1
|
Zhu T, Li Y, Wang Y, Li D. The Application of Dendritic Cells Vaccines in Tumor Therapy and Their Combination with Biomimetic Nanoparticles. Vaccines (Basel) 2025; 13:337. [PMID: 40333202 PMCID: PMC12031636 DOI: 10.3390/vaccines13040337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 05/09/2025] Open
Abstract
Dendritic cells (DCs) act as a bridge between innate and adaptive immunity by presenting antigens to effector immune cells and have shown broad application potential in tumor immunotherapy. However, the clinical translation of DC vaccines encounters significant challenges, such as the immunosuppressive tumor microenvironment (TME) and the sub-optimal DC function and vaccine efficacy in vivo. In this review, our investigation has uncovered the latest developments in DC vaccines and their potential in cancer immunotherapy, with a special emphasis on the integration of nanotechnology. Several types of nanomaterials, including protein cage nanoparticles (NPs), biomimetic NPs, and targeted multifunctional NPs, have been developed to enhance the antigen presentation ability of DCs and their stimulatory effects on T cells. In addition, we have also summarized the synergistic anti-cancer effects of DC vaccines with immune checkpoint inhibitors, chemotherapy, and radiotherapy. In addition, recent advances in nanotechnology have made it possible to develop novel biomarkers that can enhance the antigen presentation capacity of DCs and stimulate T cells. These biomarkers not only improve the accuracy and precision of DC vaccine design but also provide new insights into understanding the mechanisms of the DC-mediated immune response. Despite challenges pertaining to technical complexities and individual adaptation in the design and production of DC vaccines, personalized immunotherapy based on DCs is expected to become an important part of cancer treatment with rapid developments in biotechnology and immunology. This review provides new perspectives and potential solutions for the optimal design and application of DC vaccines in cancer therapy.
Collapse
Affiliation(s)
- Tong Zhu
- Panjin Central Hospital, Panjin 124010, China;
| | - Yuexin Li
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin 150081, China;
| | - Yutao Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing 100000, China
| | - Danyang Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
2
|
Pan J, Zeng W, Jia J, Shi Y, Wang D, Dong J, Fang Z, He J, Yang X, Zhang R, He M, Huang M, Fu B, Zhong B, Liu H. A Novel Therapeutic Tumor Vaccine Targeting MUC1 in Combination with PD-L1 Elicits Specific Anti-Tumor Immunity in Mice. Vaccines (Basel) 2022; 10:vaccines10071092. [PMID: 35891256 PMCID: PMC9325010 DOI: 10.3390/vaccines10071092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Dendritic cells (DCs), as professional antigen-presenting cells (APCs), play a key role in the initiation and regulation of humoral and cellular immunity. DC vaccines loaded with different tumor-associated antigens (TAAs) have been widely used to study their therapeutic effects on cancer. A number of clinical trials have shown that DCs are safe as an antitumor vaccine and can activate certain anti-tumor immune responses; however, the overall clinical efficacy of DC vaccine is not satisfactory, so its efficacy needs to be enhanced. MUC1 is a TAA with great potential, and the immune checkpoint PD-L1 also has great potential for tumor treatment. Both of them are highly expressed on the surface of various tumors. In this study, we generated a novel therapeutic MUC1-Vax tumor vaccine based on the method of PD-L1-Vax vaccine we recently developed; this novel PD-L1-containing MUC1-Vax vaccine demonstrated an elevated persistent anti-PD-L1 antibody production and elicited a much stronger protective cytotoxic T lymphocyte (CTL) response in immunized mice. Furthermore, the MUC1-Vax vaccine exhibited a significant therapeutic anti-tumor effect, which significantly inhibited tumor growth by expressing a high MUC1+ and PD-L1+ level of LLC and Panc02 tumor cells, and prolonged the survival of cancer-bearing animals. Taken together, our study provides a new immunotherapy strategy for improving the cross-presentation ability of therapeutic vaccine, which may be applicable to pancreatic cancer, lung cancer and for targeting other types of solid tumors that highly express MUC1 and PD-L1.
Collapse
Affiliation(s)
- Jiayi Pan
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- Clinical Laboratory, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Science, Guangzhou 510080, China
| | - Wuyi Zeng
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Jiangtao Jia
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Yi Shi
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Danni Wang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Jun Dong
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Zixuan Fang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Jiashan He
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Xinyu Yang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Rong Zhang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Menghua He
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Maoping Huang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
| | - Bishi Fu
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou 510260, China
| | - Bei Zhong
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- Correspondence: (B.Z.); (H.L.); Tel./Fax: +86-020-8320-5013 (H.L.)
| | - Hui Liu
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou 510182, China; (J.P.); (W.Z.); (J.J.); (Y.S.); (D.W.); (J.D.); (Z.F.); (J.H.); (X.Y.); (R.Z.); (M.H.); (M.H.); (B.F.)
