1
|
Dendritic Cell-Based Immunotherapy in Hot and Cold Tumors. Int J Mol Sci 2022; 23:ijms23137325. [PMID: 35806328 PMCID: PMC9266676 DOI: 10.3390/ijms23137325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells mediate innate and adaptive immune responses and are directly involved in the activation of cytotoxic T lymphocytes that kill tumor cells. Dendritic cell-based cancer immunotherapy has clinical benefits. Dendritic cell subsets are diverse, and tumors can be hot or cold, depending on their immunogenicity; this heterogeneity affects the success of dendritic cell-based immunotherapy. Here, we review the ontogeny of dendritic cells and dendritic cell subsets. We also review the characteristics of hot and cold tumors and briefly introduce therapeutic trials related to hot and cold tumors. Lastly, we discuss dendritic cell-based cancer immunotherapy in hot and cold tumors.
Collapse
|
2
|
Mixed cultures of allogeneic dendritic cells are phenotypically and functionally stable - a potential for primary cell-based "off the shelf" product generation. Cent Eur J Immunol 2021; 46:152-161. [PMID: 34764784 PMCID: PMC8568021 DOI: 10.5114/ceji.2021.107555] [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: 06/30/2020] [Accepted: 01/12/2021] [Indexed: 11/29/2022] Open
Abstract
Vaccination against tumors using antigen-pulsed dendritic cell (DC) vaccines has greatly evolved over the last decade, with hundreds of active human clinical trials well on the way. The use of an autologous source for DC-based vaccine therapeutics remains the obvious choice in the majority of clinical studies; however, novel evidence suggests that an allogeneic source of DCs can yield success if administered in the right context. One of the challenges facing successful DC vaccination protocols is the generation of large enough numbers of DCs intended for vaccination and standardization of these procedures. In addition, variations in the quality of DC vaccines due to donor-to-donor variation represent an important therapeutic factor. To this day it has not been shown whether DCs from different donors can readily co-exist within the same co-culture for the extended periods required for vaccine manufacture. We demonstrate that generation of allogeneic DC co-cultures, generated from multiple unrelated donors, allows the preservation of their phenotypical and functional properties in vitro for up to 72 hours. Therefore, in the case of an allogeneic vaccination approach, one could ensure large numbers of DCs generated from a primary cell source intended for multiple vaccinations. By generating large amounts of ex vivo manufactured DCs from multiple donors, this would represent the possibility to ensure sufficient amounts of equipotent “off the shelf” product that could e.g. be used for an entire cohort of patients within a study.
Collapse
|
3
|
Song MJ, Pan QZ, Ding Y, Zeng J, Dong P, Zhao JJ, Tang Y, Li J, Zhang Z, He J, Yang J, Huang Y, Peng R, Wang QJ, Gu JM, He J, Li YQ, Chen SP, Huang R, Zhou ZQ, Yang C, Han Y, Chen H, Liu H, Xia S, Wan Y, Weng DS, Xia L, Zhou FJ, Xia JC. The efficacy and safety of the combination of axitinib and pembrolizumab-activated autologous DC-CIK cell immunotherapy for patients with advanced renal cell carcinoma: a phase 2 study. Clin Transl Immunology 2021; 10:e1257. [PMID: 33717483 PMCID: PMC7927618 DOI: 10.1002/cti2.1257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/15/2021] [Accepted: 02/01/2021] [Indexed: 12/22/2022] Open
Abstract
Objectives Although axitinib has achieved a preferable response rate for advanced renal cell carcinoma (RCC), patient survival remains unsatisfactory. In this study, we evaluated the efficacy and safety of a combination treatment of axitinib and a low dose of pembrolizumab‐activated autologous dendritic cells–co‐cultured cytokine‐induced killer cells in patients with advanced RCC. Methods All adult patients, including treatment‐naive or pretreated with VEGF‐targeted agents, were enrolled from May 2016 to March 2019. Patients received axitinib 5 mg twice daily and pembrolizumab‐activated dendritic cells–co‐cultured cytokine‐induced killer cells intravenously weekly for the first four cycles, every 2 weeks for the next four cycles, and every month thereafter. Results The 43 patients (22 untreated and 21 previously treated) showed a median progression‐free survival (mPFS) of 14.7 months (95% CI, 11.16–18.30). mPFS in treatment‐naive patients was 18.2 months, as compared with 14.4 months in pretreated patients (log‐rank P‐value = 0.07). Overall response rates were 25.6% (95% CI, 13.5–41.2%). Grade 3 or higher adverse events occurred in 5% of patients included hypertension (11.6%) and palmar‐plantar erythrodysesthesia (7.0%). Peripheral blood lymphocyte immunophenotype and serum cytokine profile analyses demonstrated increased antitumor immunity after combination treatment particularly in patients with a long‐term survival benefit, while those with a minimal survival benefit demonstrated an elevated proportion of peripheral CD8+TIM3+ T cells and lower serum‐level immunostimulatory cytokine profile. Conclusions The combination therapy was active and well tolerated for treatment of advanced RCC, either as first‐ or second‐line treatment following other targeted agents. Changes in immunophenotype and serum cytokine profile may be used as prognostic biomarkers.
