressed in order to avoid over treatment and prevent relapse.
Collapse
Affiliation(s)
- M Hinterberger-Fischer
- Ludwig Boltzmann Institute for Stem Cell Transplantation, 2nd Dept. of Int. Medicine, Donauspital, Langobardenstrasse 122, A-1220 Vienna, Austria.
| | | |
Collapse
|
47
|
Bruserud Ø, Wendelboe Ø. Biological treatment in acute myelogenous leukaemia: how should T-cell targeting immunotherapy be combined with intensive chemotherapy? Expert Opin Biol Ther 2001; 1:1005-16. [PMID: 11728232 DOI: 10.1517/14712598.1.6.1005] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
T-cell targeting immunotherapy is now considered as a possible strategy in the treatment of acute myelogenous leukaemia (AML). Clinical importance of antileukaemic T-cell reactivity after allogeneic stem cell transplantation (SCT) is well established and the early experience from IL-2 therapy suggests that even autologous T-cells can mediate antileukaemic reactivity. The clinical experience also indicates that immunotherapy should begin when the leukaemia cell burden is minimal, and the detection of an operative cellular immune system, even in patients with chemotherapy-induced cytopenia, further suggests that it is possible to begin T-cell targeting therapy early after chemotherapy while patients are still cytopenic. However, adult patients in particular have a T-cell defect after chemotherapy that may last for several months. For this reason immunotherapy should probably be continued or repeated until a maximal effect is achieved when the patients no longer have a T-cell defect. This treatment approach may also be considered in combination with autologous SCT. T-cell targeting regimens should include, if possible, several therapeutic components. Firstly, native AML blasts can function as accessory cells during T-cell activation and in vivo therapy with T-cell growth factors (e.g., IL-2, IL-15) may then enhance antileukaemic reactivity or non-specific cytotoxicity against the AML cells; and secondly, a further enhancement of AML-specific reactivity may be achieved by vaccination with AML-specific peptides, immunisation with AML-blasts expressing a dendritic cell phenotype, or exposure to normal antigen-presenting cells (APC) pulsed with or expressing AML-specific peptide sequences.
Collapse
Affiliation(s)
- Ø Bruserud
- Division of Hematology, Department of Medicine, Haukeland University Hospital and the University of Bergen, Norway.
| | | |
Collapse
|
48
|
Kim YJ, Broxmeyer HE. Therapeutic potential of 4-1BB (CD137) as a regulator for effector CD8(+) T cells. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2001; 10:441-9. [PMID: 11522228 DOI: 10.1089/15258160152509064] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A fundamental problem of antitumor immunity is tumor-induced immunosuppression. Tumor cells often down-regulate expression of co-stimulatory molecules, tumor antigens, and major histocompatibility complex (MHC) molecules on tumor cells, secrete immunosuppressive substance such as transforming growth factor-beta (TGF-beta) or interleukin-4 (IL-4), and induce apoptosis of effector T cells to escape surveillance. A major goal of antitumor or antivirus immunotherapy is to generate long-lived protective T cells that enable killing of target cells. In this review, we discuss the importance of 4-1BB for development or survival of functionally active effector CD8(+) T cells against tumors, virus infection, and allogeneic immune responses and for potential therapeutic application.
Collapse
Affiliation(s)
- Y J Kim
- Department of Microbiology and Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|