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou 510260, China
- Correspondence: (B.Z.); (H.L.); Tel./Fax: +86-020-8320-5013 (H.L.)
| |
Collapse
|
3
|
Nava S, Lisini D, Frigerio S, Bersano A. Dendritic Cells and Cancer Immunotherapy: The Adjuvant Effect. Int J Mol Sci 2021; 22:ijms222212339. [PMID: 34830221 PMCID: PMC8620771 DOI: 10.3390/ijms222212339] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) are immune specialized cells playing a critical role in promoting immune response against antigens, and may represent important targets for therapeutic interventions in cancer. DCs can be stimulated ex vivo with pro-inflammatory molecules and loaded with tumor-specific antigen(s). Protocols describing the specific details of DCs vaccination manufacturing vary widely, but regardless of the employed protocol, the DCs vaccination safety and its ability to induce antitumor responses is clearly established. Many years of studies have focused on the ability of DCs to provide overall survival benefits at least for a selection of cancer patients. Lessons learned from early trials lead to the hypothesis that, to improve the efficacy of DCs-based immunotherapy, this should be combined with other treatments. Thus, the vaccine’s ultimate role may lie in the combinatorial approaches of DCs-based immunotherapy with chemotherapy and radiotherapy, more than in monotherapy. In this review, we address some key questions regarding the integration of DCs vaccination with multimodality therapy approaches for cancer treatment paradigms.
Collapse
|
4
|
Chimeric antigen receptor therapy in hematological malignancies: antigenic targets and their clinical research progress. Ann Hematol 2020; 99:1681-1699. [PMID: 32388608 DOI: 10.1007/s00277-020-04020-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 04/02/2020] [Indexed: 12/20/2022]
Abstract
Chimeric antigen receptor (CAR)-based immunotherapy has achieved dramatic success in the treatment of B cell malignancies, based on the summary of current research data, and has shown good potential in early phase cancer clinical trials. Modified constructs are being optimized to recognize and destroy tumor cells more effectively. By targeting the proper B-lineage-specific antigens such as CD19 and CD20, adoptive immunotherapy has demonstrated promising clinical results and already plays a role in the treatment of several lymphoid malignancies, which highlights the importance of target selection for other CAR therapies. The high efficacy of CAR-T cells has resulted in the approval of anti-CD19-directed CAR-T cells for the treatment of B cell malignancies. In this review, we focus on the basic structure and current clinical application of CAR-T cells, detail the research progress of CAR-T for different antigenic targets in hematological malignancies, and further discuss the current barriers and proposed solutions, investigating the possible mechanisms of recurrence of CAR-T cell therapy. A summary of the paper is also given to overview as the prospects for this therapy.
Collapse
|
5
|
Wang L, Yao R, Zhang L, Fan C, Ma L, Liu J. Chimeric antigen receptor T cell therapy and other therapeutics for malignancies: Combination and opportunity. Int Immunopharmacol 2019; 70:498-503. [PMID: 30875561 DOI: 10.1016/j.intimp.2019.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy provides possibility for the treatment of malignancies since clinical trials have shown that CAR-T therapy has a significant anti-tumor effect. Although many efforts have been made to improve the efficacy and reduce the side effects of CAR-T therapy, there are still many problems to solve. With the rapid development of this field, combination immunotherapy has been proved to improve the efficacy of CAR-T therapy. Studies have shown that radiotherapy, chemotherapy, oncolytic virotherapy, BTK inhibitors and immune checkpoint blockade-based therapy may further enhance the efficacy of CAR-T therapy while CRISPR/Cas9 technology and IL-1 blockade may improve the safety. In this review, we summarized the advantages and the mechanisms of the combination immunotherapy based on CAR-T cell therapy.
Collapse
Affiliation(s)
- Luyao Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Ruixue Yao
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Lifa Zhang
- 401 Hospital of the People's Liberation Army, China
| | - Chuanbo Fan
- Department of Hematology, Qingdao Hiser Medical Center, China
| | - Leina Ma
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China.
| |
Collapse
|
6
|
Liu J, Zhou J, Wu M, Hu C, Yang J, Li D, Wu P, Chen Y, Chen P, Lin S, Cui Y, Fu S, Wu J. Low-Dose Total Body Irradiation Can Enhance Systemic Immune Related Response Induced by Hypo-Fractionated Radiation. Front Immunol 2019; 10:317. [PMID: 30873170 PMCID: PMC6401363 DOI: 10.3389/fimmu.2019.00317] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/06/2019] [Indexed: 12/20/2022] Open
Abstract
A systemic immune related response (SIME) of radiotherapy has been occasionally observed on metastatic tumors, but the clinical outcomes remain poor. Novel treatment approaches are therefore needed to improve SIME ratio. We used a combination of hypo-fractionated radiation therapy (H-RT) with low-dose total body irradiation (L-TBI) in a syngeneic mouse model of breast and colon carcinoma. The combination therapy of H-RT and L-TBI potentially enhanced SIME by infiltration of CD8+ T cell and altering the immunosuppressive microenvironment in non-irradiated subcutaneous tumor lesions. The frequency of IFN-γ, as a tumor-specific CD8+ T cells producing, significantly inhibited the secondary tumor growth of breast and colon. Our findings suggest that L-TBI could serve as a potential therapeutic agent for metastatic breast and colon cancer and, together with H-RT, their therapeutic potential is enhanced significantly.