Collapse
Affiliation(s)
- Meng-Jia Song
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Qiu-Zhong Pan
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Ya Ding
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Jianxiong Zeng
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Pei Dong
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Urology Sun Yat-sen University Cancer Center Guangzhou China
| | - Jing-Jing Zhao
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Yan Tang
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Jingjing Li
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Zhiling Zhang
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Urology Sun Yat-sen University Cancer Center Guangzhou China
| | - Junyi He
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Jieying Yang
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Yue Huang
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Ruiqing Peng
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Qi-Jing Wang
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Jia-Mei Gu
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Jia He
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Yong-Qiang Li
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Shi-Ping Chen
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Rongxing Huang
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Zi-Qi Zhou
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Chaopin Yang
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Yulong Han
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Hao Chen
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Heping Liu
- Guangzhou Yiyang Bio-technology Co., Ltd Guangzhou China
| | - Shangzhou Xia
- Guangzhou Yiyang Bio-technology Co., Ltd Guangzhou China
| | - Yang Wan
- Guangzhou Yiyang Bio-technology Co., Ltd Guangzhou China
| | - De-Sheng Weng
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| | - Liming Xia
- Guangzhou Yiyang Bio-technology Co., Ltd Guangzhou China
| | - Fang-Jian Zhou
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Urology Sun Yat-sen University Cancer Center Guangzhou China
| | - Jian-Chuan Xia
- Collaborative Innovation Center for Cancer Medicine State Key Laboratory of Oncology in South China Sun Yat-sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-sen University Cancer Center Guangzhou China
| |
Collapse
|
4
|
Naseri M, Zöller M, Hadjati J, Ghods R, Ranaei Pirmardan E, Kiani J, Eini L, Bozorgmehr M, Madjd Z. Dendritic cells loaded with exosomes derived from cancer stem cell-enriched spheroids as a potential immunotherapeutic option. J Cell Mol Med 2021; 25:3312-3326. [PMID: 33634564 PMCID: PMC8034455 DOI: 10.1111/jcmm.16401] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/30/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are responsible for therapeutic resistance and recurrence in colorectal cancer. Despite advances in immunotherapy, the inability to specifically eradicate CSCs has led to treatment failure. Hence, identification of appropriate antigen sources is a major challenge in designing dendritic cell (DC)‐based therapeutic strategies against CSCs. Here, in an in vitro model using the HT‐29 colon cancer cell line, we explored the efficacy of DCs loaded with exosomes derived from CSC‐enriched colonospheres (CSCenr‐EXOs) as an antigen source in activating CSC‐specific T‐cell responses. HT‐29 lysate, HT‐29‐EXOs and CSCenr lysate were independently assessed as separate antigen sources. Having confirmed CSCs enrichment in spheroids, CSCenr‐EXOs were purified and characterized, and their impact on DC maturation was investigated. Finally, the impact of the antigen‐pulsed DCs on the proliferation rate and also spheroid destructive capacity of autologous T cells was assessed. CSCenr‐EXOs similar to other antigen groups had no suppressive/negative impacts on phenotypic maturation of DCs as judged by the expression level of costimulatory molecules. Notably, similar to CSCenr lysate, CSCenr‐EXOs significantly increased the IL‐12/IL‐10 ratio in supernatants of mature DCs. CSCenr‐EXO‐loaded DCs effectively promoted T‐cell proliferation. Importantly, T cells stimulated with CSCenr‐EXOs disrupted spheroids' structure. Thus, CSCenr‐EXOs present a novel and promising antigen source that in combination with conventional tumour bulk‐derived antigens should be further explored in pre‐clinical immunotherapeutic settings for the efficacy in hampering recurrence and metastatic spread.
Collapse
Affiliation(s)
- Marzieh Naseri
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Margot Zöller
- Section Pancreas Research, University Hospital of Surgery, Heidelberg, Germany
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ehsan Ranaei Pirmardan
- Department of Radiology, Molecular Biomarkers Nano-imaging Laboratory, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Leila Eini
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Basic Science, Faculty of Veterinary, Science and Research Branch of Islamic, Azad University, Tehran, Iran
| | - Mahmood Bozorgmehr
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
5
|
Kwiatkowski AJ, Stewart JM, Cho JJ, Avram D, Keselowsky BG. Nano and Microparticle Emerging Strategies for Treatment of Autoimmune Diseases: Multiple Sclerosis and Type 1 Diabetes. Adv Healthc Mater 2020; 9:e2000164. [PMID: 32519501 DOI: 10.1002/adhm.202000164] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/12/2020] [Indexed: 02/06/2023]
Abstract
Autoimmune diseases affect 10% of the world's population, and 1 in 200 people worldwide suffer from either multiple sclerosis (MS) or type 1 diabetes (T1D). While the targeted organ systems are different, MS and T1D share similarities in terms of autoreactive immune cells playing a critical role in pathogenesis. Both diseases can be managed only symptomatically without curative remission, and treatment options are limited and non-specific. Most current therapies cause some degree of systemic immune suppression, leaving the patients susceptible to opportunistic infections and other complications. Thus, there is considerable interest in the development of immunotherapies not associated with generalized immune suppression for these diseases. This review presents current and preclinical strategies for MS and T1D treatment, emphasizing those aimed to modulate the immune response, including the most recent strategies for tolerance induction. A central focus is on the emerging approaches using nano- and microparticle platforms, their evolution as immunotherapeutic carriers, including those incorporating specific antigens to induce tolerance and reduce unwanted generalized immune suppression.
Collapse
Affiliation(s)
- Alexander J Kwiatkowski
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Joshua M Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Jonathan J Cho
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Dorina Avram
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
6
|
Cho JJ, Stewart JM, Drashansky TT, Brusko MA, Zuniga AN, Lorentsen KJ, Keselowsky BG, Avram D. An antigen-specific semi-therapeutic treatment with local delivery of tolerogenic factors through a dual-sized microparticle system blocks experimental autoimmune encephalomyelitis. Biomaterials 2017; 143:79-92. [PMID: 28772190 DOI: 10.1016/j.biomaterials.2017.07.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/11/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Antigen-specific treatments are highly desirable for autoimmune diseases in contrast to treatments which induce systemic immunosuppression. A novel antigen-specific therapy has been developed which, when administered semi-therapeutically, is highly efficacious in the treatment of the mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). The treatment uses dual-sized, polymeric microparticles (dMPs) loaded with specific antigen and tolerizing factors for intra- and extra-cellular delivery, designed to recruit and modulate dendritic cells toward a tolerogenic phenotype without systemic release. This approach demonstrated robust efficacy and provided complete protection against disease. Therapeutic efficacy required encapsulation of the factors in controlled-release microparticles and was antigen-specific. Disease blocking was associated with a reduction of infiltrating CD4+ T cells, inflammatory cytokine-producing pathogenic CD4+ T cells, and activated macrophages and microglia in the central nervous system. Furthermore, CD4+ T cells isolated from dMP-treated mice were anergic in response to disease-specific, antigen-loaded splenocytes. Additionally, the frequency of CD86hiMHCIIhi dendritic cells in draining lymph nodes of EAE mice treated with Ag-specific dMPs was reduced. Our findings highlight the efficacy of microparticle-based drug delivery platform to mediate antigen-specific tolerance, and suggest that such a multi-factor combinatorial approach can act to block autoimmunity.
