1
|
Spring J, Gurbuxani S, Golovkina T. Microbiota does not influence tumor development in two models of heritable cancer. mBio 2025; 16:e0386624. [PMID: 39969175 PMCID: PMC11898629 DOI: 10.1128/mbio.03866-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
Microbial impact on tumorigenesis of heritable cancers proximal to the gut is well-documented. Whether the microbiota influences cancers arising from inborn mutations at sites distal to the gut is undetermined. Using two models of heritable cancer, Trp53-deficient mice and Wnt1-transgenic mice, and a gnotobiotic approach, we found the microbiota to be inconsequential for tumor development. This work furthers our understanding of the degree of the microbial impact on tumor development. IMPORTANCE The influence of the microbiome on the development of cancer is well-documented with many if not all published studies reporting either a positive or a negative impact. None of the published studies, however, presented data on the influence of the microbiome on the development of heritable cancer. We find that the microbiota has no influence on cancer development in two models of spontaneous cancers driven by germline Trp53 deficiency and constitutive Wnt1 signaling.
Collapse
Affiliation(s)
- Jessica Spring
- Committee on Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Sandeep Gurbuxani
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Tatyana Golovkina
- Committee on Microbiology, University of Chicago, Chicago, Illinois, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
- Committee on Genetics, Genomics and System Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
2
|
Zhang T, Yu W, Cheng X, Yeung J, Ahumada V, Norris PC, Pearson MJ, Yang X, van Deursen W, Halcovich C, Nassar A, Vesely MD, Zhang Y, Zhang JP, Ji L, Flies DB, Liu L, Langermann S, LaRochelle WJ, Humphrey R, Zhao D, Zhang Q, Zhang J, Gu R, Schalper KA, Sanmamed MF, Chen L. Up-regulated PLA2G10 in cancer impairs T cell infiltration to dampen immunity. Sci Immunol 2024; 9:eadh2334. [PMID: 38669316 DOI: 10.1126/sciimmunol.adh2334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 10/19/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
T cells are often absent from human cancer tissues during both spontaneously induced immunity and therapeutic immunotherapy, even in the presence of a functional T cell-recruiting chemokine system, suggesting the existence of T cell exclusion mechanisms that impair infiltration. Using a genome-wide in vitro screening platform, we identified a role for phospholipase A2 group 10 (PLA2G10) protein in T cell exclusion. PLA2G10 up-regulation is widespread in human cancers and is associated with poor T cell infiltration in tumor tissues. PLA2G10 overexpression in immunogenic mouse tumors excluded T cells from infiltration, resulting in resistance to anti-PD-1 immunotherapy. PLA2G10 can hydrolyze phospholipids into small lipid metabolites, thus inhibiting chemokine-mediated T cell mobility. Ablation of PLA2G10's enzymatic activity enhanced T cell infiltration and sensitized PLA2G10-overexpressing tumors to immunotherapies. Our study implicates a role for PLA2G10 in T cell exclusion from tumors and suggests a potential target for cancer immunotherapy.
Collapse
Affiliation(s)
- Tianxiang Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Weiwei Yu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoxiao Cheng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jacky Yeung
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Viviana Ahumada
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Xuan Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Christina Halcovich
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ala Nassar
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew D. Vesely
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Yu Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jian-Ping Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lan Ji
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | - Dejian Zhao
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | - Qiuyu Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jindong Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Runxia Gu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Program of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Spring J, Gurbuxani S, Golovkina T. Microbiota may affect the tumor type but not overall tumor development in two models of heritable cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.561890. [PMID: 37873087 PMCID: PMC10592741 DOI: 10.1101/2023.10.11.561890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Microbial impact on tumorigenesis of heritable cancers proximal to the gut is well documented. Whether the microbiota influences cancers arising from inborn mutations at sites distal to the gut is undetermined. Using two models of heritable cancer, we found the microbiota to be inconsequential for tumor development. However, the type of tumor that develops may be influenced by the microbiota. This work furthers our understanding of the microbial impact on tumor development.
Collapse
|
4
|
Jiang T, Chen X, Ren X, Yang JM, Cheng Y. Emerging role of autophagy in anti-tumor immunity: Implications for the modulation of immunotherapy resistance. Drug Resist Updat 2021; 56:100752. [PMID: 33765484 DOI: 10.1016/j.drup.2021.100752] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapies such as CAR-T cell transfer and antibody-targeted therapy have produced promising clinical outcomes in patients with advanced and metastatic cancer that are resistant to conventional therapies. However, with increasing use of cancer immunotherapy in clinical treatment, multiple therapy-resistance mechanisms have gradually emerged. The tumor microenvironment (TME), an integral component of cancer, can significantly influence the therapeutic response. Thus, it is worth exploring the potential of TME in modulating therapy resistance, in the hope to devise novel strategies to reinforcing anti-cancer treatments such as immunotherapy. As a crucial recycling process in the complex TME, the role of autophagy in tumor immunity has been increasingly appreciated. Firstly, autophagy in tumor cells can affect their immune response through modulating MHC-I-antigen complexes, thus modulating immunogenic tumor cell death, changing functions of immune cells via secretory autophagy, reducing the NK- and CTL-mediated cell lysis and degradation of immune checkpoint proteins. Secondly, autophagy is critical for the differentiation, maturation and survival of immune cells in the TME and can significantly affect the immune function of these cells, thereby regulating the anti-tumor immune response. Thirdly, alteration of autophagic activity in stromal cells, especially in fibroblasts, can reconstruct the three-dimensional stromal environment and metabolic reprogramming in the TME. A number of studies have demonstrated that optimal induction or inhibition of autophagy may lead to effective therapeutic regimens when combined with immunotherapy. This review discusses the important roles of autophagy in tumor cells, immune cells and stromal cells in the context of tumor immunity, and the potential of combining the autophagy-based therapy with immunotherapy as novel therapeutic approaches against cancer.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xisha Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, Department of Pharmacology, and Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, Department of Pharmacology, and Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
5
|
Immunological ignorance is an enabling feature of the oligo-clonal T cell response to melanoma neoantigens. Proc Natl Acad Sci U S A 2019; 116:23662-23670. [PMID: 31685621 DOI: 10.1073/pnas.1906026116] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The impact of intratumoral heterogeneity (ITH) and the resultant neoantigen landscape on T cell immunity are poorly understood. ITH is a widely recognized feature of solid tumors and poses distinct challenges related to the development of effective therapeutic strategies, including cancer neoantigen vaccines. Here, we performed deep targeted DNA sequencing of multiple metastases from melanoma patients and observed ubiquitous sharing of clonal and subclonal single nucleotide variants (SNVs) encoding putative HLA class I-restricted neoantigen epitopes. However, spontaneous antitumor CD8+ T cell immunity in peripheral blood and tumors was restricted to a few clonal neoantigens featuring an oligo-/monoclonal T cell-receptor (TCR) repertoire. Moreover, in various tumors of the 4 patients examined, no neoantigen-specific TCR clonotypes were identified despite clonal neoantigen expression. Mature dendritic cell (mDC) vaccination with tumor-encoded amino acid-substituted (AAS) peptides revealed diverse neoantigen-specific CD8+ T responses, each composed of multiple TCR clonotypes. Isolation of T cell clones by limiting dilution from tumor-infiltrating lymphocytes (TILs) permitted functional validation regarding neoantigen specificity. Gene transfer of TCRαβ heterodimers specific for clonal neoantigens confirmed correct TCR clonotype assignments based on high-throughput TCRBV CDR3 sequencing. Our findings implicate immunological ignorance of clonal neoantigens as the basis for ineffective T cell immunity to melanoma and support the concept that therapeutic vaccination, as an adjunct to checkpoint inhibitor treatment, is required to increase the breadth and diversity of neoantigen-specific CD8+ T cells.
Collapse
|
6
|
Kim TK, Herbst RS, Chen L. Defining and Understanding Adaptive Resistance in Cancer Immunotherapy. Trends Immunol 2018; 39:624-631. [PMID: 29802087 DOI: 10.1016/j.it.2018.05.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/01/2018] [Accepted: 05/03/2018] [Indexed: 12/31/2022]
Abstract
Despite the unprecedented tumor regression and long-term survival benefit observed with anti-programmed death (PD) [anti-PD-1 or anti-B7-homolog 1 (B7-H1)] therapy in patients with advanced cancers, a large portion of patients do not benefit from such treatment and a fraction of responders relapse. Current efforts to overcome resistance and improve efficacy of anti-PD therapy require a clear understanding of resistance and should precede current avenues using random combinations with available treatment regimens. Here, we categorized three types of resistance, namely target-missing, primary, and acquired resistance. This categorization requires reliable, accurate tissue sampling and appropriate interpretation of results based on the four classifications of tumor immunity in the microenvironment (TIME). We believe that fundamental understanding of these complex tumor-immune interactions and of the cellular and molecular mechanisms underlying these types of true resistance is the key for targeting the right targets in combination with or beyond anti-PD therapy in the future.
