1
|
Au KM, Swinnea JS, Wang AZ. Intratumoral Injectable Redox-Responsive Immune Niche Improves the Abscopal Effect in Radiotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411330. [PMID: 39501983 PMCID: PMC11710980 DOI: 10.1002/adma.202411330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/03/2024] [Indexed: 01/11/2025]
Abstract
Radiotherapy (XRT) is often utilized to improve the immune checkpoint blockade response in cancer management. Such combination treatment can enhance the abscopal effect, facilitating a prolonged and durable systemic response. However, despite intense research efforts, only a minority of patients respond to this approach, and novel strategies to increase the abscopal effect are urgently needed. Here, the development of an intratumoral (i.t.) injectable nanofiber (NF)-based tumor immune niche (TIN) that converts XRT-treated tumors into an in situ cancer vaccine, eliciting robust systemic antitumor immunity, is reported. This NF-based immune niche incorporates redox-degradable anti-CTLA-4 (α-CTLA-4) nanogels (NGs) and interleukin-2 (IL-2) NGs for controlled release in hypoxic irradiated tumors, reversing the immunosuppressive tumor microenvironment into a pro-inflammatory microenvironment, and expanding the tumor-infiltrating CD8+ T cell population. Additionally, it is functionalized with polyinosinic-polycytidylic acid (poly(I:C)) to promote antigen-presenting cell maturation and prime neoantigen-specific CD8+ T cells. In vitro studies demonstrate TIN's ability to prime antigen-specific CD8+ T cells and increase antigen-specific cell-killing efficiency under in vitro immunosuppressive conditions. In vivo studies confirm TIN's ability to elicit robust systemic anticancer activity in mouse melanoma and colorectal cancer models without inducing severe immune-related adverse events.
Collapse
Affiliation(s)
- Kin Man Au
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75230, USA
| | - J. Steven Swinnea
- Department of Chemical Engineering, University of Texas at Austin, 200 E Dean Keeton St, Austin, TX 78712, USA
| | - Andrew Z. Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75230, USA
| |
Collapse
|
2
|
Bommakanti KK, Kosaraju N, Tam K, Chai-Ho W, St John M. Management of Cutaneous Head and Neck Squamous and Basal Cell Carcinomas for Immunocompromised Patients. Cancers (Basel) 2023; 15:3348. [PMID: 37444461 DOI: 10.3390/cancers15133348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
The incidence of non-melanoma skin cancer (NMSC) continues to rise, and more than one million cases are diagnosed in the United States each year. The increase in prevalence has been attributed to increased lifespan and improvements in survival for conditions that increase the risk of these malignancies. Patients who are immunocompromised have a higher risk of developing NMSC compared to the general population. In immunosuppressed patients, a combination of prevention, frequent surveillance, and early intervention are necessary to reduce morbidity and mortality. In this review, we collate and summarize current knowledge regarding pathogenesis of head and neck cutaneous SCC and BCC within immunocompromised patients, examine the potential role of the immune response in disease progression, and detail the role of novel immunotherapies in this subset of patients.
Collapse
Affiliation(s)
- Krishna K Bommakanti
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
- UCLA Head and Neck Cancer Program (HNCP), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
| | - Nikitha Kosaraju
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
| | - Kenric Tam
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
- UCLA Head and Neck Cancer Program (HNCP), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
| | - Wanxing Chai-Ho
- UCLA Head and Neck Cancer Program (HNCP), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
| | - Maie St John
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
- UCLA Head and Neck Cancer Program (HNCP), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1624, USA
| |
Collapse
|
3
|
Amôr NG, Santos PSDS, Campanelli AP. The Tumor Microenvironment in SCC: Mechanisms and Therapeutic Opportunities. Front Cell Dev Biol 2021; 9:636544. [PMID: 33634137 PMCID: PMC7900131 DOI: 10.3389/fcell.2021.636544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Squamous cell carcinoma (SCC) is the second most common skin cancer worldwide and, despite the relatively easy visualization of the tumor in the clinic, a sizeable number of SCC patients are diagnosed at advanced stages with local invasion and distant metastatic lesions. In the last decade, immunotherapy has emerged as the fourth pillar in cancer therapy via the targeting of immune checkpoint molecules such as programmed cell-death protein-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). FDA-approved monoclonal antibodies directed against these immune targets have provide survival benefit in a growing list of cancer types. Currently, there are two immunotherapy drugs available for cutaneous SCC: cemiplimab and pembrolizumab; both monoclonal antibodies (mAb) that block PD-1 thereby promoting T-cell activation and/or function. However, the success rate of these checkpoint inhibitors currently remains around 50%, which means that half of the patients with advanced SCC experience no benefit from this treatment. This review will highlight the mechanisms by which the immune checkpoint molecules regulate the tumor microenvironment (TME), as well as the ongoing clinical trials that are employing single or combinatory therapeutic approaches for SCC immunotherapy. We also discuss the regulation of additional pathways that might promote superior therapeutic efficacy, and consequently provide increased survival for those patients that do not benefit from the current checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Nádia Ghinelli Amôr
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Paulo Sérgio da Silva Santos
- Department of Surgery, Stomatology, Pathology, and Radiology, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Ana Paula Campanelli
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| |