Collapse
Affiliation(s)
- Jonathan J Cho
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Joshua M Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Theodore T Drashansky
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Maigan A Brusko
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ashley N Zuniga
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kyle J Lorentsen
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - Dorina Avram
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
7
|
KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol Immunother 2017; 66:1175-1187. [PMID: 28451792 PMCID: PMC5579171 DOI: 10.1007/s00262-017-2005-z] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 04/18/2017] [Indexed: 12/28/2022]
Abstract
It was reported that PD-L1 expression was correlated with genetic alterations. Whether PD-L1 was regulated by mutant Kirsten rat sarcoma viral oncogene homolog (KRAS) in non-small-cell lung cancer (NSCLC) and the underlying molecular mechanism were largely unknown. In this study, we investigated the correlation between PD-L1 expression and KRAS mutation and the functional significance of PD-1/PD-L1 blockade in KRAS-mutant lung adenocarcinoma. We found that PD-L1 expression was associated with KRAS mutation both in the human lung adenocarcinoma cell lines and tissues. PD-L1 was up-regulated by KRAS mutation through p-ERK but not p-AKT signaling. We also found that KRAS-mediated up-regulation of PD-L1 induced the apoptosis of CD3-positive T cells which was reversed by anti-PD-1 antibody (Pembrolizumab) or ERK inhibitor. PD-1 blocker or ERK inhibitor could recover the anti-tumor immunity of T cells and decrease the survival rates of KRAS-mutant NSCLC cells in co-culture system in vitro. However, Pembrolizumab combined with ERK inhibitor did not show synergistic effect on killing tumor cells in co-culture system. Our study demonstrated that KRAS mutation could induce PD-L1 expression through p-ERK signaling in lung adenocarcinoma. Blockade of PD-1/PD-L1 pathway may be a promising therapeutic strategy for human KRAS-mutant lung adenocarcinoma.
Collapse
|
8
|
Hong S, Chen N, Fang W, Zhan J, Liu Q, Kang S, He X, Liu L, Zhou T, Huang J, Chen Y, Qin T, Zhang Y, Ma Y, Yang Y, Zhao Y, Huang Y, Zhang L. Upregulation of PD-L1 by EML4-ALK fusion protein mediates the immune escape in ALK positive NSCLC: Implication for optional anti-PD-1/PD-L1 immune therapy for ALK-TKIs sensitive and resistant NSCLC patients. Oncoimmunology 2015; 5:e1094598. [PMID: 27141355 DOI: 10.1080/2162402x.2015.1094598] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/03/2015] [Accepted: 09/10/2015] [Indexed: 01/06/2023] Open
Abstract
Driver mutations were reported to upregulate programmed death-ligand 1 (PD-L1) expression. However, how PD-L1 expression and immune function was affected by ALK-TKIs and anti-PD-1/PD-L1 treatment in ALK positive non-small-cell lung cancer (NSCLC) remains poorly understood. In the present study, western-blot, real-time PCR, flow cytometry and immunofluorescence were employed to explore how PD-L1 was regulated by ALK fusion protein. ALK-TKIs and relevant inhibitors were used to identify the downstream signaling pathways involved in PD-L1 regulation. Cell apoptosis, viability and Elisa test were used to study the immune suppression by ALK activation and immune reactivation by ALK-TKIs and/or PD-1 blocking in tumor cells and DC-CIK cells co-culture system. We found that PD-L1 expression was associated with EGFR mutations and ALK fusion genes in NSCLC cell lines. Over-expression of ALK fusion protein increased PD-L1 expression. PD-L1 mediated by ALK fusion protein increased the apoptosis of T cells in tumor cells and DC-CIK cells co-culture system. Inhibiting ALK by sensitive TKIs could enhance the production of IFNγ. Anti-PD-1 antibody was effective in both crizotinib sensitive and resistant NSCLC cells. Synergistic tumor killing effects were not observed with ALK-TKIs and anti-PD-1 antibody combination in co-culture system. ALK-TKIs not only directly inhibited tumor viability but also indirectly enhanced the antitumor immunity via the downregulation of PD-L1. Anti-PD-1/PD-L1 antibodies could be an optional therapy for crizotinib sensitive, especially crizotinib resistant NSCLC patients with ALK fusion gene. Combination of ALK-TKIs and anti-PD-1/PD-L1 antibodies treatment for ALK positive NSCLC warrants more data before moving into clinical practice.