Collapse
Affiliation(s)
- Tae Kon Kim
- Section of Hematology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Roy S Herbst
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lieping Chen
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Ding X, Zhang J, Liu D, Xu W, Lu DY, Zhang LP, Su B. Serum expression level of IL-6 at the diagnosis time contributes to the long-term prognosis of SCLC patients. J Cancer 2018; 9:792-796. [PMID: 29581757 PMCID: PMC5868143 DOI: 10.7150/jca.22656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 12/09/2017] [Indexed: 11/05/2022] Open
Abstract
Cytokines are vital mediators involved in tumor immunity. We aimed to explore whether the expression levels of IL-1β, TNF-α and IL-6 have impacts on prognosis of SCLC patients. In this study, we concluded 707 non-operable SCLC patients at stage III or IV into this study and analyzed the relationships between interleukins and OS/PFS by cox regression analysis and Kaplan-Meier analysis (log-rank test). As a result, under current standard chemotherapy, SCLC patients with higher IL-6 expression level had a shortened OS compared with those with normal level (HR: 0.381, 95%CI: 0.177-0.822, p=0.014). Furthermore, IL-6 expression level contributed mostly to patients without a smoking history. Non-smoking patients with a high IL-6 level showed a 6 months shortened OS than those with normal IL-6 level (10.50 vs 16.90 months, p=0.003 by Log-Rank test in Kaplan-Meier analysis). IL-6 had no obvious impacts on first-line PFS in these SCLC patients. To conclude, IL-6 acts as an independent factor of long-term prognosis of SCLC patients under current therapy.
Collapse
Affiliation(s)
- Xi Ding
- Department of General Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University
| | - Jie Zhang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University
| | - Di Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University
| | - Wen Xu
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University
| | - De-Yi Lu
- Department of Bioengineering, University of Illinois at Chicago
| | - Li-Ping Zhang
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University
| | - Bo Su
- Department of Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University
| |
Collapse
|
8
|
CD133, Selectively Targeting the Root of Cancer. Toxins (Basel) 2016; 8:toxins8060165. [PMID: 27240402 PMCID: PMC4926132 DOI: 10.3390/toxins8060165] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 12/20/2022] Open
Abstract
Cancer stem cells (CSC) are capable of promoting tumor initiation and self-renewal, two important hallmarks of carcinoma formation. This population comprises a small percentage of the tumor mass and is highly resistant to chemotherapy, causing the most difficult problem in the field of cancer research, drug refractory relapse. Many CSC markers have been reported. One of the most promising and perhaps least ubiquitous is CD133, a membrane-bound pentaspan glycoprotein that is frequently expressed on CSC. There is evidence that directly targeting CD133 with biological drugs might be the most effective way to eliminate CSC. We have investigated two entirely unrelated, but highly effective approaches for selectively targeting CD133. The first involves using a special anti-CD133 single chain variable fragment (scFv) to deliver a catalytic toxin. The second utilizes this same scFv to deliver components of the immune system. In this review, we discuss the development and current status of these CD133 associated biological agents. Together, they show exceptional promise by specific and efficient CSC elimination.
Collapse
|
9
|
Abstract
Spontaneous remission (SR) of acute myeloid leukemia (AML) is rare. We collected all 46 reported cases of AML with SR. Fever occurred in 91.3% of cases before remission, which was largely due to pneumonia (54.5%) and bacteremia (24.2%). Pneumonia and bacteremia were significantly more common among those who achieved complete remission (CR) compared to those who achieved only a partial remission (p = 0.032). Although 88.6% of remissions were CR, the median duration of remission was only 5 months. Eight cases did not relapse during the follow-up period. The mechanism of SR in AML likely involves the stimulatory effect of systemic febrile infection on the immune system. Immediate treatment of infections and fever may contribute to the rarity of SR in AML. The results of this review improve our understanding of the important role of the immune system in countermanding AML and may provide new ideas for immunotherapy.
Collapse
Affiliation(s)
- Armin Rashidi
- Division of Oncology, Washington University School of Medicine , St. Louis, MO , USA
| | | |
Collapse
|
10
|
Yang Z, Zhao T, Liu Y. Upregulation of tumor suppressor WWOX promotes immune response in glioma. Cell Immunol 2013; 285:1-5. [PMID: 24044959 DOI: 10.1016/j.cellimm.2013.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 06/24/2013] [Accepted: 07/29/2013] [Indexed: 10/26/2022]
Abstract
Previous studies demonstrate that human glioma cells could evade the host's immune surveillance system, result in aggressive proliferation. WWOX, a tumor suppressor gene affected in multiple cancers, induces tumor apoptosis and suppresses growth in vitro and in vivo. However, the effect of WWOX expression in glioma cells to immune cells is still unknown. In the present study, we transduced WWOX into human glioma cell line U251, and cocultured with Jurkat T cells together. We demonstrated that upregulation of WWOX could increase proliferation of Jurkat T cells and decrease the FasL and TGF-β expression of U251 cells, result in inhibiting apoptosis of Jurkat T cells. Therefore, our results suggested that loss of WWOX expression not only resulted in glioma carcinogenesis, but also suppressed immune cell attack by inducing Fas/FasL mediated apoptotic signaling.
Collapse
Affiliation(s)
- Zhao Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | | | | |
Collapse
|
11
|
Wu YL, Liang J, Zhang W, Tanaka Y, Sugiyama H. Immunotherapies: the blockade of inhibitory signals. Int J Biol Sci 2012; 8:1420-30. [PMID: 23197939 PMCID: PMC3509335 DOI: 10.7150/ijbs.5273] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Accepted: 11/06/2012] [Indexed: 12/30/2022] Open
Abstract
T lymphocytes require signaling by the T cell receptor and by nonclonotypic cosignaling receptors. The costimulatory and inhibitory signals profoundly influence the course of immune responses by amplifying or reducing the transcriptional effects of T cell receptor triggering. The inhibitory receptors such as CTLA-4, PD-1, and BTLA have recently drawn much attention as potential targets for immunotherapies. This review focuses on the progress that has been made with the mentioned receptors in the field of immunotherapies for autoimmune diseases, malignancies, infectious diseases, and transplantation.
Collapse
MESH Headings
- Abatacept
- Animals
- Antigens, Differentiation/chemistry
- Antigens, Differentiation/genetics
- Antigens, Differentiation/immunology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/metabolism
- B7-H1 Antigen/physiology
- CD28 Antigens/immunology
- CD28 Antigens/metabolism
- CTLA-4 Antigen/chemistry
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Immunoconjugates/pharmacology
- Immunoconjugates/therapeutic use
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- Immunotherapy/trends
- Mice
- Programmed Cell Death 1 Receptor
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/physiology
- Signal Transduction/drug effects
- Transplantation Immunology
Collapse
Affiliation(s)
- Yan-Ling Wu
- 1. Virus Inspection Department of Zhejiang Provincial Center for Disease Control and Prevention, 630 Xincheng Road, Hangzhou, 310051, China
| | - Jing Liang
- 2. Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China
| | - Wen Zhang
- 2. Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, 310014, China
| | - Yoshimasa Tanaka
- 3. Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Hiroshi Sugiyama
- 4. Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| |
Collapse
|
12
|
Preise D, Scherz A, Salomon Y. Antitumor immunity promoted by vascular occluding therapy: lessons from vascular-targeted photodynamic therapy (VTP). Photochem Photobiol Sci 2011; 10:681-8. [DOI: 10.1039/c0pp00315h] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
13
|
Antitumor efficacy of a photodynamic therapy-generated dendritic cell glioma vaccine. Med Oncol 2010; 28 Suppl 1:S453-61. [PMID: 20960074 DOI: 10.1007/s12032-010-9713-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Accepted: 10/04/2010] [Indexed: 12/15/2022]
Abstract
The objective of this study is to generate dendritic cell (DC) vaccines by exposing DCs to C6 glioma cancer cell antigenic (tumor) peptides following the exposure of C6 cells to photodynamic therapy (PDT) and acid elution. Effects of these DCs on host immunity were assessed by measuring cytokine induction (following adaptive transfer into rats) and assessing DC-induced cytotoxic T lymphocyte (CTL)-mediated lysis of C6 target cells. Precursor dendritic cells were purified from rat bone marrow and matured in vitro. C6 cells were stimulated with PDT, and adherent cells were acid-eluted to obtain cell surface antigens, whole cell antigens were also isolated from supernatants. C6 cells not stimulated with PDT were also used to isolate antigens by acid elution or freeze-thaw methods for comparison purposes. The isolated antigens from the respective purification methods were used to sensitize DCs for the generation of DC vaccines subsequently transferred into SD rats. Following adoptive transfer, the changes in interleukin (IL)-12, IL-10, and TNF-α expression were measured in rat serum by ELISA. CTL-mediated lysis was assessed using the MTT assay. PDT-generated antigens further purified by acid elution had the greatest stimulatory effect on DCs based on the elevated serum IL-12 and TNF-α levels and decreased serum IL-10 levels. CTL activity in this group was also highest (percent lysis 95.5% ± 0.016) compared with that elicited by PDT-supernatants, acid elution, and freeze-thawing (or the control group), which had 90.2% ± 0.024, 73.3% ± 0.027, 63.6% ± 0.049, or 0.4% ± 0.063 lysis, respectively. PDT significantly enhanced tumor cell immunogenicity. These data suggested that DC vaccines prepared by treating tumor cells with PDT to generate antigen-specific CTL responses can be developed as novel cancer immunotherapeutic strategies.