Collapse
|
4
|
Sisakht M, Darabian M, Mahmoodzadeh A, Bazi A, Shafiee SM, Mokarram P, Khoshdel Z. The role of radiation induced oxidative stress as a regulator of radio-adaptive responses. Int J Radiat Biol 2020; 96:561-576. [PMID: 31976798 DOI: 10.1080/09553002.2020.1721597] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Purpose: Various sources of radiation including radiofrequency, electromagnetic radiation (EMR), low- dose X-radiation, low-level microwave radiation and ionizing radiation (IR) are indispensable parts of modern life. In the current review, we discussed the adaptive responses of biological systems to radiation with a focus on the impacts of radiation-induced oxidative stress (RIOS) and its molecular downstream signaling pathways.Materials and methods: A comprehensive search was conducted in Web of Sciences, PubMed, Scopus, Google Scholar, Embase, and Cochrane Library. Keywords included Mesh terms of "radiation," "electromagnetic radiation," "adaptive immunity," "oxidative stress," and "immune checkpoints." Manuscripts published up until December 2019 were included.Results: RIOS induces various molecular adaptors connected with adaptive responses in radiation exposed cells. One of these adaptors includes p53 which promotes various cellular signaling pathways. RIOS also activates the intrinsic apoptotic pathway by depolarization of the mitochondrial membrane potential and activating the caspase apoptotic cascade. RIOS is also involved in radiation-induced proliferative responses through interaction with mitogen-activated protein kinases (MAPks) including p38 MAPK, ERK, and c-Jun N-terminal kinase (JNK). Protein kinase B (Akt)/phosphoinositide 3-kinase (PI3K) signaling pathway has also been reported to be involved in RIOS-induced proliferative responses. Furthermore, RIOS promotes genetic instability by introducing DNA structural and epigenetic alterations, as well as attenuating DNA repair mechanisms. Inflammatory transcription factors including macrophage migration inhibitory factor (MIF), nuclear factor κB (NF-κB), and signal transducer and activator of transcription-3 (STAT-3) paly major role in RIOS-induced inflammation.Conclusion: In conclusion, RIOS considerably contributes to radiation induced adaptive responses. Other possible molecular adaptors modulating RIOS-induced responses are yet to be divulged in future studies.
Collapse
Affiliation(s)
- Mohsen Sisakht
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Darabian
- Department of Radiology, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Bazi
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Sayed Mohammad Shafiee
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Khoshdel
- Department of Medical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Faleiro R, Karunarathne DS, Horne-Debets JM, Wykes M. The Contribution of Co-signaling Pathways to Anti-malarial T Cell Immunity. Front Immunol 2018; 9:2926. [PMID: 30631323 PMCID: PMC6315188 DOI: 10.3389/fimmu.2018.02926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/29/2018] [Indexed: 12/30/2022] Open
Abstract
Plasmodium spp., the causative agent of malaria, caused 212 million infections in 2016 with 445,000 deaths, mostly in children. Adults acquire enough immunity to prevent clinical symptoms but never develop sterile immunity. The only vaccine for malaria, RTS,S, shows promising protection of a limited duration against clinical malaria in infants but no significant protection against severe disease. There is now abundant evidence that T cell functions are inhibited during malaria, which may explain why vaccine are not efficacious. Studies have now clearly shown that T cell immunity against malaria is subdued by multiple the immune regulatory receptors, in particular, by programmed cell-death-1 (PD-1). Given there is an urgent need for an efficacious malarial treatment, compounded with growing drug resistance, a better understanding of malarial immunity is essential. This review will examine molecular signals that affect T cell-mediated immunity against malaria.
Collapse
Affiliation(s)
- Rebecca Faleiro
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | | | - Michelle Wykes
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
6
|
B-Cell Depletion Reduces the Maturation of Cerebral Cavernous Malformations in Murine Models. J Neuroimmune Pharmacol 2016; 11:369-77. [PMID: 27086141 DOI: 10.1007/s11481-016-9670-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/30/2016] [Indexed: 02/05/2023]
Abstract
Cerebral cavernous malformations (CCMs) are relatively common vascular malformations, characterized by increased Rho kinase (ROCK) activity, vascular hyper-permeability and the presence of blood degradation products including non-heme iron. Previous studies revealed robust inflammatory cell infiltration, selective synthesis of IgG, in situ antigen driven B-cell clonal expansion, and deposition of immune complexes and complement proteins within CCM lesions. We aimed to evaluate the impact of suppressing the immune response on the formation and maturation of CCM lesions, as well as lesional iron deposition and ROCK activity. Two murine models of heterozygous Ccm3 (Pdcd10), which spontaneously develop CCM lesions with severe and milder phenotypes, were either untreated or received anti-mouse BR3 to deplete B cells. Brains from anti-mouse BR3-treated mice exhibited significantly fewer mature CCM lesions and smaller lesions compared to untreated mice. B cell depletion halted the progression of lesions into mature stage 2 lesions but did not prevent their genesis. Non-heme iron deposition and ROCK activity was decreased in lesions of B cell depleted mice. This represents the first report of the therapeutic benefit of B-cell depletion in the development and progression of CCMs, and provides a proof of principle that B cells play a critical role in CCM lesion genesis and maturation. These findings add biologics to the list of potential therapeutic agents for CCM disease. Future studies would characterize the putative antigenic trigger and further define the mechanism of immune response in the lesions.