Collapse
Affiliation(s)
- Shaodong Hong
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Nan Chen
- Department of Medical Oncology, the Fifth Affiliated Hospital of Sun Yat-sen University , Zhuhai, Guangdong, China
| | - Wenfeng Fang
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jianhua Zhan
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qing Liu
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shiyang Kang
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiaobo He
- Department of Medical Oncology, the Fifth Affiliated Hospital of Sun Yat-sen University , Zhuhai, Guangdong, China
| | - Lin Liu
- Department of Medical Oncology, the Fifth Affiliated Hospital of Sun Yat-sen University , Zhuhai, Guangdong, China
| | - Ting Zhou
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jiaxing Huang
- Department of Medical Oncology, the Fifth Affiliated Hospital of Sun Yat-sen University , Zhuhai, Guangdong, China
| | - Ying Chen
- Department of Medical Oncology, the Fifth Affiliated Hospital of Sun Yat-sen University , Zhuhai, Guangdong, China
| | - Tao Qin
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yaxiong Zhang
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yuxiang Ma
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yunpeng Yang
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yuanyuan Zhao
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yan Huang
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Li Zhang
- State Key laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Li C, Liang S, Zhang C, Liu Y, Yang M, Zhang J, Zhi X, Pan F, Cui D. Allogenic dendritic cell and tumor cell fused vaccine for targeted imaging and enhanced immunotherapeutic efficacy of gastric cancer. Biomaterials 2015; 54:177-87. [DOI: 10.1016/j.biomaterials.2015.03.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/13/2015] [Accepted: 03/15/2015] [Indexed: 12/26/2022]
|
10
|
Gustafson MP, Lin Y, Bleeker JS, Warad D, Tollefson MK, Crispen PL, Bulur PA, Harrington SM, Laborde RR, Gastineau DA, Leibovich BC, Cheville JC, Kwon ED, Dietz AB. Intratumoral CD14+ Cells and Circulating CD14+HLA-DRlo/neg Monocytes Correlate with Decreased Survival in Patients with Clear Cell Renal Cell Carcinoma. Clin Cancer Res 2015; 21:4224-33. [PMID: 25999436 DOI: 10.1158/1078-0432.ccr-15-0260] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 05/04/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Immunotherapeutic strategies to treat patients with renal cell carcinoma (RCC) offer new opportunities for disease management. Further improvements to immunotherapy will require additional understanding of the host response to RCC development. EXPERIMENTAL DESIGN Using a novel approach to understanding the immune status of cancer patients, we previously showed that patients with a certain immune profile had decreased overall survival. Here, we examine in more detail the phenotypic changes in peripheral blood and the potential consequences of these changes in RCC patients. RESULTS We found that CD14(+)HLA-DR(lo/neg) monocytes were the most predominant phenotypic change in peripheral blood of RCC patients, elevated nearly 5-fold above the average levels measured in healthy volunteers. Intratumoral and peritumoral presence of CD14 cells was an independent prognostic factor for decreased survival in a cohort of 375 RCC patients. The amount of peripheral blood CD14(+)HLA-DR(lo/neg) monocytes was found to correlate with the intensity of CD14 staining in tumors, suggesting that the measurement of these cells in blood may be a suitable surrogate for monitoring patient prognosis. The interaction of monocytes and tumor cells triggers changes in both cell types with a loss of HLA-DR expression in monocytes, increases of monocyte survival factors such as GM-CSF in tumors, and increased production of angiogenic factors, including FGF2. CONCLUSIONS Our results suggest a model of mutually beneficial interactions between tumor cells and monocytes that adversely affect patient outcome.
Collapse
Affiliation(s)
- Michael P Gustafson
- Human Cellular Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | | | - Deepti Warad
- Division of Pediatric Hematology/Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Paul L Crispen
- Department of Urology, University of Florida, Gainesville, Florida
| | - Peggy A Bulur
- Human Cellular Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Rebecca R Laborde
- Human Cellular Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Dennis A Gastineau
- Human Cellular Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota. Division of Hematology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Eugene D Kwon
- Department of Urology, Mayo Clinic, Rochester, Minnesota
| | - Allan B Dietz
- Human Cellular Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
11
|
Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2014; 1:1111-1134. [PMID: 23170259 PMCID: PMC3494625 DOI: 10.4161/onci.21494] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) occupy a central position in the immune system, orchestrating a wide repertoire of responses that span from the development of self-tolerance to the elicitation of potent cellular and humoral immunity. Accordingly, DCs are involved in the etiology of conditions as diverse as infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. During the last decade, several methods have been developed to load DCs with tumor-associated antigens, ex vivo or in vivo, in the attempt to use them as therapeutic anticancer vaccines that would elicit clinically relevant immune responses. While this has not always been the case, several clinical studies have demonstrated that DC-based anticancer vaccines are capable of activating tumor-specific immune responses that increase overall survival, at least in a subset of patients. In 2010, this branch of clinical research has culminated with the approval by FDA of a DC-based therapeutic vaccine (sipuleucel-T, Provenge®) for use in patients with asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer. Intense research efforts are currently dedicated to the identification of the immunological features of patients that best respond to DC-based anticancer vaccines. This knowledge may indeed lead to personalized combination strategies that would extend the benefit of DC-based immunotherapy to a larger patient population. In addition, widespread enthusiasm has been generated by the results of the first clinical trials based on in vivo DC targeting, an approach that holds great promises for the future of DC-based immunotherapy. In this Trial Watch, we will summarize the results of recently completed clinical trials and discuss the progress of ongoing studies that have evaluated/are evaluating DC-based interventions for cancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Cicchelero L, de Rooster H, Sanders NN. Various ways to improve whole cancer cell vaccines. Expert Rev Vaccines 2014; 13:721-35. [PMID: 24758597 DOI: 10.1586/14760584.2014.911093] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunotherapy based on whole cancer cell vaccines is regarded as a promising avenue for cancer treatment. However, limited efficacy in the first human clinical trials calls for more optimized whole cancer cell vaccines and better patient selection. It is suggested that whole cancer cell vaccines consist preferably of immunogenically killed autologous cancer stem cells associated with dendritic cells. Adjuvants should stimulate both immune effector cells and memory cells, which could be achieved through their correct dosage and timing of administration. There are indications that whole cancer cell vaccination is less effective in patients who are immunocompromised, who have specific genetic defects in their immune or cancer cells, as well as in patients in an advanced cancer stage. However, such patients form the bulk of enrolled patients in clinical trials, prohibiting an objective evaluation of the true potential of whole cancer cell immunotherapy. Each key point will be discussed.
Collapse
Affiliation(s)
- Laetitia Cicchelero
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | | | | |
Collapse
|
13
|
Autologous CIK cell immunotherapy in patients with renal cell carcinoma after radical nephrectomy. Clin Dev Immunol 2013; 2013:195691. [PMID: 24382970 PMCID: PMC3872096 DOI: 10.1155/2013/195691] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/13/2013] [Indexed: 11/17/2022]
Abstract
Objective. To evaluate the efficacy of autologous cytokine-induced killer (CIK) cells in patients with renal cell carcinoma (RCC). Methods. 20 patients diagnosed with TNM stage I or II RCC were randomly divided into two groups, a CIK cell treatment group and a control group. The endpoint was progression-free survival (PFS) evaluated by Kaplan-Meier analyses. Results. CD3+, CD3+/CD8+, CD3+/CD4+, and CD3+/CD56+ levels increased after CIK cell culture (P < 0.01). The median PFS in CIK cell treatment group was significantly longer than that in control group (PFS, 32.2 months versus 21.6 months; log-rank, P = 0.032), all patients were alive during the course of followup, and there are no statistically significant differences between two groups in OS (log-rank, P = 0.214). Grade III or greater adverse events were not observed. Conclusions. CIK cells treatment could prolong survival in patients with RCC after radical nephrectomy and showed acceptable curative effect with potential enhancement of cellular immune function. This trial is registered with Clinicaltrials.gov NCT01799083.