Collapse
|
14
|
Chemical diversity of biologically active metabolites in the sclerotia of Inonotus obliquus and submerged culture strategies for up-regulating their production. Appl Microbiol Biotechnol 2010; 87:1237-54. [DOI: 10.1007/s00253-010-2682-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/11/2010] [Accepted: 05/11/2010] [Indexed: 01/10/2023]
|
15
|
Wang S, Chen L. Immunobiology of cancer therapies targeting CD137 and B7-H1/PD-1 cosignal pathways. Curr Top Microbiol Immunol 2010; 344:245-67. [PMID: 20582531 DOI: 10.1007/82_2010_81] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is finally entering a new era with manipulation of cosignaling pathways as a therapeutic approach, for which the principle was proved nearly two decades ago. In addition to CTLA-4, CD137 and B7-H1/PD-1 pathways are two new targets in the stage. CD137 pathway is costimulatory and its agonistic antibody delivers potent signal to drive T cell growth and activation. On the other hand, blockade of B7-H1/PD-1 pathway with antagonistic antibody has shown to protect ongoing T cell responses from impairment by immune evasion mechanism in cancer microenvironment. With these tools in hand, a mechanism-based design of combined immunotherapy with high efficacy is becoming a reality.
Collapse
Affiliation(s)
- Shengdian Wang
- Center for Infection and Immunity, Institute for Biophysics, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
16
|
Thacker EE, Nakayama M, Smith BF, Bird RC, Muminova Z, Strong TV, Timares L, Korokhov N, O'Neill AM, de Gruijl TD, Glasgow JN, Tani K, Curiel DT. A genetically engineered adenovirus vector targeted to CD40 mediates transduction of canine dendritic cells and promotes antigen-specific immune responses in vivo. Vaccine 2009; 27:7116-24. [PMID: 19786146 DOI: 10.1016/j.vaccine.2009.09.055] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 09/02/2009] [Accepted: 09/16/2009] [Indexed: 01/06/2023]
Abstract
Targeting viral vectors encoding tumor-associated antigens to dendritic cells (DCs) in vivo is likely to enhance the effectiveness of immunotherapeutic cancer vaccines. We have previously shown that genetic modification of adenovirus (Ad) 5 to incorporate CD40 ligand (CD40L) rather than native fiber allows selective transduction and activation of DCs in vitro. Here, we examine the capacity of this targeted vector to induce immune responses to the tumor antigen CEA in a stringent in vivo canine model. CD40-targeted Ad5 transduced canine DCs via the CD40-CD40L pathway in vitro, and following vaccination of healthy dogs, CD40-targeted Ad5 induced strong anti-CEA cellular and humoral responses. These data validate the canine model for future translational studies and suggest targeting of Ad5 vectors to CD40 for in vivo delivery of tumor antigens to DCs is a feasible approach for successful cancer therapy.
Collapse
Affiliation(s)
- Erin E Thacker
- Division of Human Gene Therapy, Department of Medicine, Birmingham, AL 35294, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Gibbons C, Sykes M. Manipulating the immune system for anti-tumor responses and transplant tolerance via mixed hematopoietic chimerism. Immunol Rev 2008; 223:334-60. [PMID: 18613846 PMCID: PMC2680695 DOI: 10.1111/j.1600-065x.2008.00636.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SUMMARY Stem cells (SCs) with varying potentiality have the capacity to repair injured tissues. While promising animal data have been obtained, allogeneic SCs and their progeny are subject to immune-mediated rejection. Here, we review the potential of hematopoietic stem cells (HSCs) to promote immune tolerance to allogeneic and xenogeneic organs and tissues, to reverse autoimmunity, and to be used optimally to cure hematologic malignancies. We also review the mechanisms by which hematopoietic cell transplantation (HCT) can promote anti-tumor responses and establish donor-specific transplantation tolerance. We discuss the barriers to clinical translation of animal studies and describe some recent studies indicating how they can be overcome. The recent achievements of durable mixed chimerism across human leukocyte antigen barriers without graft-versus-host disease and of organ allograft tolerance through combined kidney and bone marrow transplantation suggest that the potential of this approach for use in the treatment of many human diseases may ultimately be realized.
Collapse
Affiliation(s)
- Carrie Gibbons
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | |
Collapse
|
18
|
Ruybal P, Gravisaco MJ, Barcala V, Escalada A, Di Sciullo P, Waldner C, Mongini C. Complete rejection of a T-cell lymphoma due to synergism of T-cell receptor costimulatory molecules, CD80, CD40L, and CD40. Vaccine 2007; 26:697-705. [PMID: 18155328 DOI: 10.1016/j.vaccine.2007.11.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 10/20/2007] [Accepted: 11/16/2007] [Indexed: 11/17/2022]
Abstract
The equal importance of the qualitative and quantitative characteristics of antigen presentation as well as the set of costimulatory signals provided by antigen presenting cells to T-cells in determining the outcome of T-cell responses at the time of antigen recognition is now clear. Moreover, an important function in innate mechanisms has been recently attributed to costimulatory molecules demonstrating their relevant role in different stages of immune response. In this paper, we demonstrated the ability of CD40L (CD154) and CD80 costimulatory molecules expression in a T-cell lymphoma to induce both T-cell dependent and independent immune responses leading to an important anti-tumor effect. CD40 expression by LBC cells enhanced only T-cell dependent anti-tumor immune response resulting in tumor rejection. Furthermore, this work represents the first report to describe complete tumor rejection after co-inoculation of lymphoma cells transfected with CD40L and CD80 in either presence or absence of CD40 expressing lymphoma cells. In addition, this synergistic effect resulted in long lasting immunity to parental tumor cells. Co-inoculation of tumor cells each genetically modified to express a different costimulatory molecule circumvents the need to co-transfect genetically unstable tumor cells and represents an option for those weakly or non-immunogenic tumors where either treatment alone proved to be inefficient. This strategy represents a promising approach for inducing anti-tumor immunity and provides a new rational design of cancer therapies.
Collapse
Affiliation(s)
- Paula Ruybal
- Laboratorio de Inmunología Celular y Molecular, Centro de Estudios Farmacológicos y Botánicos, CEFyBO-CONICET, Universidad de Buenos Aires, Facultad de Medicina, Paraguay 2155, 1121 Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
19
|
Mazzolini G, Murillo O, Atorrasagasti C, Dubrot J, Tirapu I, Rizzo M, Arina A, Alfaro C, Azpilicueta A, Berasain C, Perez-Gracia JL, Gonzalez A, Melero I. Immunotherapy and immunoescape in colorectal cancer. World J Gastroenterol 2007; 13:5822-31. [PMID: 17990348 PMCID: PMC4205429 DOI: 10.3748/wjg.v13.i44.5822] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy encompasses a variety of interventions and techniques with the common goal of eliciting tumor cell destructive immune responses. Colorectal carcinoma often presents as metastatic disease that impedes curative surgery. Novel strategies such as active immunization with dendritic cells (DCs), gene transfer of cytokines into tumor cells or administration of immunostimulatory monoclonal antibodies (such as anti-CD137 or anti-CTLA-4) have been assessed in preclinical studies and are at an early clinical development stage. Importantly, there is accumulating evidence that chemotherapy and immunotherapy can be combined in the treatment of some cases with colorectal cancer, with synergistic potentiation as a result of antigens cross-presented by dendritic cells and/or elimination of competitor or suppressive T lymphocyte populations (regulatory T-cells). However, genetic and epigenetic unstable carcinoma cells frequently evolve mechanisms of immunoevasion that are the result of either loss of antigen presentation, or an active expression of immunosuppressive substances. Some of these actively immunosuppressive mechanisms are inducible by cytokines that signify the arrival of an effector immune response. For example, induction of 2, 3 indoleamine dioxygenase (IDO) by IFNγ in colorectal carcinoma cells. Combinational and balanced strategies fostering antigen presentation, T-cell costimulation and interference with immune regulatory mechanisms will probably take the stage in translational research in the treatment of colorectal carcinoma.
Collapse
|
20
|
B cells in glomerulonephritis: focus on lupus nephritis. Semin Immunopathol 2007; 29:337-53. [PMID: 17943287 DOI: 10.1007/s00281-007-0092-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Accepted: 09/28/2007] [Indexed: 01/10/2023]
Abstract
The production of pathogenic antibody has been traditionally viewed as the principle contribution of B cells to the pathogenesis of immune-mediated glomerulonephritis. However, it is increasingly appreciated that B cells play a much broader role in such diseases, functioning as antigen-presenting cells, regulators of T cells, dendritic cells, and macrophages and orchestrators of local lymphatic expansion. In this review, we provide an overview of basic B cell biology and consider the evidence implicating B cells in one of the archetypal immune-mediated glomerulonephritides, lupus nephritis.