Collapse
|
7
|
Schramm HM. Should EMT of Cancer Cells Be Understood as Epithelial-Myeloid Transition? J Cancer 2014; 5:125-32. [PMID: 24494030 PMCID: PMC3909767 DOI: 10.7150/jca.8242] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 01/02/2014] [Indexed: 12/11/2022] Open
Abstract
Cancer cells express epithelial markers, and when progressing in malignancy they may express markers of the mesenchymal cell type. Therefore an epithelial-mesenchymal transition of the cancer cells is assumed. However the mesenchymal markers can equally well be interpreted as myeloid markers since they are common in both types of cell lineages. Moreover, cancer cells express multiple specific markers of the myeloid lineages thus giving rise to the hypothesis that the transition of cancer cells may be from epithelial to myeloid cells and not to mesenchymal cells. This interpretation would better explain why cancer cells, often already in their primary cancer site, frequently show properties common to those of macrophages, platelets and pre-/osteoclasts.
Collapse
Affiliation(s)
- Henning M. Schramm
- Institute Hiscia, Society for Cancer Research, CH-4144 Arlesheim/Switzerland
| |
Collapse
|
8
|
Singh TP, Mayer G, Wolf P. In vivo siRNA targeting of CD28 reduces UV-induced DNA damage and inflammation. J Invest Dermatol 2013; 134:861-864. [PMID: 24042341 DOI: 10.1038/jid.2013.397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Tej Pratap Singh
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Gerlinde Mayer
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Peter Wolf
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
9
|
Xiang H, Zhao W, Sun Y, Qian W, Xing J, Zhou Y, Yao J, Xu J, Wang Y, Yao H, Hu Z. CD86 gene variants and susceptibility to pancreatic cancer. J Cancer Res Clin Oncol 2012; 138:2061-7. [PMID: 22821131 DOI: 10.1007/s00432-012-1289-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 07/02/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pancreatic cancer is one of the most lethal cancers worldwide. CD86 (B7-2) is a costimulatory molecule on antigen-presenting cells and plays critical roles in tumor immunity. It has been reported that polymorphisms in CD86 gene can be involved in the development of various cancers. Here, we investigated the association of two CD86 polymorphisms, +1057G/A (rs1129055) and +2379G/C (rs17281995), with pancreatic cancer in the Chinese population. METHODS The two polymorphisms were identified in 369 pancreatic cancer patients and 412 healthy controls using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Data were analyzed by chi-square test and adjusted for body mass index, smoking, drinking, and diabetes status. RESULTS Results showed that the frequency of the +1057A allele was significantly higher in pancreatic cancer cases than in controls (59.8 vs. 52.8 %, p = 0.021). Comparison of genotype frequencies showed that +1057GA and +1057AA genotypes were significantly increased in the pancreatic cancer group (odds ratio (OR) = 1.52; 95 % confidence interval (CI), 1.09-2.38; p = 0.026; and OR = 1.90; 95 % CI, 1.21-3.01; p = 0.007). We did not find any association between the +2379G/C polymorphism and pancreatic cancer. Analysis of haplotypes indicated that the AG (+1057, +2379) haplotype was correlated with the susceptibility to this disease (p = 0.019). CONCLUSIONS These results suggest that the CD86 +1057G/A polymorphism and AG (+1057, +2379) haplotype are genetic risk factors for pancreatic cancer.
Collapse
Affiliation(s)
- Honggang Xiang
- Department of General Surgery, Shanghai Changzheng Hospital, Second Military Medical University, 440 Chengdu North Road, Shanghai, 200003, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Ghafouri-Fard S, Ghafouri-Fard S. Immunotherapy in nonmelanoma skin cancer. Immunotherapy 2012; 4:499-510. [DOI: 10.2217/imt.12.29] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Nonmelanoma skin cancer is the most common type of cancer in humans. The role of the immune system in the prevention and regression of cancer is significant. UV radiation, being the most important risk factor in the development of skin cancer, has a suppressive effect on local and systemic immune effectors. Different immunotherapeutic approaches have been used for the treatment of nonmelanoma skin cancer including adoptive T-cell therapies, vaccine-based strategies, cytokines and monoclonal antibodies. The most important advancement with promising effects in the field of nonmelanoma skin cancer immunotherapy is the topical immune response modifier imiquimod. In addition, immunoprevention has been successfully applied for autosomal dominant basal cell nevus syndrome. Immunotherapeutic approaches provide a new modality for the treatment of recurrent or multiple nonmelanoma skin tumors.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| | - Somayyeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran
| |
Collapse
|
11
|
Abstract
Regulatory T cells (Tregs), either thymic derived or peripherally induced, suppress a variety of physiological and pathological immune responses, and the absence of this cell subset has been shown to result in severe systemic autoimmunity. Since their acceptance almost two decades ago, intensive research aiming to characterize the phenotype, to elucidate the suppressive activity, and to decipher the migratory behavior of Tregs has been performed. A substantial number of studies, however, focused on understanding whether defects in Treg numbers and function contribute to the development and progression of inflammatory, autoimmune, and malignant disorders, and how Treg numbers/function might be modulated to treat patients with autoimmune diseases or cancer. In the skin, an organ that is constantly exposed to the environment, Tregs are known to be critically involved not only in the maintenance of skin homeostasis but also in the regulation of cutaneous immune responses. In this review, we present an overview on recent data concerning Treg development and expansion, the molecular mechanisms underlying their immunosuppressive activity, and the modulation of Treg function. Furthermore, we discuss the role of Tregs in cutaneous inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Karin Loser
- Department of Dermatology, Interdisciplinary Center of Clinical Research, University of Münster, Münster, Germany.