Collapse
|
14
|
Chen X, Liu Z, Huang Y, Li R, Zhang H, Dong S, Ge C, Zhang Z, Wang Y, Wang Y, Xue Y, Li Z, Song X. Superior anti-tumor protection and therapeutic efficacy of vaccination with dendritic cell/tumor cell fusion hybrids for murine Lewis lung carcinoma. Autoimmunity 2013; 47:46-56. [PMID: 24191684 DOI: 10.3109/08916934.2013.850080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The development of protocols for the ex vivo generation of dendritic cells (DCs) has led to intensive research into their potential use in immunotherapy in the treatment of cancer. In this study, we examined the efficacy of dendritic cell-tumor cell fusion hybrid vaccines in eliciting an immune response against Lewis lung carcinoma (LLC) cells, as compared to other types of tumor vaccines. In addition, we also tested whether the efficacy of the vaccines was affected by the route of administration. Four different tumor vaccines were compared: (1) HC (hybrid cell), consisting of DC/LLC hybrids; (2) DC+LLC (DCs pulsed with apoptotic LLCs); (3) DC without antigen loading/pulsing; (4) LLC (apoptotic/irradiated tumor cells). We also compared four different routes of administration for each vaccine: (1) Preimmunization; (2) Vaccination therapy; (3) Adoptive immunotherapy; (4) Vaccination therapy combined with adoptive immunotherapy. Anti-tumor immunity was assessed in vivo and the CTL (cytotoxic T lymphocyte) response as well as the expression of key cytokines, IFN-γ and IL-10 were further evaluated using in vitro assays. RESULTS Our data demonstrate that vaccination with HC hybrids provides more effective anti-tumor protective immunity and significantly greater therapeutic immunity than vaccination with DC+LLC, DC or LLC. Most remarkably, vaccination therapy with HC hybrids was more successful than combination (vaccination + adoptive) therapy for the induction of anti-tumor responses. Splenocytes harvested from mice immunized with HC hybrids demonstrated the greatest cytotoxic T lymphocyte (CTL) activity and their production of IFN-γ was high, while their production of IL-10 was very low. CONCLUSIONS Our results suggest that vaccination therapy with DC-tumor cell fusion hybrids provides more effective protection against lung cancer.
Collapse
|
15
|
Cintolo JA, Datta J, Mathew SJ, Czerniecki BJ. Dendritic cell-based vaccines: barriers and opportunities. Future Oncol 2013; 8:1273-99. [PMID: 23130928 DOI: 10.2217/fon.12.125] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have several characteristics that make them an ideal vehicle for tumor vaccines, and with the first US FDA-approved DC-based vaccine in use for the treatment of prostate cancer, this technology has become a promising new therapeutic option. However, DC-based vaccines face several barriers that have limited their effectiveness in clinical trials. A major barrier includes the activation state of the DC. Both DC lineage and maturation signals must be selected to optimize the antitumor response and overcome immunosuppressive effects of the tumor microenvironment. Another barrier to successful vaccination is the selection of target antigens that will activate both CD8(+) and CD4(+) T cells in a potent, immune-specific manner. Finally, tumor progression and immune dysfunction limit vaccine efficacy in advanced stages, which may make DC-based vaccines more efficacious in treating early-stage disease. This review underscores the scientific basis and advances in the development of DC-based vaccines, focuses on current barriers to success and highlights new research opportunities to address these obstacles.
Collapse
Affiliation(s)
- Jessica A Cintolo
- Department of Surgery & Harrison Department of Surgical Research, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
16
|
Wang J, Liao L, Tan J. Dendritic cell-based vaccination for renal cell carcinoma: challenges in clinical trials. Immunotherapy 2013; 4:1031-42. [PMID: 23148755 DOI: 10.2217/imt.12.107] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After decades of research, dendritic cell (DC)-based vaccines for renal cell carcinoma have progressed from preclinical rodent models and safety assessments to Phase I/II clinical trials. DC vaccines represent a promising therapy that has produced measurable immunological responses and prolonged survival rates. However, there is still much room to improve in terms of therapeutic efficacy. The key issues that affect the efficiency and reliability of DC therapy include the selection of patients who will respond best to treatment, the proper preparation and administration of DC vaccines, and a combination of DC vaccination with other immune-enhancing therapies (e.g., removal of Tregs, CTLA-4 blockade and lymphodepletion). Additional antiangiogenic agents will hopefully lead to greater survival benefits for patients in early disease stages. This review focuses on the different approaches of DC-based vaccination against renal cell carcinoma and potential strategies to enhance the efficacy of DC vaccination.