Collapse
|
21
|
Chu Y, Yang X, Xu W, Wang Y, Guo Q, Xiong S. In situ expression of IFN-gamma-inducible T cell alpha chemoattractant in breast cancer mounts an enhanced specific anti-tumor immunity which leads to tumor regression. Cancer Immunol Immunother 2007; 56:1539-49. [PMID: 17659370 PMCID: PMC11030116 DOI: 10.1007/s00262-007-0296-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 01/23/2007] [Indexed: 10/23/2022]
Abstract
Increased evidence indicates that chemokines are involved in tumor growth. ITAC, a key member of chemokines, possesses the ability to recruit T cells and enhance immune responses. Therefore, ITAC might contribute to antitumor immunity. In this study, we evaluated the relationship between the expression of ITAC and human breast cancer advancement. We further investigated whether forced expression of ITAC in tumor sites could mediate enhanced antitumor immunity in a murine breast cancer model. Results showed that ITAC expression level was down-regulated in 31 breast cancer specimens compared to normal mammary tissues, and associated negatively with the stages of breast cancer. Contrarily, forced expression of ITAC in murine 4T1 tumor cells resulted in tumor regression after initial growth upon injection into naïve Balb/c mice. More lymphocytes were recruited to the site of tumor inoculated by 4T1-ITAC and more than 80% of these T cells expressed the ITAC receptor, CXCR3. ITAC-recruited TILs exhibited 4T1-specific proliferation and cytotoxicity, and an increased IFN-gamma but decreased IL-4 production. Importantly, forced expression of ITAC in 4T1 tumor nodules inhibited tumor growth. These findings demonstrated that the decreased expression of ITAC is associated with the advancement of breast cancer in patients. Forced expression of ITAC in tumor site not only induces increased T cell-recruitment and elicits a specific antitumor immunity, but also mediates regression of established 4T1 tumors, indicating the potential application of ITAC-expressing tumor cells in cancer immunotherapy and vaccine designing.
Collapse
Affiliation(s)
- Yiwei Chu
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Xiuli Yang
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Wei Xu
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Ying Wang
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Qiang Guo
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
| | - Sidong Xiong
- Department of Immunology of Shanghai Medical College and Institute for Immunobiology, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 Peoples Republic of China
- Key Laboratory of Molecular Medicine of Ministry of Education, Fudan University, Shanghai, 200032 Peoples Republic of China
- Immunology Division, E-Institutes of Shanghai Universities, Shanghai, 200025 Peoples Republic of China
| |
Collapse
|
22
|
Altin JG, Parish CR. Liposomal vaccines--targeting the delivery of antigen. Methods 2007; 40:39-52. [PMID: 16997712 DOI: 10.1016/j.ymeth.2006.05.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 05/05/2006] [Indexed: 01/05/2023] Open
Abstract
Vaccines that can prime the adaptive immune system for a quick and effective response against a pathogen or tumor cells, require the generation of antigen (Ag)-specific memory T and B cells. The unique ability of dendritic cells (DCs) to activate naïve T cells, implies a key role for DCs in this process. The generation of tumor-specific CD8(+) cytotoxic T cells (CTLs) is dependent on both T cell stimulation with Ag (peptide-MHC-complexes) and costimulation. Interestingly, tumor cells that lack expression of T cell costimulatory molecules become highly immunogenic when transfected to express such molecules on their surface. Adoptive immunotherapy with Ag-pulsed DCs also is a strategy showing promise as a treatment for cancer. The use of such cell-based vaccines, however, is cumbersome and expensive to use clinically, and/or may carry risks due to genetic manipulations. Liposomes are particulate vesicular lipid structures that can incorporate Ag, immunomodulatory factors and targeting molecules, and hence can serve as potent vaccines. Similarly, Ag-containing plasma membrane vesicles (PMV) derived from tumor cells can be modified to incorporate a T cell costimulatory molecule to provide both TCR stimulation, and costimulation. PMVs also can be modified to contain IFN-gamma and molecules for targeting DCs, permitting delivery of both Ag and a DC maturation signal for initiating an effective immune response. Our results show that use of such agents as vaccines can induce potent anti-tumor immune responses and immunotherapeutic effects in tumor models, and provide a strategy for the development of effective vaccines and immunotherapies for cancer and infectious diseases.
Collapse
Affiliation(s)
- Joseph G Altin
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia.
| | | |
Collapse
|
23
|
Abstract
Human telomerase reverse transcriptase (hTERT) represents a universal tumor-associated antigen to activate specific immune response in cancer immune therapy. Peptides derived from hTERT are presented by major histocompatibility complex (MHC) class I alleles to T lymphocytes, and CD8+ cytotoxic T lymphocytes (CTLs) specific for the hTERT-derived antigenic epitopes lyse hTERT-positive tumors from multiple histologies. These findings identify hTERT as an important tumor antigen widely applicable for anti-cancer immunotherapeutic strategies. The hTERT antigen-specific immunotherapy involves both active vaccination and adoptive immunotherapy approaches. Most importantly, the anti-tumor immune responses have been observed in the absence of toxicity, underlying the ongoing endeavors to develop immunotherapy directed against hTERT antigen. This chapter discusses most promising results and the approaches for investigation to target hTERT peptides as tumor antigens.
Collapse
Affiliation(s)
- He Li
- Centre for Functional Genomics and Human Disease, Monash University, Melbourne, Australia
| | | | | |
Collapse
|
24
|
Murakami M, Ito H, Harada E, Enoki T, Sykes M, Hamano K. Long-term survival of xenogeneic heart grafts achieved by costimulatory blockade and transient mixed chimerism. Transplantation 2006; 82:275-81. [PMID: 16858292 DOI: 10.1097/01.tp.0000226221.53161.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Xenotransplantation holds great promise in clinical medicine, but is limited by the vigorous rejection response elicited against solid organs transplanted across species barriers. In this study, we investigated the role of anti-CD40L monoclonal antibody (mAb) in inducing xenogeneic mixed chimerism and donor-specific heart transplantation tolerance. METHODS One day before heart transplantation, mice were injected intraperitoneally with anti-mouse CD8/NK1.1/Thy1.2 mAbs. On day 0, the mice received 3 Gy total body irradiation (TBI), an intravenous injection of unseparated bone marrow (BM) harvested from F344 rats, and an intraperitoneal injection of hamster antimouse CD40L mAb, MR1. Heart grafts from F344 rats were heterotopically transplanted into the abdomen of B6 mouse recipients. Using flow cytometric analysis of peripheral white blood cells, we assessed donor hematopoiesis at various times after bone marrow transplantation (BMT). RESULTS Chimerism subsided gradually and disappeared completely 18 weeks after BMT. The cardiac graft survived permanently, even after the mixed chimerism disappeared. To determine if the mice acquired donor-specific tolerance, second rat heart grafts were transplanted 120 days after the first heart transplantation. The second transplanted hearts were also accepted over 60 days. Histological analysis revealed no remarkable vasculopathy in the coronary vessels at any stage. CONCLUSIONS These findings clearly show that costimulatory blockade plays an important role in inducing xenochimerism, and that transient mixed chimerism can induce permanent acceptance of rat to mouse cardiac xenografts. Transplantation of xenogeneic bone marrow cells under costimulatory blockade at the time of heart transplantation may induce transplantation tolerance.
Collapse
Affiliation(s)
- Masanori Murakami
- Department of Surgery and Clinical Science, Yamaguchi University, Graduate School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Zheng W, Gao X, Chen C, Tan R. Total flavonoids of Daphne genkwa root significantly inhibit the growth and metastasis of Lewis lung carcinoma in C57BL6 mice. Int Immunopharmacol 2006; 7:117-27. [PMID: 17178377 DOI: 10.1016/j.intimp.2006.07.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2006] [Revised: 06/19/2006] [Accepted: 07/09/2006] [Indexed: 11/26/2022]
Abstract
Daphne genkwa root has been traditionally used as an effective remedy to treat various tumors. However, the active constituents for its antitumor potency have not been well documented. During the screening for antitumor constituents, it was found that the total flavonoids of D. genkwa root (TFDR) were responsible for the inhibition of tumor growth and metastasis. In this study, TFDR was investigated for its chemical composition and activities against tumor growth and metastasis. HPLC indicates that daphnodorin B, containing 42.79% of the total, represents the predominant constituent in TFDR. Treatment of LLC-bearing mice with TFDR evidently protected peripheral lymphocytes from tumor-induced reduction, and increased lymphocyte proliferation potential and cytolytic activity of NK, and inhibited tumor progression and metastasis either 7 days before, or simultaneous with, or 7 days after LLC transplantation. TFDR also suggested higher cytotoxicity to a number of tumor cell lines than that to normal human kidney cell K293. TFDR also induced an enhancement on peripheral release of TNF-alpha at doses between 25 and 75 mg/kg. These results indicated that TFDR inhibited tumor growth and metastasis by protecting host immunocyte viability and its proliferation potential, and selectively inhibiting tumor cell proliferation, and improving cytolytic activity of NK cells, and enhancing TNF release in LLC-bearing mice. Daphnodorin B and its analogues in TFDR are the active constituents in the roots of D. genkwa, contributing to the inhibition of tumor growth and metastasis.