Collapse
Affiliation(s)
- Jing Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Jie Zhou
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - ChuanFei Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Juan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Dong Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Peng Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Yue Chen
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ping Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - Sheng Lin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - YongXia Cui
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - ShaoZhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| | - JingBo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
| |
Collapse
|
7
|
van Gulijk M, Dammeijer F, Aerts JGJV, Vroman H. Combination Strategies to Optimize Efficacy of Dendritic Cell-Based Immunotherapy. Front Immunol 2018; 9:2759. [PMID: 30568653 PMCID: PMC6289976 DOI: 10.3389/fimmu.2018.02759] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/09/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells (APCs) that are essential for the activation of immune responses. In various malignancies, these immunostimulatory properties are exploited by DC-therapy, aiming at the induction of effective anti-tumor immunity by vaccination with ex vivo antigen-loaded DCs. Depending on the type of DC-therapy used, long-term clinical efficacy upon DC-therapy remains restricted to a proportion of patients, likely due to lack of immunogenicity of tumor cells, presence of a stromal compartment, and the suppressive tumor microenvironment (TME), thereby leading to the development of resistance. In order to circumvent tumor-induced suppressive mechanisms and unleash the full potential of DC-therapy, considerable efforts have been made to combine DC-therapy with chemotherapy, radiotherapy or with checkpoint inhibitors. These combination strategies could enhance tumor immunogenicity, stimulate endogenous DCs following immunogenic cell death, improve infiltration of cytotoxic T lymphocytes (CTLs) or specifically deplete immunosuppressive cells in the TME, such as regulatory T-cells and myeloid-derived suppressor cells. In this review, different strategies of combining DC-therapy with immunomodulatory treatments will be discussed. These strategies and insights will improve and guide DC-based combination immunotherapies with the aim of further improving patient prognosis and care.
Collapse
Affiliation(s)
- Mandy van Gulijk
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
8
|
[Radiation therapy and immunomodulation: Focus on experimental data]. Cancer Radiother 2015; 19:515-8. [PMID: 26293415 DOI: 10.1016/j.canrad.2015.05.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 05/20/2015] [Indexed: 12/13/2022]
Abstract
The immunosuppressive effects of radiation therapy have long been the only ones considered. It has been demonstrated that exposure to ionizing radiation induces the release of tumour antigens which activates both the innate immune system and the adaptive immune response of the host. The purpose of tumour immunotherapy is based on the principle that reversal of tolerance to immunogenic tumours would be able to activate an immune response against tumour cells. Preclinical data and clinical studies early phase suggest a potential therapeutic benefit of immunotherapy combined with radiation therapy. The objective of this article is to review how tumour cells interact with the immune system and how ionizing radiation modulate this interaction and finally the combination of perspectives of immunotherapy and ionizing radiation by focusing on existing clinical data.
Collapse
|
9
|
Zheng Y, Dou Y, Duan L, Cong C, Gao A, Lai Q, Sun Y. Using chemo-drugs or irradiation to break immune tolerance and facilitate immunotherapy in solid cancer. Cell Immunol 2015; 294:54-9. [PMID: 25687508 DOI: 10.1016/j.cellimm.2015.02.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
The immunity is dual host-protective and tumor-promoting in cancer development and progression. Many immune suppressive cells and cytokines in microenvironment can prevent cytotoxic T lymphocytes (CTL) and natural killer cells (NK) from killing tumor cells. Chemotherapy drugs and irradiation have been reported helpful in breaking immune tolerance and creating microenvironment for adoptive cell therapy. Low-dose cyclophosphamide or gemcitabine therapy can selectively deplete T regulatory cells (Treg). Paclitaxel can alter cytokine network at the tumor site, and 5-fluorouracil shows a pronounced effect on myeloid-derived suppressor cells (MDSC) depletion. Local tumor irradiation and total body irradiation (TBI) can also affect tumor microenvironment and facilitate immunotherapy. In this review, we summarize the particular effects of these agents and methods on immunomodulation, as well as their potential values in immunotherapy. The combination with immunotherapy represents a novel therapeutic strategy.