Collapse
Affiliation(s)
- Jin Wang
- Organ Transplant Institute, Fuzhou General Hospital, Xiamen University, Fuzhou, China
| | | | | |
Collapse
|
17
|
Flörcken A, Kopp J, van Lessen A, Movassaghi K, Takvorian A, Jöhrens K, Möbs M, Schönemann C, Sawitzki B, Egerer K, Dörken B, Pezzutto A, Westermann J. Allogeneic partially HLA-matched dendritic cells pulsed with autologous tumor cell lysate as a vaccine in metastatic renal cell cancer: a clinical phase I/II study. Hum Vaccin Immunother 2013; 9:1217-27. [PMID: 23458999 DOI: 10.4161/hv.24149] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Multi-kinase inhibitors have been established for the treatment of advanced renal cell cancer, but long-term results are still disappointing and immunotherapeutic approaches remain an interesting experimental option particularly in patients with a low tumor burden. DC are crucial for antigen-specific MHC-restricted T cell immunity. Furthermore, allogeneic HLA-molecules pose a strong immunogenic signal and may help to induce tumor-specific T cell responses. In this phase I/II trial, 7 patients with histologically confirmed progressive metastatic RCC were immunized repetitively with 1 × 10 (7) allogeneic partially HLA-matched DC pulsed with autologous tumor lysate following a schedule of 8 vaccinations over 20 weeks. Patients also received 3 Mio IE IL-2 s.c. once daily starting in week 4. Primary endpoints of the study were feasibility and safety. Secondary endpoints were immunological and clinical responses. Vaccination was feasible and safe with no severe toxicity being observed. No objective response could be documented. However, while all patients had documented progress at study entry, 29% of the patients showed SD throughout the study with a mean TTP of 24.6 weeks (range 5 to 96 weeks). In 3/7 patients, TH1-polarized immune responses against RCC-associated antigens were observed. In one patient showing a minimal clinical response and a TTP of 96 weeks, clonally proliferated T cells against yet undefined antigens were induced by the vaccine. Vaccination with tumor antigen loaded DC remains an interesting experimental approach, but should rather be applied in the situation of minimal residual disease after systemic therapy. Additional depletion of regulatory cells might be a promising strategy.
Collapse
Affiliation(s)
- Anne Flörcken
- Department of Hematology; Oncology, and Tumor Immunology; Charité University Medicine Berlin; Campus Virchow- Klinikum; Berlin, Germany; Department of Hematology; Oncology, and Tumor Immunology; Charité University Medicine Berlin; Campus Benjamin Franklin; Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Pohla H, Buchner A, Stadlbauer B, Frankenberger B, Stevanovic S, Walter S, Frank R, Schwachula T, Olek S, Kopp J, Willimsky G, Stief CG, Hofstetter A, Pezzutto A, Blankenstein T, Oberneder R, Schendel DJ. High immune response rates and decreased frequencies of regulatory T cells in metastatic renal cell carcinoma patients after tumor cell vaccination. Mol Med 2013; 18:1499-508. [PMID: 23269976 DOI: 10.2119/molmed.2012.00221] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/19/2012] [Indexed: 11/06/2022] Open
Abstract
Our previously reported phase I clinical trial with the allogeneic gene-modified tumor cell line RCC-26/CD80/IL-2 showed that vaccination was well tolerated and feasible in metastatic renal cell carcinoma (RCC) patients. Substantial disease stabilization was observed in most patients despite a high tumor burden at study entry. To investigate alterations in immune responses that might contribute to this effect, we performed an extended immune monitoring that included analysis of reactivity against multiple antigens, cytokine/chemokine changes in serum and determination of the frequencies of immune suppressor cell populations, including natural regulatory T cells (nTregs) and myeloid-derived suppressor cell subsets (MDSCs). An overall immune response capacity to virus-derived control peptides was present in 100% of patients before vaccination. Vaccine-induced immune responses to tumor-associated antigens occurred in 75% of patients, demonstrating the potent immune stimulatory capacity of this generic vaccine. Furthermore, some patients reacted to peptide epitopes of antigens not expressed by the vaccine, showing that epitope-spreading occurred in vivo. Frequencies of nTregs and MDSCs were comparable to healthy donors at the beginning of study. A significant decrease of nTregs was detected after vaccination (p = 0.012). High immune response rates, decreased frequencies of nTregs and a mixed T helper 1/T helper 2 (T(H)1/T(H)2)-like cytokine pattern support the applicability of this RCC generic vaccine for use in combination therapies.
Collapse
Affiliation(s)
- Heike Pohla
- Laboratory of Tumor Immunology, LIFE Center, Ludwig Maximilians University, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bhargava A, Mishra D, Banerjee S, Mishra PK. Dendritic cell engineering for tumor immunotherapy: from biology to clinical translation. Immunotherapy 2012; 4:703-18. [PMID: 22853757 DOI: 10.2217/imt.12.40] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Dendritic cells (DCs) are the most potent APCs, with the ability to orchestrate a repertoire of immune responses. DCs play a pivotal role in the initiation, programming and regulation of tumor-specific immune responses, as they are poised to take up, process and present tumor antigens to naive or effector T lymphocytes. Although, to an extent, DC-based immunotherapeutic strategies have successfully induced specific anti-tumor responses in animal models, their clinical efficacy has rarely been translated into the clinic. This article attempts to present a complete picture of recent developments of DC-based therapeutic strategies addressing multiple components of tumor immunoenvironment. It also showcases certain practical intricacies in order to explore novel strategies for providing new impetus to DC-based cancer vaccination.
Collapse
Affiliation(s)
- Arpit Bhargava
- Division of Translational Research, Tata Memorial Centre, ACTREC, India
| | | | | | | |
Collapse
|
20
|
Kroeze SG, Daenen LG, Nijkamp MW, Roodhart JM, de Gast GC, Bosch JR, Jans JJ. Radio Frequency Ablation Combined with Interleukin-2 Induces an Antitumor Immune Response to Renal Cell Carcinoma in a Murine Model. J Urol 2012; 188:607-14. [DOI: 10.1016/j.juro.2012.03.116] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Indexed: 11/28/2022]
Affiliation(s)
| | - Laura G.M. Daenen
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten W. Nijkamp
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeanine M.L. Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gijsbert C. de Gast
- Department of Medical Oncology and Immunotherapy, Dutch Cancer Institute, Amsterdam, The Netherlands
| | - J.L.H. Ruud Bosch
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Judith J.M. Jans
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
21
|
Abstract
Although several cytokines have shown antitumor activity in renal cell carcinoma (RCC), the most consistent results have been reported with interleukin-2 (IL-2) and interferon (IFN). Recent insights into how the immune response to a tumor is regulated hold the promise of allowing patients to obtain a durable response to immunotherapy, perhaps without the significant toxicity associated with conventional approaches. This review describes how improvements in patient selection, combination therapy, and investigational agents might expand and better define the role of immunotherapy in metastatic RCC.