Collapse
Affiliation(s)
- Weifa Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, Xuzhou Normal University, Xuzhou, China.
| | | | | | | |
Collapse
|
26
|
Schillaci R, Salatino M, Cassataro J, Proietti CJ, Giambartolomei GH, Rivas MA, Carnevale RP, Charreau EH, Elizalde PV. Immunization with murine breast cancer cells treated with antisense oligodeoxynucleotides to type I insulin-like growth factor receptor induced an antitumoral effect mediated by a CD8+ response involving Fas/Fas ligand cytotoxic pathway. THE JOURNAL OF IMMUNOLOGY 2006; 176:3426-37. [PMID: 16517711 DOI: 10.4049/jimmunol.176.6.3426] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have demonstrated that in vivo administration of phosphorothioate antisense oligodeoxynucleotides (AS[S]ODNs) to type I insulin-like growth factor receptor (IGF-IR) mRNA resulted in inhibition of C4HD breast cancer growth in BALB/c mice. The present study focused on whether in vivo administration of C4HD tumor cells pretreated with IGF-IR AS[S]ODN and irradiated could provide protection against C4HD wild-type tumor challenge and also on elucidating the mechanism mediating this effect. Our results showed that mice immunized with IGF-IR AS[S]ODN-treated C4HD cells experienced a growth inhibition of 53.4%, 61.6%, and 60.2% when compared with PBS-treated mice, wild-type C4HD cell-injected mice, or phosphorothioate sense oligodeoxynucleotide-treated C4HD cell-injected mice, respectively. The protective effect was C4HD-specific, because no cross-protection was observed against other syngeneic mammary tumor lines. The lack of protection against tumor formation in nude mice indicated that T cells were involved in the antitumoral response. Furthermore, cytotoxicity and splenocyte proliferation assays demonstrated that a cellular CD8(+)-dependent immune response, acting through the Fas/Fas ligand death pathway, could be mediating the antitumor effect induced by immunization with AS[S]ODN-treated cells. Immunization also induced splenocytes to produce Ag-dependent IFN-gamma, indicating the presence of a type 1 response. We demonstrated for the first time that IGF-IR AS[S]ODN treatment of breast cancer cells induced expression of CD86 and heat shock protein 70 molecules, both involved in the induction of the immunogenic phenotype. Immunization with these tumor immunogens imparted protection against parental tumor growth through activation of a specific immune response.
Collapse
Affiliation(s)
- Roxana Schillaci
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
It is not unusual for antigens and potentially responsive T cells to co-exist in the same organism while these T cells remain silent and do not mount life-threatening immune responses. A rich array of mechanisms has been proposed to explain these observations. T cell silencing is controlled in multiple levels. Initially, dendritic cells and regulatory T cells appear to play critical roles. In addition, T cell immunity is tightly regulated by a molecular network of cytokines and cell receptor interactions by the opposed surfaces of antigen-presenting cells and T cells. Recognition of a specific antigen is therefore shaped and tuned by co-stimulatory and co-inhibitory receptor-ligand pairs. At last, immunologists are beginning to exploit the rules governing these assorted sounds of T cell silence.
Collapse
Affiliation(s)
- Ignacio Melero
- Department of Medicine, CIMA and Clinica Universitaria, University of Navarra, Pamplona, Spain.
| | | | | |
Collapse
|
28
|
Lu J, Zhao J, Song Q, Cui ZY, Zhao GQ, Yang HY, Huang YT, Zhao JM, Dong ZM. Combination effect of human B7-2 and dentric cells vaccines in anti-esophageal cancer in vitro. Shijie Huaren Xiaohua Zazhi 2005; 13:1382-1385. [DOI: 10.11569/wcjd.v13.i12.1382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate the roles of human B7-2 combined with dentric cell (DC) vaccine in inducing anti-tumor immunity against esophageal cancer in vitro.
METHODS: Human esophageal cancer cell line EC9706 was transfected with the vector of pEGFP-N3-B7-2 by lipofectamine method. The mononuclear cells (MNCs) were separated from cord blood by density gradient centrifugation (Ficall-Hypaque) and then were induced to differentiate by cell factors. On the 3rd day, EC9706 cytolysis antigen was added into the medium, in which DCs gradually matured and expressed special tumor antigen. After the matured DCs were co-cultured with autologous T cells derived from cord blood for 3 days, the T cells were activated to become tumor specific cytotoxic T lymphocytes (CTL). The inhibition of CTL on the transfected and untransfected EC9706 cells was detected by methyl thiazolyl tetrazolium (MTT) assay.
RESULTS: The EGFP-B7-2 fusion gene was expressed mainly on the membrane of EC9706 cells, which proved the transfection was successful. Mature DCs with tumor cytolysis antigene was able to activate naive T cells to become tumor specialized CTL, and the CTL had significant inhibitory effect or killing response on EC9706 cells transfected with pEGFP-N3-B7-2(F = 21.672,P = 0.000).
CONCLUSION: The human B7-2 combined with DC vaccine can induce significant killing response on esophageal cancer cell.
Collapse
|
29
|
Vonderheide RH, Domchek SM, Schultze JL, George DJ, Hoar KM, Chen DY, Stephans KF, Masutomi K, Loda M, Xia Z, Anderson KS, Hahn WC, Nadler LM. Vaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytes. Clin Cancer Res 2004; 10:828-39. [PMID: 14871958 DOI: 10.1158/1078-0432.ccr-0620-3] [Citation(s) in RCA: 191] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE High-level expression of the telomerase reverse transcriptase (hTERT) in >85% of human cancers, in contrast with its restricted expression in normal adult tissues, points to hTERT as a broadly applicable molecular target for anticancer immunotherapy. CTLs recognize peptides derived from hTERT and kill hTERT+ tumor cells of multiple histologies in vitro. Moreover, because survival of hTERT+ tumor cells requires functionally active telomerase, hTERT mutation or loss as a means of escape may be incompatible with sustained tumor growth. EXPERIMENTAL DESIGN A Phase I clinical trial was performed to evaluate the clinical and immunological impact of vaccinating advanced cancer patients with the HLA-A2-restricted hTERT I540 peptide presented with keyhole limpet hemocyanin by ex vivo generated autologous dendritic cells. RESULTS As measured by peptide/MHC tetramer, enzyme-linked immunospot, and cytotoxicity assays, hTERT-specific T lymphocytes were induced in 4 of 7 patients with advanced breast or prostate carcinoma after vaccination with dendritic cells pulsed with hTERT peptide. Tetramer-guided high-speed sorting and polyclonal expansion achieved highly enriched populations of hTERT-specific cells that killed tumor cells in an MHC- restricted fashion. Despite concerns of telomerase activity in rare normal cells, no significant toxicity was observed. Partial tumor regression in 1 patient was associated with the induction of CD8+ tumor infiltrating lymphocytes. CONCLUSIONS These results demonstrate the immunological feasibility of vaccinating patients against telomerase and provide rationale for targeting self-antigens with critical roles in oncogenesis.
Collapse
Affiliation(s)
- Robert H Vonderheide
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2004; 12:1976-1978. [DOI: 10.11569/wcjd.v12.i8.1976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
|
31
|
Bhattacharyya A, Mandal D, Lahiry L, Sa G, Das T. Black tea protects immunocytes from tumor-induced apoptosis by changing Bcl-2/Bax ratio. Cancer Lett 2004; 209:147-54. [PMID: 15159016 DOI: 10.1016/j.canlet.2003.12.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2003] [Revised: 12/29/2003] [Accepted: 12/30/2003] [Indexed: 10/26/2022]
Abstract
It is known that cancer is associated with altered immune function. We demonstrated earlier that black tea inhibits tumor growth in a dose-dependent manner. Here, we report that apoptosis was the cause of immunocyte death in Ehrlich's ascites carcinoma (EAC)-bearing mice and anti-tumor dose of black tea restored EAC-induced immunosuppression by inhibiting apoptosis. A search for the molecular mechanism revealed that EAC burden increased the expression of the pro-apoptotic proteins p53 and Bax in splenic lymphocytes although did not change the level of pro-proliferative protein Bcl-2. Interestingly, anti-tumor dose of black tea down-regulated p53, decreased Bax while augmenting Bcl-2 in these cells. As a result, Bcl-2/Bax ratio was increased and the immunocytes were protected from tumor-induced apoptosis. Thus, unlike many other anti-cancer agents, black tea is not only devoid of immunosuppressive effect but also acts as immuno-restorer in tumor-bearing host. These results, thus, raise the possibility of inclusion of black tea in successful therapeutic regimen against cancer.