| | | |
Collapse
|
12
|
Abstract
The immune system can identify and destroy nascent tumor cells in a process termed cancer immunosurveillance, which functions as an important defense against cancer. Recently, data obtained from numerous investigations in mouse models of cancer and in humans with cancer offer compelling evidence that particular innate and adaptive immune cell types, effector molecules, and pathways can sometimes collectively function as extrinsic tumor-suppressor mechanisms. However, the immune system can also promote tumor progression. Together, the dual host-protective and tumor-promoting actions of immunity are referred to as cancer immunoediting. In this review, we discuss the current experimental and human clinical data supporting a cancer immunoediting process that provide the fundamental basis for further study of immunity to cancer and for the rational design of immunotherapies against cancer.
Collapse
Affiliation(s)
- Matthew D Vesely
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
13
|
Wang W, Song H, Liu J, Song B, Cao X. CD86 + 1057G/A polymorphism and susceptibility to osteosarcoma. DNA Cell Biol 2011; 30:925-9. [PMID: 21563968 DOI: 10.1089/dna.2011.1211] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CD86 (B7-2), one of the costimulatory molecules on antigen-presenting cells, plays essential roles not only in autoimmunity and transplantation but also in tumor immunity. CD86 + 1057G/A polymorphism (rs1129055) is associated with various diseases. The objective of this study was to investigate the association between CD86 + 1057G/A polymorphism and susceptibility to osteosarcoma in a Chinese population. The CD86 + 1057G/A mutation was detected by polymerase chain reaction-restriction fragment length polymorphism in 205 osteosarcoma cases and 216 age-matched healthy controls. Frequencies of CD86 + 1057 AA genotype and +1057 A allele were significantly increased in osteosarcoma patients than in healthy controls (odds ratio = 2.18, 95% confidence interval, 1.21-3.93, p = 0.008; and odds ratio = 1.43, 95% confidence interval, 1.08-1.88, p = 0.011). Our data suggest that the +1057G/A polymorphism of the CD86 gene is associated with increased susceptibility to osteosarcoma.
Collapse
Affiliation(s)
- Wen Wang
- Traumatic Orthopedic Department, General Hospital of Jinan Military Command, Jinan, P.R. China
| | | | | | | | | |
Collapse
|
14
|
|
15
|
Pan XM, Gao LB, Liang WB, Liu Y, Zhu Y, Tang M, Li YB, Zhang L. CD86 +1057 G/A polymorphism and the risk of colorectal cancer. DNA Cell Biol 2010; 29:381-6. [PMID: 20380573 DOI: 10.1089/dna.2009.1003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
CD86 (B7-2), one of the costimulatory molecules on antigen-presenting cells, plays essential roles not only in autoimmunity and transplantation but also in tumor immunity. The purpose of this study was to investigate whether CD86 gene polymorphism was involved in predisposing an individual to colorectal cancer (CRC). The CD86 +1057 G/A polymorphism was genotyped by performing polymerase chain reaction-restriction fragment length polymorphism in 273 patients with CRC and 292 healthy controls. There were significant differences in the genotype and allele distribution of +1057 G/A polymorphism of the CD86 gene between cases and controls. The +1057 AA genotype was associated with a significantly increased risk of CRC when compared with the GG genotype (odds ratio [OR] = 2.16; 95% confidence interval [CI], 1.31-3.58). Using the G allele as a reference, a significant correlation was detected between the presence of the A allele and a risk of developing CRC (OR = 1.42; 95% CI, 1.12-1.80). Interestingly, the A allele in female patients with CRC was significantly higher than that in male patients after stratified analysis (OR = 1.49; 95% CI, 1.04-2.14). These data suggest that CD86 +1057G/A polymorphism may contribute to genetic susceptibility to CRC in a Chinese population.