Collapse
Affiliation(s)
- Yawen Zheng
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yan Dou
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Lili Duan
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Changsheng Cong
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Aiqin Gao
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Qinghua Lai
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
10
|
Radiation-induced autophagy potentiates immunotherapy of cancer via up-regulation of mannose 6-phosphate receptor on tumor cells in mice. Cancer Immunol Immunother 2014; 63:1009-21. [PMID: 24943275 DOI: 10.1007/s00262-014-1573-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/10/2014] [Indexed: 12/13/2022]
Abstract
There is a significant body of evidence demonstrating that radiation therapy (XRT) enhances the effect of immune therapy. However, the precise mechanisms by which XRT potentiates the immunotherapy of cancer remain elusive. Here, we report that XRT potentiates the effect of immune therapy via induction of autophagy and resultant trafficking of mannose-6-phopsphate receptor (MPR) to the cell surface. Irradiation of different tumor cells caused substantial up-regulation of MPR on the cell surface in vitro and in vivo. Down-regulation of MPR in tumor cells with shRNA completely abrogated the combined effect of XRT and immunotherapy (CTLA4 antibody) in B16F10-bearing mice without changes in the tumor-specific responses of T cells. Radiation-induced MPR up-regulation was the result of redistribution of the receptor to the cell surface. This effect was caused by autophagy with redirection of MPR to autophagosomes in a clathrin-dependent manner. In autophagosomes, MPR lost its natural ligands, which resulted in subsequent trafficking of empty receptor(s) back to the surface. Together, our data demonstrated a novel mechanism by which XRT can enhance the effect of immunotherapy and the molecular mechanism of this process.
Collapse
|
11
|
Concurrent dendritic cell vaccine and strontium-89 radiation therapy in the management of multiple bone metastases. Ir J Med Sci 2014; 184:457-61. [PMID: 24876093 DOI: 10.1007/s11845-014-1145-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 05/12/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND In addition to its direct cytotoxic effects, radiation therapy renders tumor cells more susceptible to T cell-mediated cytotoxicity by modulating cell surface molecules involved in antigen presentation. The purpose of the present study was to determine the benefit of combined 89Sr radiation and dendritic cell (DC) vaccine therapy in bone metastasis patients. METHODS Patients were treated with intravenous 89Sr at a dose of 40 μCi/kg of body weight on the first day after the peripheral blood mononuclear cell collection. Seven days later, patients received DCs once a week for 6 weeks. The first three vaccines were administered by intravenous infusion, and the last three vaccines were administered by 24-point intradermal injection. Clinical response was evaluated by the number of bone metastatic foci demonstrated on bone scintigraphy; cell-mediated cytotoxicity response was evaluated by delayed-type hypersensitivity (DTH) reaction. All treatment-related toxicities including vaccine-induced fever and 89Sr-associated hematological toxicity were carefully monitored. RESULTS Twenty-six patients with histologically diagnosed with primary cancers and multiple bone metastases demonstrated on bone scintigraphy were studied. The overall survival rate was 58.3%. The total positive DTH rate was 50%. The efficiency rate for pain relief was 60% (6/10), for quantity of life was 80%, and for clinic responses was 90%. Out of 10 cases, the Grade 1 or 2 of hematological depression in 4, erythema in 1, and fever in 7 were observed. CONCLUSIONS The study has important implications for that combined 89Sr radiation, and DC vaccine therapy can benefit cancer patients with bone metastasis.
Collapse
|
12
|
Cell transfer therapy for cancer: past, present, and future. J Immunol Res 2014; 2014:525913. [PMID: 24741604 PMCID: PMC3987872 DOI: 10.1155/2014/525913] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/26/2013] [Indexed: 12/30/2022] Open
Abstract
Cell transfer therapy for cancer has made a rapid progress recently and the immunotherapy has been recognized as the fourth anticancer modality after operation, chemotherapy, and radiotherapy. Lymphocytes used for cell transfer therapy include dendritic cells, natural killer (NK) cells, and T lymphocytes such as tumor-infiltrating lymphocytes (TILs) and cytotoxic T lymphocytes (CTLs). In vitro activated or engineered immune cells can traffic to cancer tissues to elicit persistent antitumor immune response which is very important especially after immunosuppressive treatments such as chemotherapy. In this review, we overviewed recent advances in the exploration of dendritic cells, NK cells, and T cells for the treatment of human cancer cells.