Collapse
|
22
|
Zhang X, de Chickera SN, Willert C, Economopoulos V, Noad J, Rohani R, Wang AY, Levings MK, Scheid E, Foley R, Foster PJ, Dekaban GA. Cellular magnetic resonance imaging of monocyte-derived dendritic cell migration from healthy donors and cancer patients as assessed in a scid mouse model. Cytotherapy 2011; 13:1234-48. [PMID: 21923625 DOI: 10.3109/14653249.2011.605349] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS. The use of dendritic cells (DC) as an adjuvant in cell-based immunotherapeutic cancer vaccines is a growing field of interest. A reliable and non-invasive method to track the fate of autologous DC following their administration to patients is required in order to confirm that clinically sufficient numbers are reaching the lymph node (LN). We demonstrate that an immunocompromised mouse model can be used to conduct translational studies employing cellular magnetic resonance imaging (MRI). Such studies can provide clinically relevant information regarding the migration potential of clinical-grade DC used in cancer immunotherapies. METHODS. Human monocyte-derived dendritic cells (mo-DC) were generated from negatively selected monocytes obtained from either healthy donors or cancer patients. DC were labeled with superparamagnetic iron oxide (SPIO) nanoparticles in order to track them in vivo in a CB17scid mouse model using cellular MRI. SPIO did not have any adverse effects on DC phenotype or function, independent of donor type. Cellular MRI readily detected migration of SPIO-loaded DC in CB17scid mice. No differences in migration were observed between DC obtained from healthy donors and those obtained from donors undergoing autologous stem cell transplant for cancer therapy. CONCLUSIONS. Cellular MRI provided semi-quantitative image data that corresponded with data obtained by digital morphometry, validating cellular MRI's potential to assess DC migration in DC-based cancer immunotherapy clinical trials.
Collapse
Affiliation(s)
- Xizhong Zhang
- BioTherapeutics Research Laboratory, University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Luo W, Zhang XB, Huang YT, Hao PP, Jiang ZM, Wen Q, Zhou MQ, Jin Q, Ma L. Development of genetically engineered CD4+ and CD8+ T cells expressing TCRs specific for a M. tuberculosis 38-kDa antigen. J Mol Med (Berl) 2011; 89:903-13. [PMID: 21556811 DOI: 10.1007/s00109-011-0760-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 03/16/2011] [Accepted: 04/04/2011] [Indexed: 10/18/2022]
Abstract
Cell-mediated immunity is critical to the clearance of Mycobacterium tuberculosis due to the primarily intracellular niche of this pathogen. Adoptive transfer of M. tuberculosis-specific effector T cells has been shown to confer immunity to M. tuberculosis-infected recipients resulting in M. tuberculosis clearance. However, it is difficult to generate sufficient numbers of M. tuberculosis antigen-specific T cells in a short time. Recent studies have developed T cell receptor (TCR) gene-modified T cells that allow for the rapid generation of large numbers of antigen-specific T cells. Many TCRs that target various tumor and viral antigens have now been isolated and shown to have functional activity. Nevertheless, TCRs specific for intracellular bacterial antigens (including M. tuberculosis antigens) have yet to be isolated and their functionality confirmed. We isolated M. tuberculosis 38-kDa antigen-specific HLA class I and class II-restricted TCRs and modified the TCR gene C regions by substituting nine amino acids with their murine TCR homologs (minimal murinization). Results showed that both wild-type and minimal murinized TCR genes were successfully cloned into retroviral vectors and transduced into primary CD4(+) and CD8(+) T cells and displayed anti-M. tuberculosis activity. As expected, minimal murinized TCRs displayed higher cell surface expression levels and stronger anti-M. tuberculosis activity than wild-type TCRs. To the best of our knowledge, this is the first report describing TCRs targeting M. tuberculosis antigens and this investigation provides the basis for future TCR gene-based immunotherapies that can be designed for the treatment of immunocompromised M. tuberculosis-infected patients.
Collapse
Affiliation(s)
- Wei Luo
- Institute of Molecular Immunology, School of Biotechnology, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Immunologic monitoring of cellular responses by dendritic/tumor cell fusion vaccines. J Biomed Biotechnol 2011; 2011:910836. [PMID: 21541197 PMCID: PMC3085507 DOI: 10.1155/2011/910836] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 11/11/2010] [Accepted: 02/27/2011] [Indexed: 12/22/2022] Open
Abstract
Although dendritic cell (DC)- based cancer vaccines induce effective antitumor activities in murine models, only limited therapeutic results have been obtained in clinical trials. As cancer vaccines induce antitumor activities by eliciting or modifying immune responses in patients with cancer, the Response Evaluation Criteria in Solid Tumors (RECIST) and WHO criteria, designed to detect early effects of cytotoxic chemotherapy in solid tumors, may not provide a complete assessment of cancer vaccines. The problem may, in part, be resolved by carrying out immunologic cellular monitoring, which is one prerequisite for rational development of cancer vaccines. In this review, we will discuss immunologic monitoring of cellular responses for the evaluation of cancer vaccines including fusions of DC and whole tumor cell.
Collapse
|
25
|
Balan S, Kale VP, Limaye LS. A large number of mature and functional dendritic cells can be efficiently generated from umbilical cord blood-derived mononuclear cells by a simple two-step culture method. Transfusion 2011; 50:2413-23. [PMID: 20497510 DOI: 10.1111/j.1537-2995.2010.02706.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Advances in the past two decades in dendritic cell (DC) biology paved the way to exploit them as a promising tool in cancer immunotherapy. The prerequisite for DC vaccine preparations is large-scale in vitro generations of homogeneous, mature, and functional DCs. Frequent improvements are being made in the existing in vitro DC production protocols to achieve this goal. In our previous study we reported a large-scale generation of mature, functional DCs from umbilical cord blood (UCB) CD34+ cells. Here we report that this method can be used for the efficient generation of DCs from UCB mononuclear cells (MNCs) and thus the hematopoietic stem cell isolation step is not essential. STUDY DESIGN AND METHODS MNCs or CD34+ cells isolated from the same cord blood (CB) samples were used for the generation of DCs. DCs were characterized for morphology, phenotype, and functional assays including antigen uptake, chemotaxis, and mixed leukocyte reaction. Similarly DCs generated from the MNCs of same fresh and frozen CB units were compared. RESULTS The morphologic, phenotypic, and functional characterization of the DCs generated from various sets show that they were comparable in nature irrespective of the starting population used. CONCLUSION We conclude that the CD34+ isolation step is not essential for the generation of mature, functional DCs and thus can be eliminated. More importantly, we show that DCs can be generated with equal efficiency from the MNCs of frozen CB units. Our culture method will be useful for exploiting the potential of UCB as an additional source for allogeneic DCs in the clinical settings.