Collapse
Affiliation(s)
- Xin-Min Pan
- Department of Forensic Biology, Sichuan University, Chengdu, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
de Gruijl FR, Koehl GE, Voskamp P, Strik A, Rebel HG, Gaumann A, de Fijter JW, Tensen CP, Bavinck JNB, Geissler EK. Early and late effects of the immunosuppressants rapamycin and mycophenolate mofetil on UV carcinogenesis. Int J Cancer 2010; 127:796-804. [PMID: 19998342 DOI: 10.1002/ijc.25097] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Increased skin cancer risk in organ transplant recipients has been experimentally emulated with enhanced UV carcinogenesis from administering conventional immunosuppressants. However, newer generation immunosuppressive drugs, rapamycin (Rapa) and mycophenolate mofetil (MMF), have been shown to impair angiogenesis and outgrowth of tumor implants. To ascertain the overall effect on UV carcinogenesis, Rapa and MMF were admixed into the food pellets of hairless SKH1 mice receiving daily sub-sunburn UV dosages. With immunosuppressive blood levels neither of the drugs affected onset of tumors (<2 mm), but in contrast to MMF, Rapa significantly increased latency of large tumors (>or=4 mm, medians of 190 vs 125 days) and reduced their multiplicity (1.6 vs 4.5 tumors per mouse at 200 days). Interestingly, tumors (>2 mm) from the Rapa-fed group showed a reduction in UV-signature p53 mutations (39% vs 90%) in favor of mutations from putative base oxidation. This shift in mutation spectrum was not essentially linked to the reduction in large tumors because it was absent in large tumors similarly reduced in number when feeding Rapa in combination with MMF, possibly owing to an antioxidant effect of MMF. Significantly fewer tumor cells were Vegf-positive in the Rapa-fed groups, but a correspondingly reduced expression of Hif1alpha target genes (Vegf, Ldha, Glut1, Pdk1) that would indicate altered glucose metabolism with increased oxidative stress was not found. Remarkably, we observed no effect of the immunosuppressants on UV-induced tumor onset, and with impaired tumor outgrowth Rapa could therefore strongly reduce skin carcinoma morbidity and mortality rates in organ transplant recipients.
Collapse
Affiliation(s)
- F R de Gruijl
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Singh TP, Schön MP, Wallbrecht K, Michaelis K, Rinner B, Mayer G, Schmidbauer U, Strohmaier H, Wang XJ, Wolf P. 8-methoxypsoralen plus ultraviolet A therapy acts via inhibition of the IL-23/Th17 axis and induction of Foxp3+ regulatory T cells involving CTLA4 signaling in a psoriasis-like skin disorder. THE JOURNAL OF IMMUNOLOGY 2010; 184:7257-67. [PMID: 20488788 DOI: 10.4049/jimmunol.0903719] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To elucidate the molecular action of 8-methoxypsoralen plus UVA (PUVA), a standard dermatological therapy, we used K5.hTGF-beta1 transgenic mice exhibiting a skin phenotype and cytokine abnormalities with strong similarities to human psoriasis. We observed that impaired function of CD4+CD25+ regulatory T cells (Tregs) and increased cytokine levels of the IL-23/Th17 pathway were responsible for the psoriatic phenotype in this mouse model. Treatment of K5.hTGF-beta1 transgenic mice with PUVA suppressed the IL-23/Th17 pathway, Th1 milieu, as well as transcription factors STAT3 and orphan nuclear receptor RORgammat. PUVA induced the Th2 pathway and IL-10-producing CD4+CD25+Foxp3+Tregs with disease-suppressive activity that was abolished by anti-CTLA4 mAb treatment. These findings were paralleled by macroscopic and microscopic clearance of the diseased murine skin. Anti-IL-17 mAb treatment also diminished the psoriatic phenotype of the mice. This indicated that both induced Tregs involving CTLA4 signaling and inhibition of the IL-23/Th17 axis are central for the therapeutic action of PUVA.
Collapse
Affiliation(s)
- Tej Pratap Singh
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Duncan FJ, Martin JR, Wulff BC, Stoner GD, Tober KL, Oberyszyn TM, Kusewitt DF, Van Buskirk AM. Topical treatment with black raspberry extract reduces cutaneous UVB-induced carcinogenesis and inflammation. Cancer Prev Res (Phila) 2009; 2:665-72. [PMID: 19584078 DOI: 10.1158/1940-6207.capr-08-0193] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Light in the UVB spectrum (280-320 nm) induces a number of changes in the epidermis and dermis of mice and humans, resulting in a robust inflammatory response. A standardized black raspberry extract (BRE) has been effective in reducing signaling pathways commonly initiated by inflammatory stimuli. In this study, we determined whether this extract could reduce cutaneous UVB-induced inflammation and carcinogenesis. In our carcinogenesis model, female SKH-1 hairless mice were exposed to one minimal erythemal dose of UVB thrice weekly on nonconsecutive days for 25 weeks. Immediately after each exposure, the mice were treated topically with either BRE dissolved in vehicle or with vehicle only. Beginning on week 19, mice treated with BRE had a significant reduction in tumor number and in average tumor size. This reduction correlated with a significant reduction in tumor-infiltrating CD3(+)foxp3(+) regulatory T-cells. In the acute model, mice were exposed to a single minimal erythemal dose of UVB and treated topically with BRE or with vehicle. At 48 hours post-UVB exposure, topical BRE treatment significantly reduced edema, p53 protein levels, oxidative DNA damage, and neutrophil activation. The ability of topical BRE to reduce acute UVB-induced inflammation and to decrease tumor development in a long-term model provides compelling evidence to explore the clinical efficacy of BRE in the prevention of human skin cancers.