Collapse
|
13
|
Hasumi K, Aoki Y, Wantanabe R, Mann DL. Clinical response of advanced cancer patients to cellular immunotherapy and intensity-modulated radiation therapy. Oncoimmunology 2013; 2:e26381. [PMID: 24349874 PMCID: PMC3858384 DOI: 10.4161/onci.26381] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 09/04/2013] [Accepted: 09/05/2013] [Indexed: 12/20/2022] Open
Abstract
Patients afflicted with advanced cancers were treated with the intratumoral injection of autologous immature dendritic cells (iDCs) followed by activated T-cell infusion and intensity-modulated radiation therapy (IMRT). A second round of iDCs and activated T cells was then administered to patients after the last radiation cycle. This complete regimen was repeated for new and recurring lesions after 6 weeks of follow-up. One year post therapy, outcome analyses were performed to evaluate treatment efficacy. Patients were grouped according to both the number and size of tumors and clinical parameters at treatment initiation, including recurrent disease after standard cancer therapy, Stage IV disease, and no prior therapy. Irrespective of prior treatment status, 23/37 patients with ≤ 5 neoplastic lesions that were ≤ 3 cm in diameter achieved complete responses (CRs), and 5/37 exhibited partial responses (PRs). Among 130 individuals harboring larger and more numerous lesions, CRs were observed in 7/74 patients that had received prior SCT and in 2/56 previously untreated patients. Some patients manifested immune responses including an increase in CD8+CD56+ lymphocytes among circulating mononuclear cells in the course of treatment. To prospectively explore the therapeutic use of these cells, CD8+ cells were isolated from patients that had been treated with cellular immunotherapy and IMRT, expanded in vitro, and injected into recurrent metastatic sites in 13 individuals who underwent the same immunoradiotherapeutic regimens but failed to respond. CRs were achieved in 34 of 58 of such recurrent lesions while PRs in 17 of 58. These data support the expanded use of immunoradiotherapy in advanced cancer patients exhibiting progressive disease.
Collapse
Affiliation(s)
- Kenichiro Hasumi
- Hasumi International Research Foundation; Tokyo Research Center; Tokyo, Japan
| | - Yukimasa Aoki
- Hasumi International Research Foundation; Tokyo Research Center; Tokyo, Japan
| | - Ryuko Wantanabe
- Hasumi International Research Foundation; Tokyo Research Center; Tokyo, Japan
| | - Dean L Mann
- Department of Pathology; University of Maryland School of Medicine; Baltimore, MD USA
| |
Collapse
|
14
|
Zheng YW, Li RM, Zhang XW, Ren XB. Current Adoptive Immunotherapy in Non-Small Cell Lung Cancer and Potential Influence of Therapy Outcome. Cancer Invest 2013; 31:197-205. [DOI: 10.3109/07357907.2013.775294] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
15
|
Wei S, Egenti MU, Teitz-Tennenbaum S, Zou W, Chang AE. Effects of tumor irradiation on host T-regulatory cells and systemic immunity in the context of adoptive T-cell therapy in mice. J Immunother 2013; 36:124-32. [PMID: 23377667 PMCID: PMC3607501 DOI: 10.1097/cji.0b013e31828298e6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In this study, we used a murine D5 melanoma model to study the effects of local tumor irradiation on the therapeutic efficacy of adoptive T-cell therapy. Tumor irradiation was delivered in 5 daily fractions (8.5 Gy) to subcutaneous tumors on days 7-11 after tumor inoculation. After the last radiation dose, activated tumor-draining lymph node cells were transferred intravenously followed by intraperitoneal IL-2 administration. Tumor irradiation alone had no significant effect on tumor growth; however, it synergistically enhanced the therapeutic efficacy of T-cell therapy. For 2 days after tumor irradiation there was a significant reduction in T cells, B cells, and CD11c(+) dendritic cells in both the tumor microenvironment and the systemic lymphoid compartments. By days 4-6 after irradiation, the relative reduction in the number of Treg cells within the tumor and the systemic compartments was greater than the reduction in conventional T cells. Furthermore, the suppressive function of the Tregs was significantly impaired in irradiated versus untreated mice. Using effector T cells derived from congenic mice, we found that local tumor irradiation resulted in increased proliferation of donor T cells within the tumor and the systemic lymphoid compartments. Radiation was associated with increased expression of the effector cytokines IFN-γ and TNF-α by donor and host CD4(+) and CD8(+) T cells. Altogether, our data indicate that local tumor irradiation has a distinct modulatory effect on Tregs and can enhance systemic antitumor immunity associated with adoptive T-cell therapy.
Collapse
Affiliation(s)
- Shuang Wei
- Department of Surgery, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109-5932, USA
| | | | | | | | | |
Collapse
|
16
|
Levy A, Chargari C, Cheminant M, Simon N, Bourgier C, Deutsch E. Radiation therapy and immunotherapy: implications for a combined cancer treatment. Crit Rev Oncol Hematol 2012; 85:278-87. [PMID: 23036459 DOI: 10.1016/j.critrevonc.2012.09.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/17/2012] [Accepted: 09/04/2012] [Indexed: 01/29/2023] Open
Abstract
Ionizing radiation (IR) is used as primary treatment for numerous localized cancers. Although it is usually described as an immunosuppressive modality, there are new preclinical evidences suggesting that IR could have also generated substantial changes in the tumor microenvironment, including triggering an inflammatory process. This finding implies that radiotherapy could both modulate tumor immunity and have out-of-field activity by recruiting biological effectors. There are numerous uncertainties regarding the true biological impact of radiation on tumor immunogenicity, but some preclinical studies established the proof of concept that combining IR with strategies modifying immunology could enhance antitumor effects. From these findings, clinical trials are now analyzing how immunotherapy and radiation work while given together, with promising preliminary results. This review aims at summarizing the recent developments regarding the impact of IR on tumor immunity, with focus on potential therapeutic targets.