Collapse
|
32
|
Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol 2004; 4:336-47. [PMID: 15122199 DOI: 10.1038/nri1349] [Citation(s) in RCA: 986] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Lieping Chen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.
| |
Collapse
|
33
|
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) is an important source of cancer-related morbidity and mortality worldwide. Current treatment options are founded on various combinations of surgery, radiation therapy, and chemotherapy, with implementation largely based on patient/disease-specific factors and provider/institutional bias. Despite improvements in surgical techniques and the development of novel chemoradiotherapy strategies for organ preservation, survival has remained relatively unchanged in the past 30 years. Additionally, the impact of these novel treatment approaches on the quality of life of patients remains largely unknown. Improvements in survival and the quality of life for patients with SCCHN will likely require the development of new therapeutic alternatives based on the improved understanding of the molecular pathogenesis of SCCHN and associated changes in the immune response.
Collapse
Affiliation(s)
- Scott E Strome
- Department of Otolaryngology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | |
Collapse
|
34
|
Yu P, Lee Y, Liu W, Chin RK, Wang J, Wang Y, Schietinger A, Philip M, Schreiber H, Fu YX. Priming of naive T cells inside tumors leads to eradication of established tumors. Nat Immunol 2004; 5:141-9. [PMID: 14704792 DOI: 10.1038/ni1029] [Citation(s) in RCA: 289] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2003] [Accepted: 11/13/2003] [Indexed: 12/22/2022]
Abstract
The tumor barrier comprised of nonantigenic stromal cells may contribute to the failure of tumor rejection. The tumor-necrosis factor superfamily member LIGHT (also known as TNFSF-14) is a ligand of stromal cell-expressed lymphotoxin-beta receptor and T cell-expressed herpes viral entry mediator (HVEM). Here we show that forced expression of LIGHT in the tumor environment induces a massive infiltration of naive T lymphocytes that correlates with an upregulation of both chemokine production and expression of adhesion molecules. Activation of these infiltrating T cells, possibly through HVEM, leads to the rejection of established, highly progressive tumors at local and distal sites. Our study indicates that targeting the tumor barrier may be an effective strategy for cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Cell Adhesion Molecules/biosynthesis
- Chemokines/biosynthesis
- Female
- Gene Expression
- Graft Rejection
- Immunotherapy/methods
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Models, Immunological
- Neoplasm Transplantation
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor, Member 14
- Receptors, Virus/immunology
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/therapy
- T-Lymphocytes/immunology
- Transduction, Genetic
- Tumor Necrosis Factor Ligand Superfamily Member 14
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Vaccination
Collapse
Affiliation(s)
- Ping Yu
- The Committee on Immunology and Department of Pathology, University of Chicago, MC3083, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sinibaldi Vallebona P, Rasi G, Pierimarchi P, Bernard P, Guarino E, Guadagni F, Garaci E. Vaccination with a synthetic nonapeptide expressed in human tumors prevents colorectal cancer liver metastases in syngeneic rats. Int J Cancer 2004; 110:70-5. [PMID: 15054870 DOI: 10.1002/ijc.20063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In previous studies, the antigen CSH-275 (RTNKEASIC) was found expressed in tissue specimens from colorectal cancer but not in normal colonic mucosa. It was also naturally expressed in the DHD-K12 experimental colorectal cancer in BDIX rats. In this study, we describe the effect of vaccination with the synthetic nonapeptide CSH-275 in preventing tumor growth in a model closely mimicking the clinical situation of liver metastases, after surgical resection of primary colorectal cancer. A vaccination protocol using CSH-275, conjugated with complete or incomplete Freund's adjuvant, was carried out to determine the effect in preventing the progression of liver metastases induced by DHD-K12 cells injected in the splenic vein (preventive vaccine). An additional vaccination procedure was carried out to determine the effect on s.c. tumor growth (therapeutic vaccine). A significant improvement in survival along with the prevention of liver metastases formation and reduced growth of s.c. tumor were observed. CSH-275 vaccination resulted in a significant increase in CTL activity against autologous DHD-K12 cells in DHD-K12 tumor-bearing rats and the generation of a CTL response against DHD-K12 cells in DHD-K12 naive rats. Vaccination also induced massive infiltration of CD8(+) cells in tumor. These results demonstrate that CSH-275 is a new molecular target for colorectal cancer immunotherapy; it is also an excellent candidate for preclinical studies because it is naturally expressed on tumors in a fully competent syngeneic animal, which reproduces the clinical pattern of cancer progression.
Collapse
Affiliation(s)
- Paola Sinibaldi Vallebona
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Chang CJ, Liao CH, Wang FH, Lin CM. Transforming growth factor-beta induces apoptosis in antigen-specific CD4+ T cells prepared for adoptive immunotherapy. Immunol Lett 2003; 86:37-43. [PMID: 12600743 DOI: 10.1016/s0165-2478(02)00307-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transforming growth factor-beta (TGF-beta), found at the site of most tumors, has been recognized as one of the mechanisms involved in tumor immunological escape. To evaluate its impact on adoptive immunotherapy against cancer, we examined the susceptibility of tumor-specific T cells to TGF-beta in the setting of these T cells being prepared for adoptive transfer. Hepatitis B virus (HBV)-specific CD4(+) T cells were ex vivo generated by activating with HBV-transfected dendritic cells and selecting with antibodies to CD25 activation molecules, and then expanded with antibodies to CD3/CD28. These T cells expressed higher levels of the type II TGF-beta receptor than nai;ve T cells and exhibited enhanced apoptosis when exposed to TGF-beta. The underlying apoptotic pathway was linked to the dissipation of the mitochondrial inner membrane potential and activation of caspase-9. The absence of caspase-8 activity in TGF-beta-treated T cells suggests that the death receptor system may not be involved in this type of apoptosis. Interleukin-2 (IL-2), which is concomitantly administered with tumor-specific T cells in adoptive immunotherapy, was unable to protect HBV-specific CD4(+) T cells from the pro-apoptotic effect of TGF-beta when added simultaneously with TGF-beta. Interesting, IL-2-pretreated T cells displayed the type II TGF-beta receptor at lower levels and were more resistant to TGF-beta. Together, our findings indicate that the effectiveness of adoptive cancer immunotherapy may be impaired by tumor-derived TGF-beta and appropriate manipulation of exogenous IL-2 might overcome this hurdle.
Collapse
Affiliation(s)
- Chun-Jung Chang
- Department of Microbiology, Soochow University Taipei, Wai Shuang Hsi, Shih Lin, Taipei 11102, Taiwan, ROC
| | | | | | | |
Collapse
|
37
|
Ochsenbein AF. Principles of tumor immunosurveillance and implications for immunotherapy. Cancer Gene Ther 2002; 9:1043-55. [PMID: 12522443 DOI: 10.1038/sj.cgt.7700540] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2002] [Indexed: 01/24/2023]
Abstract
Although antigen loss variants, major histocompatibility (MHC) class I down-regulation, or the expression of inhibitory molecules may explain the failure of immunosurveillance against some tumors, this seems not to apply for many other solid peripheral or lymphohematopoietic tumors. Why then is immunosurveillance so ineffective and can it be improved? This review focuses on one important aspect of tumor immunity, namely the relevance of antigen dose and localization. Immune responses in vivo are induced in organized lymphoid tissues, i.e., in lymph nodes and spleen. The antigen dose that reaches secondary lymphoid organs over time is a crucial parameter that drives antiviral and antitumoral immune responses. Tumors use various strategies to prevent efficient presentation of their antigens in lymphoid organs. A major obstacle to the induction of an endogenous tumor-specific cytotoxic T lymphocyte (CTL) response is the inefficient presentation of tumor antigen on MHC class I molecules of professional antigen-presenting cells. Peripheral solid tumors that develop outside lymphoid organs are, therefore, often ignored by the immune system. In other situations, tumors - especially of lymphohematopoietic origin - may tolerize specific CTLs. Understanding tumor immunosurveillance is key to the design of efficient antitumor vaccines. Attempts to improve immunity to tumors include vaccination strategies to (a) provide the tumor antigen to secondary lymphoid organs using recombinant viruses or dendritic cells as carriers, (b) express costimulatory signals on tumor cells, or (c) improve the efficiency of cross-priming.