Collapse
Affiliation(s)
- F J Duncan
- Department of Pathology, The Ohio State University, 1645 Neil Avenue, 129 Hamilton Hall, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Flies DB, Chen L. Modulation of Immune Response by B7 Family Molecules in Tumor Microenvironments. Immunol Invest 2009; 35:395-418. [PMID: 16916759 DOI: 10.1080/08820130600755017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The importance of co-stimulatory and co-inhibitory molecules has been confirmed on a grand scale; with the identification of new B7 family molecules, possessing both immune activating and inhibiting functions, this family has exploded onto the scene of immune regulation. Nowhere, however, has the role of B7 family members been more apparent than in the fight against cancer. In this review, we will discuss recent data regarding the essential and complex role of B7 family members in regulating the immune response within tumor microenvironment.
Collapse
Affiliation(s)
- Dallas B Flies
- Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | |
Collapse
|
20
|
Bour-Jordan H, Bluestone JA. Regulating the regulators: costimulatory signals control the homeostasis and function of regulatory T cells. Immunol Rev 2009; 229:41-66. [PMID: 19426214 PMCID: PMC2714548 DOI: 10.1111/j.1600-065x.2009.00775.x] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SUMMARY Costimulation is a concept that goes back to the early 1980s when Lafferty and others hypothesized that cell surface and soluble molecules must exist that are essential for initiating immune responses subsequent to antigen exposure. The explosion in this field of research ensued as over a dozen molecules have been identified to function as second signals following T-cell receptor engagement. By 1994, it seemed clear that the most prominent costimulatory pathway CD28 and functionally related costimulatory molecules, such as CD154, were the major drivers of a positive immune response. Then the immunology world turned upside down. CD28 knockout mice, which were, in most cases, immunodeficient, led to increased autoimmunity when bred into the non-obese diabetic background. Another CD28 family member, cytotoxic T-lymphocyte-associated protein 4, which was presumed to be a costimulatory molecule on activated T cells, turned out to be critical in downregulating immunity. These results, coupled with the vast suppressor cell literature which had been largely rebuked, suggested that the immune system was not poised for response but controlled in such a way that regulation was dominant. Over the last decade, we have learned that these costimulatory molecules play a key role in the now classical CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) that provide critical control of unwanted autoimmune responses. In this review, we discuss the connections between costimulation and Tregs that have changed the costimulation paradigm.
Collapse
Affiliation(s)
- Hélène Bour-Jordan
- UCSF Diabetes Center, University of California at San Francisco, San Francisco, CA USA
| | - Jeffrey A. Bluestone
- UCSF Diabetes Center, University of California at San Francisco, San Francisco, CA USA
| |
Collapse
|
21
|
Loser K, Beissert S. Regulation of cutaneous immunity by the environment: an important role for UV irradiation and vitamin D. Int Immunopharmacol 2009; 9:587-9. [PMID: 19539561 DOI: 10.1016/j.intimp.2009.01.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 01/22/2009] [Indexed: 10/21/2022]
Abstract
Cutaneous immunity can be controlled by environmental factors such as ultraviolet (UV) irradiation. UV irradiation affects keratinocytes, antigen presenting cells, such as epidermal Langerhans cells (LC), and T lymphocytes. LC are specialized in antigen presentation. Upon encountering exogenous antigens they migrate to skin draining lymph nodes where they present skin-acquired antigens to naive T cells resulting in effector T cell differentiation. T cell effector functions depend on the activation state of LC, which can be influenced by UV irradiation. After completion T cell mediated cutaneous immune responses need to be downregulated. In this context, CD4(+)CD25(+) regulatory T cells have been shown to play an important role in the suppression of cellular immune responses via inhibition of T cell proliferation. Naturally occurring regulatory T cells develop in the thymus and on the molecular level members of the B7- and TNF-superfamilies are critically involved in the peripheral maintenance of CD4(+)CD25(+) T cells. Substantial evidence exists that peripheral regulatory T cells are responsive to environmental stimuli including UV irradiation. UV-induced regulatory T cells are expanded by UV-exposed cutaneous LC and recently, epidermal expression of vitamin D3 or RANKL (CD254) has been shown to connect the environment to the immune system via expansion of CD4(+)CD25(+) regulatory T cells.
Collapse
Affiliation(s)
- Karin Loser
- Department of Dermatology and Interdisciplinary Center of Clinical Research (IZKF), University of Münster, Münster, Germany.
| | | |
Collapse
|
22
|
Abstract
On the basis of experimental models and some human data, we can assume that tumor outgrowth results from the balance between immunosurveillance (the extrinsic tumor suppressor mechanisms) and immunosubversion dictated by transformed cells and/or the corrupted surrounding microenvironment. Cancer immunosurveillance relies mainly upon conventional lymphocytes exerting either lytic or secretory functions, whereas immunosubversion results from the activity of regulatory T or suppressor myeloid cells and soluble mediators. Although specific tools to target or ablate dendritic cells (DCs) became only recently available, accumulating evidence points to the critical role of the specialized DC system in dictating most of the conventional and regulatory functions of tumor-specific T lymphocytes. Although DC can be harnessed to silence tumor development, tumors in turn can exploit DC to evade immunity. Indeed, DCs harbor defects in their differentiation and stimulatory functions in cancer-bearing hosts and can actively promote T-cell tolerance to self-tumor antigens. In this review, we will focus on the dual role of DC during tumor progression and discuss pharmacoimmunological strategies to harness DC against cancer.