Collapse
Affiliation(s)
- Antonin Levy
- Inserm U1030, Radiothérapie moléculaire, Université Paris XI, le Kremlin-Bicêtre, France
| | | | | | | | | | | |
Collapse
|
17
|
Guo C, Yi H, Yu X, Zuo D, Qian J, Yang G, Foster BA, Subjeck JR, Sun X, Mikkelsen RB, Fisher PB, Wang XY. In situ vaccination with CD204 gene-silenced dendritic cell, not unmodified dendritic cell, enhances radiation therapy of prostate cancer. Mol Cancer Ther 2012; 11:2331-41. [PMID: 22896667 DOI: 10.1158/1535-7163.mct-12-0164] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Given the complexity of prostate cancer progression and metastasis, multimodalities that target different aspects of tumor biology, for example, radiotherapy in conjunction with immunotherapy, may provide the best opportunities for promoting clinical benefits in patients with high-risk localized prostate cancer. Here, we show that intratumoral administration of unmodified dendritic cells (DC) failed to synergize with fractionated radiotherapy. However, ionizing radiation combined with in situ vaccination with DCs, in which the immunosuppressive scavenger receptor A (SRA/CD204) has been downregulated by lentivirus-mediated gene silencing, profoundly suppressed the growth of two mouse prostate cancers (e.g., RM1 and TRAMP-C2) and prolonged the lifespan of tumor-bearing animals. Treatment of subcutaneous tumors with this novel combinatorial radioimmunotherapeutic regimen resulted in a significant reduction in distant experimental metastases. SRA/CD204-silenced DCs were highly efficient in generating antigen or tumor-specific T cells with increased effector functions (e.g., cytokine production and tumoricidal activity). SRA/CD204 silencing-enhanced tumor cell death was associated with elevated IFN-γ levels in tumor tissue and increased tumor-infiltrating CD8(+) cells. IFN-γ neutralization or depletion of CD8(+) cells abrogated the SRA/CD204 downregulation-promoted antitumor efficacy, indicating a critical role of IFN-γ-producing CD8(+) T cells. Therefore, blocking SRA/CD204 activity significantly enhances the therapeutic potency of local radiotherapy combined with in situ DC vaccination by promoting a robust systemic antitumor immunity. Further studies are warranted to test this novel combinatorial approach for translating into improved clinical outcomes in patients with prostate cancer.
Collapse
Affiliation(s)
- Chunqing Guo
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO Box 980033, Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Saleem SJ, Martin RK, Morales JK, Sturgill JL, Gibb DR, Graham L, Bear HD, Manjili MH, Ryan JJ, Conrad DH. Cutting edge: mast cells critically augment myeloid-derived suppressor cell activity. THE JOURNAL OF IMMUNOLOGY 2012; 189:511-5. [PMID: 22706087 DOI: 10.4049/jimmunol.1200647] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are primarily recognized for their immunosuppressive properties in malignant disease. However, their interaction with other innate immune cells and their regulation of immune responses, such as in parasitic infection, necessitate further characterization. We used our previously published mouse model of MDSC accumulation to examine the immunoregulatory role of MDSCs in B16 melanoma metastasis and Nippostrongylus brasiliensis infection. In this study, we demonstrate that the activity of MDSCs is dependent on the immune stimuli and subset induced. Monocytic MDSCs predictably suppressed antitumor immune responses but granulocytic MDSCs surprisingly enhanced the clearance of N. brasiliensis infection. Intriguingly, both results were dependent on MDSC interaction with mast cells (MCs), as demonstrated by adoptive-transfer studies in MC-deficient (Kit(Wsh)(/)(Wsh)) mice. These findings were further supported by ex vivo cocultures of MCs and MDSCs, indicating a synergistic increase in cytokine production. Thus, MCs can enhance both immunosuppressive and immunosupportive functions of MDSCs.