Collapse
|
38
|
Leggatt GR, Dunn LA, De Kluyver RL, Stewart T, Frazer IH. Interferon-gamma enhances cytotoxic T lymphocyte recognition of endogenous peptide in keratinocytes without lowering the requirement for surface peptide. Immunol Cell Biol 2002; 80:415-24. [PMID: 12225377 DOI: 10.1046/j.1440-1711.2002.01105.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Keratinocytes expressing the human papillomavirus (HPV) type 16 E7 protein, as a transgene driven by the K14 promoter, form a murine model of HPV-mediated epithelial cancers in humans. Our previous studies have shown that K14E7 transgenic skin grafts onto syngeneic mice are not susceptible to immune destruction despite the demonstrated presence of a strong, systemic CTL response directed against the E7 protein. Consistent with this finding, we now show that cultured, E7 transgenic keratinocytes (KC) express comparable endogenous levels of E7 protein to a range of CTL-sensitive E7-expressing cell lines but are not susceptible to CTL-mediated lysis in vitro. E7 transgenic and non-transgenic KC are susceptible to conventional mechanisms of CTL-mediated lysis, including perforin and Fas/FasL interaction when an excess of exogenous peptide is provided. The concentration of exogenous peptide required to render a cell susceptible to lysis was similar between KC and other conventional CTL targets (e.g. EL-4), despite large differences in H-2Db expression at the cell surface. Furthermore, exposure of KC to IFN-gamma increased H-2Db expression, but did not substantially alter the exogenous peptide concentration required to sensitize cells for half maximal lysis. In contrast, the lytic sensitivity of transgenic KC expressing endogenous E7 is modestly improved by exposure to IFN-gamma. Thus, failure of CTL to eliminate KC expressing endogenous E7, and by inference squamous tumours expressing E7, may reflect the need for a sustained, local inflammatory environment during the immune effector phase.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/virology
- Cells, Cultured/drug effects
- Cells, Cultured/immunology
- Cytotoxicity, Immunologic
- Disease Models, Animal
- Fas Ligand Protein
- H-2 Antigens/immunology
- Histocompatibility Antigen H-2D
- Interferon-gamma/pharmacology
- Keratinocytes/drug effects
- Keratinocytes/immunology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Papillomavirus E7 Proteins
- Peptide Fragments/immunology
- Perforin
- Pore Forming Cytotoxic Proteins
- Skin Neoplasms/immunology
- Skin Neoplasms/virology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- fas Receptor/physiology
Collapse
Affiliation(s)
- Graham R Leggatt
- Centre for Immunology and Cancer Research, University of Queensland, Princess Alexandra Hospital, Brisbane, Australia.
| | | | | | | | | |
Collapse
|
39
|
Knight SC, Burke F, Bedford PA. Dendritic cells, antigen distribution and the initiation of primary immune responses to self and non-self antigens. Semin Cancer Biol 2002; 12:301-8. [PMID: 12147204 DOI: 10.1016/s1044-579x(02)00016-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunity or tolerance are determined through the bone marrow-derived, antigen-presenting cells, dendritic cells (DC). Stimulation of lymphocytes by different types of DC, DC at different stages of maturity and DC producing and responding to different growth factors modulate immune responses. Innate receptors for foreign or self antigens provide scope in DC for discrimination between different antigenic stimuli. DC also transfer processed antigens to other DC. We propose that DC do not stimulate responses to antigens in their own environment but only to antigens acquired from other DC, providing a mechanism for discriminating between environmental and non-environmental antigens.
Collapse
Affiliation(s)
- Stella C Knight
- Antigen Presentation Research Group, Northwick Park Institute for Medical Research, Imperial College Faculty of Medicine, Watford Rd., Harrow HA1 3UJ, UK.
| | | | | |
Collapse
|
40
|
Chapoval AI, Zhu G, Chen L. Immunoglobulin fusion proteins as a tool for evaluation of T-cell costimulatory molecules. Mol Biotechnol 2002; 21:259-64. [PMID: 12102550 DOI: 10.1385/mb:21:3:259] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Costimulatory signal(s) in addition to engagement of T cell receptor is important for optimal activation of T lymphocytes. During last decade several dozens of new costimulatory molecules, including 4-1BB, CD40, OX40, CD27, and ICOS, have been identified. It is likely that in Post-Genome Era more molecules with potential costimulatory properties will be brought to our attention. We describe here a simple and reliable strategy for evaluating the functional activities of potential costimulatory molecules using soluble fusion protein between extracellular portion of costimulatory ligand and Fc portion of mouse IgG2a.
Collapse
|
41
|
Igney FH, Krammer PH. Immune escape of tumors: apoptosis resistance and tumor counterattack. J Leukoc Biol 2002. [DOI: 10.1189/jlb.71.6.907] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Frederik H. Igney
- Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter H. Krammer
- Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
42
|
Wilcox RA, Flies DB, Zhu G, Johnson AJ, Tamada K, Chapoval AI, Strome SE, Pease LR, Chen L. Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. J Clin Invest 2002. [PMID: 11877473 DOI: 10.1172/jci0214184] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Treatment of advanced, poorly immunogenic tumors in animal models, considered the closest simulation available thus far for conditions observed in cancer patients, remains a major challenge for cancer immunotherapy. We reported previously that established tumors in mice receiving an agonistic mAb to the T cell costimulatory molecule 4-1BB (CD137) regress due to enhanced tumor antigen-specific cytotoxic T lymphocyte responses. In this study, we demonstrate that several poorly immunogenic tumors, including C3 tumor, TC-1 lung carcinoma, and B16-F10 melanoma, once established as solid tumors or metastases, are refractory to treatment by anti-4-1BB mAb. We provide evidence that immunological ignorance, rather than anergy or deletion, of tumor antigen--specific CTLs during the progressive growth of tumors prevents costimulation by anti-4-1BB mAb. Breaking CTL ignorance by immunization with a tumor antigen-derived peptide, although insufficient to stimulate a curative CTL response, is necessary for anti--4-1BB mAb to induce a CTL response leading to the regression of established tumors. Our results suggest a new approach for immunotherapy of human cancers.
Collapse
Affiliation(s)
- Ryan A Wilcox
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Wilcox RA, Flies DB, Zhu G, Johnson AJ, Tamada K, Chapoval AI, Strome SE, Pease LR, Chen L. Provision of antigen and CD137 signaling breaks immunological ignorance, promoting regression of poorly immunogenic tumors. J Clin Invest 2002; 109:651-9. [PMID: 11877473 PMCID: PMC150893 DOI: 10.1172/jci14184] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Treatment of advanced, poorly immunogenic tumors in animal models, considered the closest simulation available thus far for conditions observed in cancer patients, remains a major challenge for cancer immunotherapy. We reported previously that established tumors in mice receiving an agonistic mAb to the T cell costimulatory molecule 4-1BB (CD137) regress due to enhanced tumor antigen-specific cytotoxic T lymphocyte responses. In this study, we demonstrate that several poorly immunogenic tumors, including C3 tumor, TC-1 lung carcinoma, and B16-F10 melanoma, once established as solid tumors or metastases, are refractory to treatment by anti-4-1BB mAb. We provide evidence that immunological ignorance, rather than anergy or deletion, of tumor antigen--specific CTLs during the progressive growth of tumors prevents costimulation by anti-4-1BB mAb. Breaking CTL ignorance by immunization with a tumor antigen-derived peptide, although insufficient to stimulate a curative CTL response, is necessary for anti--4-1BB mAb to induce a CTL response leading to the regression of established tumors. Our results suggest a new approach for immunotherapy of human cancers.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigens, CD
- Antigens, Neoplasm/metabolism
- Female
- Humans
- Immune Tolerance
- Immunotherapy/methods
- Lymphocyte Activation/drug effects
- Mice
- Mice, Inbred C57BL
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Oncogene Proteins, Viral/immunology
- Papillomavirus E7 Proteins
- Receptors, Nerve Growth Factor/immunology
- Receptors, Tumor Necrosis Factor/immunology
- Signal Transduction/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Tumor Necrosis Factor Receptor Superfamily, Member 9
Collapse
Affiliation(s)
- Ryan A Wilcox
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nguyen LT, Elford AR, Murakami K, Garza KM, Schoenberger SP, Odermatt B, Speiser DE, Ohashi PS. Tumor growth enhances cross-presentation leading to limited T cell activation without tolerance. J Exp Med 2002; 195:423-35. [PMID: 11854356 PMCID: PMC2193619 DOI: 10.1084/jem.20010032] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Using a tumor model of spontaneously arising insulinomas expressing a defined tumor-associated antigen, we investigated whether tumor growth promotes cross-presentation and tolerance of tumor-specific T cells. We found that an advanced tumor burden enhanced cross-presentation of tumor-associated antigens to high avidity tumor-specific T cells, inducing T cell proliferation and limited effector function in vivo. However, contrary to other models, tumor-specific T cells were not tolerized despite a high tumor burden. In fact, in tumor-bearing mice, persistence and responsiveness of adoptively transferred tumor-specific T cells were enhanced. Accordingly, a potent T cell-mediated antitumor response could be elicited by intravenous administration of tumor-derived peptide and agonistic anti-CD40 antibody or viral immunization and reimmunization. Thus, in this model, tumor growth promotes activation of high avidity tumor-specific T cells instead of tolerance. Therefore, the host remains responsive to T cell immunotherapy.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/immunology
- Antigen Presentation
- Antigens, Neoplasm/immunology
- Antigens, Tumor-Associated, Carbohydrate/administration & dosage
- Antigens, Tumor-Associated, Carbohydrate/immunology
- CD40 Antigens/immunology
- Cell Division
- Flow Cytometry
- Hyaluronan Receptors/immunology
- Hyaluronan Receptors/metabolism
- Hypoglycemia/complications
- Immune Tolerance
- Immunologic Surveillance
- Immunotherapy, Active
- Insulinoma/complications
- Insulinoma/immunology
- Insulinoma/pathology
- Insulinoma/therapy
- Lymph Nodes/immunology
- Lymphocyte Activation
- Mice
- Mice, Transgenic
- Radiation Chimera
- Survival Analysis
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- Time Factors
Collapse
Affiliation(s)
- Linh T Nguyen
- Departments of Immunology and Medical Biophysics, Ontario Cancer Institute, 610 University Ave., Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Lin CM, Wang FH, Lee PK. Activated human CD4+ T cells induced by dendritic cell stimulation are most sensitive to transforming growth factor-beta: implications for dendritic cell immunization against cancer. Clin Immunol 2002; 102:96-105. [PMID: 11781072 DOI: 10.1006/clim.2001.5151] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The secretion of immunosuppressive factors like transforming growth factor-beta (TGF-beta) by tumor cells has been recognized as one of the mechanisms involved in tumor immunological escape. This study aimed to examine whether dendritic cell (DC) immunization could reverse TGF-beta-induced immunosuppression by simulating the in vivo interaction among infused DCs, host T cells, and tumor-secreted TGF-beta in an in vitro study. We found that both immature and mature DCs were relatively resistant to TGF-beta. The addition of TGF-beta to naive human CD4+ T cells, which are required by genetically modified DC to elicit antitumor immunity, resulted in their hyporesponsiveness to DC stimulation in a dose-dependent manner. When activated by allogeneic DCs in the presence of TGF-beta, CD4+ T cells displayed a reduced capacity to proliferate. More importantly, activated CD4+ T cells induced by DC stimulation were very sensitive to TGF-beta, and this susceptibility was enhanced by their previous exposure to TGF-beta. The underlying mechanism was linked to TGF-beta-induced apoptosis of activated T cells. However, the presence of stimulation from DC or antibodies to CD3 plus CD28 could partly reverse the immunosuppressive effect of TGF-beta on activated CD4+ T cells. Taken together, our results indicate that the efficacy of DC immunization may be impaired by tumor-derived TGF-beta.