Collapse
|
23
|
Kuhn A, Beissert S, Krammer PH. CD4(+)CD25 (+) regulatory T cells in human lupus erythematosus. Arch Dermatol Res 2008; 301:71-81. [PMID: 18985367 DOI: 10.1007/s00403-008-0891-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 08/22/2008] [Indexed: 11/30/2022]
Abstract
Natural CD4(+)CD25(+) regulatory T cells (T(reg)) show a potent immunosuppressive function and contribute to immunologic self-tolerance by suppressing potentially auto-reactive T cells. Depletion of these cells leads to the induction of severe autoimmune diseases in animal models; more recently, several studies have also reported an impairment of T(reg) number and/or function in various human autoimmune diseases. For example, aberrant numbers of circulating CD4(+)CD25(+) T(reg) have been seen in patients with type I diabetes, mycosis fungoides, graft-versus-host-reaction, and rheumatoid arthritis. Moreover, increased numbers of functionally active CD4(+)CD25(+) T(reg) have been detected in the synovial fluid of patients with rheumatoid arthritis. In systemic lupus erythematosus (SLE), conflicting data on the role of CD4(+)CD25(+) T(reg) in human autoimmune diseases have been presented in the literature. Decreased numbers of peripheral blood T(reg) have been reported by most studies on SLE patients with active disease, but non-impaired or even increased CD4(+)CD25(+) T(reg) numbers have also been described. In addition, both deficient and normal suppressive capacity of isolated T(reg) have been observed in SLE. Analysis of CD4(+)FoxP3(+) T(reg) in skin lesions of patients with a primarily cutaneous manifestation of the disease showed a significant reduction in cell numbers as compared to other inflammatory skin diseases, suggesting the importance of analyzing T(reg) numbers in the affected tissue. In this review, we discuss the role of CD4(+)CD25(+) T(reg) in autoimmunity and recent published data on SLE. Furthermore, we highlight the need for additional studies that address specific gaps of knowledge regarding the pathophysiological mechanisms as well as the identification of future therapeutic strategies for autoimmune diseases.
Collapse
Affiliation(s)
- Annegret Kuhn
- Division of Immunogenetics, Tumor Immunology Program, German Cancer Research Center, Heidelberg, Germany.
| | | | | |
Collapse
|
24
|
Barolet D, Boucher A. LED photoprevention: reduced MED response following multiple LED exposures. Lasers Surg Med 2008; 40:106-12. [PMID: 18306161 DOI: 10.1002/lsm.20615] [Citation(s) in RCA: 269] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND AND OBJECTIVES As photoprotection with traditional sunscreen presents some limitations, the use of non-traditional treatments to increase skin resistance to ultraviolet (UV) induced damage would prove particularly appealing. The purpose of this pilot study was to test the potential of non-thermal pulsed light-emitting diode (LED) treatments (660 nm) prior to UV exposure in the induction of a state of cellular resistance against UV-induced erythema. STUDY DESIGN/MATERIALS AND METHODS Thirteen healthy subjects and two patients with polymorphous light eruption (PLE) were exposed to 5, 6, or 10 LED treatments (660 nm) on an EXPERIMENTAL anterior thigh region. Individual baseline minimal erythema doses (MED) were then determined. UV radiation was thereafter performed on the LED EXPERIMENTAL and CONTROL anterior thigh areas. Finally, 24 hours post-UV irradiation, LED pre-treated MED responses were compared to the non-treated sites. RESULTS Reduction of erythema was considered significant when erythema was reduced by >50% on the LED-treated side as opposed to CONTROL side. A significant LED treatment reduction in UV-B induced erythema reaction was observed in at least one occasion in 85% of subjects, including patients suffering from PLE. Moreover, there was evidence of a dose-related pattern in results. Finally, a sun protection factor SPF-15-like effect and a reduction in post-inflammatory hyperpigmentation were observed on the LED pre-treated side. CONCLUSIONS Results suggest that LED based therapy prior to UV exposure provided significant protection against UV-B induced erythema. The induction of cellular resistance to UV insults may possibly be explained by the induction of a state a natural resistance to the skin via specific cell signaling pathways and without the drawbacks and limitations of traditional sunscreens. These results represent an encouraging step towards expanding the potential applications of LED therapy and could be useful in the treatment of patients with anomalous reactions to sunlight.