Collapse
Affiliation(s)
- Sheinei J Saleem
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hasumi K, Aoki Y, Watanabe R, Hankey KG, Mann DL. Therapeutic response in patients with advanced malignancies treated with combined dendritic cell-activated T cell based immunotherapy and intensity-modulated radiotherapy. Cancers (Basel) 2011; 3:2223-42. [PMID: 24212806 PMCID: PMC3757414 DOI: 10.3390/cancers3022223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 04/14/2011] [Accepted: 04/19/2011] [Indexed: 12/28/2022] Open
Abstract
Successful cancer immunotherapy is confounded by the magnitude of the tumor burden and the presence of immunoregulatory elements that suppress an immune response. To approach these issues, 26 patients with advanced treatment refractory cancer were enrolled in a safety/feasibility study wherein a conventional treatment modality, intensity modulated radiotherapy (IMRT), was combined with dendritic cell-based immunotherapy. We hypothesized that radiation would lower the tumor burdens, decrease the number/function of regulatory cells in the tumor environment, and release products of tumor cells that could be acquired by intratumoral injected immature dendritic cells (iDC). Metastatic lesions identified by CT (computed tomography) were injected with autologous iDC combined with a cytokine-based adjuvant and KLH (keyhole limpet hemocyanin), followed 24 h later by IV-infused T-cells expanded with anti-CD3 and IL-2 (AT). After three to five days, each of the injected lesions was treated with fractionated doses of IMRT followed by another injection of intratumoral iDC and IV-infused AT. No toxicity was observed with cell infusion while radiation-related toxicity was observed in seven patients. Five patients had progressive disease, eight demonstrated complete resolution at treated sites but developed recurrent disease at other sites, and 13 showed complete response at various follow-up times with an overall estimated Kaplan-Meier disease-free survival of 345 days. Most patients developed KLH antibodies supporting our hypothesis that the co-injected iDC are functional with the capacity to acquire antigens from their environment and generate an adaptive immune response. These results demonstrate the safety and effectiveness of this multimodality strategy combining immunotherapy and IMRT in patients with advanced malignancies.
Collapse
Affiliation(s)
- Kenichiro Hasumi
- Hasumi International Research Foundation, Tokyo Research Center, 1-44-6 Asagaya-kita, Suginami- ku, Tokyo 166-0001, Japan; E-Mails: (K.H.); (Y.A.); (R.W.)
| | - Yukimasa Aoki
- Hasumi International Research Foundation, Tokyo Research Center, 1-44-6 Asagaya-kita, Suginami- ku, Tokyo 166-0001, Japan; E-Mails: (K.H.); (Y.A.); (R.W.)
| | - Ryuko Watanabe
- Hasumi International Research Foundation, Tokyo Research Center, 1-44-6 Asagaya-kita, Suginami- ku, Tokyo 166-0001, Japan; E-Mails: (K.H.); (Y.A.); (R.W.)
| | - Kim G. Hankey
- Department of Pathology, University of Maryland School of Medicine, MSTF Room 700, 10 South Pine Street, Baltimore, Maryland 21040, USA; E-Mail: (K.G.H.)
| | - Dean L. Mann
- Department of Pathology, University of Maryland School of Medicine, MSTF Room 700, 10 South Pine Street, Baltimore, Maryland 21040, USA; E-Mail: (K.G.H.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-410-706-1820; Fax: +1-410-706-8414
| |
Collapse
|
20
|
Kujawski M, Zhang C, Herrmann A, Reckamp K, Scuto A, Jensen M, Deng J, Forman S, Figlin R, Yu H. Targeting STAT3 in adoptively transferred T cells promotes their in vivo expansion and antitumor effects. Cancer Res 2010; 70:9599-610. [PMID: 21118964 PMCID: PMC3017475 DOI: 10.1158/0008-5472.can-10-1293] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Adoptive cell therapy with engineered T cells to improve natural immune response and antitumor functions has shown promise for treating cancer. However, the requirement for extensive ex vivo manipulation of T cells and the immunosuppressive effects of the tumor microenvironment limit this therapeutic modality. In the present study, we investigated the possibility to circumvent these limitations by engineering Stat3 -deficient CD8(+) T cells or by targeting Stat3 in the tumor microenvironment. We show that ablating Stat3in CD8(+) T cells prior to their transfer allows their efficient tumor infiltration and robust proliferation, resulting in increased tumor antigen-specific T-cell activity and tumor growth inhibition. For potential clinical translation, we combined adoptive T-cell therapy with a Food and Drug Administration-approved tyrosine kinase inhibitor, sunitinib, in renal cell carcinoma and melanoma tumor models. Sunitinib inhibited Stat3 in dendritic cells and T cells and reduced conversion of transferred FoxP3(-) T cells to tumor-associated regulatory T cells while increasing transferred CD8(+) T-cell infiltration and activation at the tumor site, leading to inhibition of primary tumor growth. These data show that adoptively transferred T cells can be expanded and activated in vivo either by engineering Stat3-silenced T cells or by targeting Stat3 systemically with small-molecule inhibitors.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Line, Tumor
- Cell Proliferation
- Combined Modality Therapy
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Immunotherapy, Adoptive/methods
- Indoles/therapeutic use
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Pyrroles/therapeutic use
- STAT3 Transcription Factor/antagonists & inhibitors
- STAT3 Transcription Factor/deficiency
- STAT3 Transcription Factor/genetics
- Sunitinib
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Burden/drug effects
- Tumor Burden/immunology
Collapse
Affiliation(s)
- Maciej Kujawski
- Department of Cancer Immunotherapeutics & Tumor Immunology, Beckman Research Institute at City of Hope, Duarte, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Mondino A, Dardalhon V, Hess Michelini R, Loisel-Meyer S, Taylor N. Redirecting the immune response: role of adoptive T cell therapy. Hum Gene Ther 2010; 21:533-41. [PMID: 20201627 DOI: 10.1089/hum.2010.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Adoptive T cell therapy is aimed at overcoming constraints of the endogenous immune response. In patients with malignancies, this approach is based on the possibility of administering sufficient numbers of tumor-reactive lymphocytes under conditions in which they will promote a therapeutic response. Although this strategy is potentially applicable to a vast number of malignancies, its efficacy, to date, has been limited. This is likely related to several factors including an insufficient persistence and reactivation of infused cells, insufficient tumor infiltration, and the presence of an immunosuppressive environment. Here, we review the importance of pretransplantation host conditioning and posttransplantation strategies that have been shown to contribute to the therapeutic efficacy of infused T lymphocytes.