Collapse
Affiliation(s)
- Chun-Ming Lin
- Department of Microbiology, Soochow University, Wai Shuang Hsi, Shih Lin, Taipei, Taiwan 11102, Republic of China.
| | | | | |
Collapse
|
47
|
Nelson DJ, Mukherjee S, Bundell C, Fisher S, van Hagen D, Robinson B. Tumor progression despite efficient tumor antigen cross-presentation and effective "arming" of tumor antigen-specific CTL. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:5557-66. [PMID: 11313395 DOI: 10.4049/jimmunol.166.9.5557] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To determine whether APC function or "arming" of CTL for lytic function are the points at which Ags from a nonimmunogenic tumor fail to induce an effective immune response, we established a murine tumor model that expressed intracellular OVA and selected a clone (cOVA-9) that remained susceptible to lysis by specific CD8(+) T cells throughout tumor growth. Viable cOVA-9 tumor cells grew in normal mice at a rate similar to the parental tumor, and vaccination with irradiated cOVA-9 cells did not induce protection against itself or the parental line, confirming its nonimmunogenic status. In vivo evaluation during tumor growth demonstrated persisting tumor Ag cross-presentation accompanied by the generation of potent, specific CTL which were detectable when tumors were barely palpable. Despite the presence of highly active CTL in the tumor-draining lymph nodes, there was no apparent lysis of tumor-associated APC. These data show that tumor-draining APC are not dysfunctional with regard to two crucial processes, in vivo tumor Ag cross-presentation and specific CTL arming, and that failure to prevent tumor growth is not in the induction phase, but in the effector phase and occurs within the tumor itself before the tumor matrix is established.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigen-Presenting Cells/pathology
- Antigen-Presenting Cells/radiation effects
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/prevention & control
- Cell Division/genetics
- Cell Division/immunology
- Cell Movement/immunology
- Cytoplasm/immunology
- Cytoplasm/metabolism
- Cytotoxicity, Immunologic/genetics
- Disease Progression
- Egg Proteins/biosynthesis
- Egg Proteins/genetics
- Egg Proteins/immunology
- Egg Proteins/metabolism
- Epitopes, T-Lymphocyte/immunology
- Female
- Histocompatibility Antigens Class I/metabolism
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation
- Ovalbumin/biosynthesis
- Ovalbumin/genetics
- Ovalbumin/immunology
- Ovalbumin/metabolism
- Peptide Fragments
- Spleen/cytology
- Spleen/immunology
- Stem Cells/cytology
- Stem Cells/immunology
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- Transfection
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/radiation effects
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- D J Nelson
- Department of Medicine, University of Western Australia, Nedlands, Queen Elizabeth II Medical Center, Perth, Western Australia, Australia.
| | | | | | | | | | | |
Collapse
|
48
|
So T, Takenoyama M, Sugaya M, Yasuda M, Eifuku R, Yoshimatsu T, Osaki T, Yasumoto K. Unfavorable prognosis of patients with non-small cell lung carcinoma associated with HLA-A2. Lung Cancer 2001; 32:39-46. [PMID: 11282427 DOI: 10.1016/s0169-5002(00)00204-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND HLA class I molecules present antigenic peptides to cytotoxic T lymphocytes and, thus, play an important role in immune surveillance. Since 1970s there have been many reports of an increased frequency of one or more HLA haplotype in association with autoimmune disease, and malignancy. We studied types of HLA class I antigens in 204 resected non-small cell lung carcinoma (NSCLC) patients and also examined its correlation with clinicopathologic features and prognosis. METHOD Serological typing for HLA class I antigens was performed using a microcytotoxicity test. The disease-free survival curves were calculated by the Kaplan-Meier method and then compared using the Logrank test. Multivariate analysis was carried out by Cox's proportional hazard method. RESULTS The difference in disease-free survival time between the HLA-A2 present group and A2 absent group was significant (P = 0.040). The 3-year disease-free survival rate of all patients was 44% in HLA-A2 present group and 66% in A2 absent group. When a comparison was made within the group with stage I, expression of HLA-A2 was the only independent factor that affected survival time by multivariate analysis (P = 0.0457). CONCLUSIONS Expression of HLA-A2 was considered as one of the unfavorable prognostic factors in NSCLC patients. Our results suggested expression of HLA-A2 in NSCLC patients was one of the mechanisms of escape from immune surveillance.
Collapse
Affiliation(s)
- T So
- Department of Surgery II, School of Medicine, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- D C Reid
- Department of Haematology, Northwick Park Hospital, Watford Road, Harrow HA1 3UJ, UK.
| |
Collapse
|
50
|
Ito H, Kurtz J, Shaffer J, Sykes M. CD4 T cell-mediated alloresistance to fully MHC-mismatched allogeneic bone marrow engraftment is dependent on CD40-CD40 ligand interactions, and lasting T cell tolerance is induced by bone marrow transplantation with initial blockade of this pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:2970-81. [PMID: 11207246 DOI: 10.4049/jimmunol.166.5.2970] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Costimulatory blockade can be used to promote allogeneic marrow engraftment and tolerance induction, but on its own is not 100% reliable. We sought to determine whether one or the other of the CD4 or CD8 T cell subsets of the recipient was primarily responsible for resistance to allogeneic marrow engraftment in mice receiving costimulatory blockade, and to use this information to develop a more reliable, minimal conditioning regimen for induction of mixed chimerism and transplantation tolerance. We demonstrate that a single anti-CD40 ligand mAb treatment is sufficient to completely overcome CD4 cell-mediated resistance to allogeneic marrow engraftment and rapidly induce CD4 cell tolerance, but does not reliably overcome CD8 CTL-mediated alloresistance. The data suggest that costimulation, which activates alloreactive CTL, is insufficient to activate alloreactive CD4 cells when the CD40 pathway is blocked. The addition of host CD8 T cell depletion to anti-CD40 ligand treatment reliably allows the induction of mixed chimerism and donor-specific skin graft tolerance in 3 Gy-irradiated mice receiving fully MHC-mismatched bone marrow grafts. Thus, despite the existence of multiple costimulatory pathways and pathways of APC activation, our studies demonstrate an absolute dependence on CD40-mediated events for CD4 cell-mediated rejection of allogeneic marrow. Exposure to donor bone marrow allows rapid tolerization of alloreactive CD4 cells when the CD40 pathway is blocked, leading to permanent marrow engraftment and intrathymic tolerization of T cells that develop subsequently.
Collapse
Affiliation(s)
- H Ito
- Bone Marrow Transplantation Section, Transplantation Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA
| | | | | | | |
Collapse
|