Collapse
Affiliation(s)
- Sreekumar Balan
- National Centre for Cell Science, Pune University Campus, Pune, India
| | | | | |
Collapse
|
26
|
Immunotherapy for renal cell carcinoma. Clin Dev Immunol 2011; 2010:284581. [PMID: 21253521 PMCID: PMC3022170 DOI: 10.1155/2010/284581] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 11/29/2010] [Indexed: 11/30/2022]
Abstract
Immunotherapy plays a significant role in the management of renal cell carcinoma (RCC) patients with metastatic disease because RCC is highly resistant to both chemotherapy and radiation therapy. Many reports illustrate various approaches to the treatment of RCC, such as cytokine-, antigen- or dendritic cell- (DC-) based immunotherapy, and the safety and effectiveness of immunotherapy have been highlighted by multiple clinical trials. Although antitumor immune responses and clinically significant outcomes have been achieved in these trials, the response rate is still low, and very few patients show long-term clinical improvement. Recently, the importance of immune regulation by antigen-presenting cells (APC) and regulatory T cells (Treg cells) has also been discussed. The authors outline the principles of cell-mediated tumor immunotherapy and discuss clinical trials of immunotherapy for RCC.
Collapse
|
27
|
Dendritic Cell-Tumor Cell Fusion Vaccines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:177-86. [DOI: 10.1007/978-94-007-0763-4_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
28
|
Regulation of tumor immunity by tumor/dendritic cell fusions. Clin Dev Immunol 2010; 2010:516768. [PMID: 21048993 PMCID: PMC2964897 DOI: 10.1155/2010/516768] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Accepted: 09/22/2010] [Indexed: 02/07/2023]
Abstract
The goal of cancer vaccines is to induce antitumor immunity that ultimately will reduce tumor burden in tumor environment. Several strategies involving dendritic cells- (DCs)- based vaccine incorporating different tumor-associated antigens to induce antitumor immune responses against tumors have been tested in clinical trials worldwide. Although DCs-based vaccine such as fusions of whole tumor cells and DCs has been proven to be clinically safe and is efficient to enhance antitumor immune responses for inducing effective immune response and for breaking T-cell tolerance to tumor-associated antigens (TAAs), only a limited success has occurred in clinical trials. This paper reviews tumor immune escape and current strategies employed in the field of tumor/DC fusions vaccine aimed at enhancing activation of TAAs-specific cytotoxic T cells in tumor microenvironment.
Collapse
|
29
|
Progress in Tumor-Dentritic Cell Hybrid Vaccines*. PROG BIOCHEM BIOPHYS 2010. [DOI: 10.3724/sp.j.1206.2010.00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Mito K, Sugiura K, Ueda K, Hori T, Akazawa T, Yamate J, Nakagawa H, Hatoya S, Inaba M, Inoue N, Ikehara S, Inaba T. IFNγ Markedly Cooperates with Intratumoral Dendritic Cell Vaccine in Dog Tumor Models. Cancer Res 2010; 70:7093-101. [DOI: 10.1158/0008-5472.can-10-0600] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
31
|
Chi N, Maranchie JK, Appleman LJ, Storkus WJ. Update on vaccine development for renal cell cancer. Res Rep Urol 2010; 2:125-41. [PMID: 24198621 PMCID: PMC3703676 DOI: 10.2147/rru.s7242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) remains a significant health concern that frequently presents as metastatic disease at the time of initial diagnosis. Current first-line therapeutics for the advanced-stage RCC include antiangiogenic drugs that have yielded high rates of objective clinical response; however, these tend to be transient in nature, with many patients becoming refractory to chronic treatment with these agents. Adjuvant immunotherapies remain viable candidates to sustain disease-free and overall patient survival. In particular, vaccines designed to optimize the activation, maintenance, and recruitment of specific immunity within or into the tumor site continue to evolve. Based on the integration of increasingly refined immunomonitoring systems in both translational models and clinical trials, allowing for the improved understanding of treatment mechanism(s) of action, further refined (combinational) vaccine protocols are currently being developed and evaluated. This review provides a brief history of RCC vaccine development, discusses the successes and limitations in such approaches, and provides a rationale for developing combinational vaccine approaches that may provide improved clinical benefits to patients with RCC.
Collapse
Affiliation(s)
- Nina Chi
- Department of immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | | |
Collapse
|
32
|
Cancer vaccine by fusions of dendritic and cancer cells. Clin Dev Immunol 2010; 2009:657369. [PMID: 20182533 PMCID: PMC2825547 DOI: 10.1155/2009/657369] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Accepted: 12/09/2009] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells and play a central role in the initiation and regulation of primary immune responses. Therefore, their use for the active immunotherapy against cancers has been studied with considerable interest. The fusion of DCs with whole tumor cells represents in many ways an ideal approach to deliver, process, and subsequently present a broad array of tumor-associated antigens, including those yet to be unidentified, in the context of DCs-derived costimulatory molecules. DCs/tumor fusion vaccine stimulates potent antitumor immunity in the animal tumor models. In the human studies, T cells stimulated by DC/tumor fusion cells are effective in lysis of tumor cells that are used as the fusion partner. In the clinical trials, clinical and immunological responses were observed in patients with advanced stage of malignant tumors after being vaccinated with DC/tumor fusion cells, although the antitumor effect is not as vigorous as in the animal tumor models. This review summarizes recent advances in concepts and techniques that are providing new impulses to DCs/tumor fusions-based cancer vaccination.
Collapse
|