Collapse
Affiliation(s)
- Anna Mondino
- Division of Immunology, Transplantation, and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | | | | | | | | |
Collapse
|
22
|
Nakai N, Hartmann G, Kishimoto S, Katoh N. Dendritic cell vaccination in human melanoma: relationships between clinical effects and vaccine parameters. Pigment Cell Melanoma Res 2010; 23:607-19. [DOI: 10.1111/j.1755-148x.2010.00736.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
23
|
Higham EM, Shen CH, Wittrup KD, Chen J. Cutting edge: delay and reversal of T cell tolerance by intratumoral injection of antigen-loaded dendritic cells in an autochthonous tumor model. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:5954-8. [PMID: 20427765 PMCID: PMC3061230 DOI: 10.4049/jimmunol.1000265] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The tumor environment exerts a powerful suppressive influence on infiltrating tumor-reactive T cells. It induces tolerance of adoptively transferred effector T cells as they enter tumors and maintains the tolerance of persisting tumor-infiltrating T cells. In an autochthonous prostate cancer model, in which tumor-reactive CD8 T cells are trackable, we demonstrate that both depletion of endogenous dendritic cells (DCs) and intratumoral injection of Ag-loaded mature DCs delayed the tolerization of tumor-infiltrating effector CD8 T cells. Intratumoral injection of Ag-loaded DCs also reactivated tolerized CD8 T cells in the tumor tissue. The observed effects lasted as long as the injected DCs persisted. These findings are consistent with a critical role of DCs in modulating T cell reactivity in the tumor environment. They also suggest new potential strategies to extend the functionality of transferred effector T cells and to restore function to tolerized tumor-infiltrating T cells for cancer immunotherapy.
Collapse
Affiliation(s)
- Eileen M. Higham
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ching-Hung Shen
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - K. Dane Wittrup
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jianzhu Chen
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
24
|
Salem ML, El-Naggar SA, Cole DJ. Cyclophosphamide induces bone marrow to yield higher numbers of precursor dendritic cells in vitro capable of functional antigen presentation to T cells in vivo. Cell Immunol 2009; 261:134-143. [PMID: 20036354 PMCID: PMC2821961 DOI: 10.1016/j.cellimm.2009.11.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 11/26/2009] [Accepted: 11/30/2009] [Indexed: 02/07/2023]
Abstract
We have shown recently that cyclophosphamide (CTX) treatment induced a marked increase in the numbers of immature dendritic cells (DCs) in blood, coinciding with enhanced antigen-specific responses of the adoptively transferred CD8(+) T cells. Because this DC expansion was preceded by DC proliferation in bone marrow (BM), we tested whether BM post CTX treatment can generate higher numbers of functional DCs. BM was harvested three days after treatment of C57BL/6 mice with PBS or CTX and cultured with GM-CSF/IL-4 in vitro. Compared with control, BM from CTX-treated mice showed faster generation and yielded higher numbers of DCs with superior activation in response to toll-like receptor (TLR) agonists. Vaccination with peptide-pulsed DCs generated from BM from CTX-treated mice induced comparable adjuvant effects to those induced by control DCs. Taken together, post CTX BM harbors higher numbers of DC precursors capable of differentiating into functional DCs, which be targeted to create host microenvironment riches in activated DCs upon treatment with TLR agonists.
Collapse
Affiliation(s)
- Mohamed L Salem
- Surgery Department and Hollings Cancer Center, Medical University of South Carolina, Charleston, 29425, USA.
| | | | | |
Collapse
|