Collapse
Affiliation(s)
- Daniel Barolet
- RoseLab, Skin Optics Laboratory, Montreal, Quebec, Canada.
| | | |
Collapse
|
25
|
Loos M, Giese NA, Kleeff J, Giese T, Gaida MM, Bergmann F, Laschinger M, W Büchler M, Friess H. Clinical significance and regulation of the costimulatory molecule B7-H1 in pancreatic cancer. Cancer Lett 2008; 268:98-109. [PMID: 18486325 DOI: 10.1016/j.canlet.2008.03.056] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 03/19/2008] [Accepted: 03/25/2008] [Indexed: 12/25/2022]
Abstract
We investigated the expression pattern and clinical significance of the costimulatory ligands B7-1, B7-2, B7-H1, and B7-DC, and their counter-receptors CTLA-4 and PD-1 in pancreatic cancer. Gene expression of all examined costimulatory molecules was significantly upregulated in pancreatic cancer tissues. B7-1, B7-2, B7-H1, and B7-DC protein was detectable in pancreatic cancer cells. Only the expression of B7-H1 significantly correlated with postoperative survival (p<0.0001). B7-H1 was inducible in cultured pancreatic cancer cells by IFN-gamma and significantly correlated with the level of IFN-gamma expression in human pancreatic cancer tissues (Spearman rho=0.4536,p=0.0029). B7-H1 positive tumors showed an increased prevalence of tumor-infiltrating regulatory T cells (Tregs) compared to B7-H1 negative tumors. Among the investigated costimulatory molecules only tumor-associated B7-H1 seems to be of prognostic relevance in pancreatic cancer. B7-H1 might, therefore, be involved in the downregulation of antitumor responses through regulation of Tregs in pancreatic cancer. Our findings also suggest a dual role of IFN-gamma in antitumor response. Through induction of B7-H1 in pancreatic cancer cells IFN-gamma might contribute to the evasion of antitumor immunity.
Collapse
Affiliation(s)
- Martin Loos
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675 Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Skin cancer constitutes one of the most frequent types of malignancies in humans with rapidly increasing incidences almost worldwide. UVR is an essential risk factor for the development of premalignant as well as malignant skin lesions. In this context UVR can function as a complete carcinogen by inducing "UV signature" DNA mutations and by suppressing protective cellular antitumoral immune responses. UV-induced DNA damage can result in impaired cutaneous cell cycle control if cell cycle regulators, such as the p53 gene, are affected. Besides interfering with cell cycle control genes, UV-induced DNA damage can result in the release of interleukin-10, a cytokine with known immunosuppressive effects on T-helper(h)-1 cells. For the development of antitumoral immune responses antigen-specific activation of effector T cells by antigen-presenting cells (APC) is required. It was demonstrated that UVR can inhibit antigen presentation both directly and indirectly via the induction of suppressive cytokines. In addition, subsets of T cells are induced upon UVR, which can actively suppress major histocompatibility complex class I/II-restricted immune responses. These UV-induced regulatory T cells appear to belong to the CD4+CD25+ T cell lineage and can express the characteristic transcription factor Foxp3, which programs for suppressor function. In mice UV-induced regulatory T cells can control the development of UV-induced skin cancer. Peripheral regulatory T cells are maintained by the expression of B7 molecules and can be expanded by APC of the skin. Recently, epidermal expression of CD254 (RANKL) has been shown to connect UVR with the expansion of regulatory CD4+CD25+ T cells. In the following, new molecular and cellular mechanisms of UV-induced skin tumor development will be described and discussed.
Collapse
Affiliation(s)
- Stefan Beissert
- Department of Dermatology, University of Münster, Münster, Germany.
| | | |
Collapse
|
27
|
Halliday GM, Rana S. Waveband and dose dependency of sunlight-induced immunomodulation and cellular changes. Photochem Photobiol 2008; 84:35-46. [PMID: 18173699 DOI: 10.1111/j.1751-1097.2007.00212.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Both the UVB and UVA wavebands within sunlight are immunosuppressive. This article reviews the relationship between wavebands and dose in UV-induced immunosuppression mainly concentrating on responses in humans. It also contrasts the effects of UVB and UVA on cellular changes involved in immunosuppression. Over physiological sunlight doses to which humans can be exposed during routine daily living or recreational pursuits, both UVA and UVB suppress immunity. While there is a linear dose relationship with UVB commencing at doses less than half of what is required to cause sunburn, UVA has a bell-shaped dose response over the range to which humans can be realistically exposed. At doses too low for either waveband to be suppressive, interactions between UVA and UVB augment each other, enabling immunosuppression to occur. At doses beyond where UVA is immunosuppressive, it still contributes to sunlight-induced immunosuppression via this interaction with UVB. While there is little research comparing the mechanisms by which UVB, UVA and their interactions can cause immunosuppression, it is likely that different chromophores and early molecular events are involved. There is evidence that both wavebands disrupt antigen presentation and effect T cell responses. Different individuals are likely to have different immunomodulatory responses to sunlight.
Collapse
Affiliation(s)
- Gary M Halliday
- Dermatology Research Laboratories, Melanoma and Skin Cancer Research Institute and Bosch Institute, University of Sydney, Sydney, NSW, Australia.
| | | |
Collapse
|
28
|
Himoudi N, Nabarro S, Yan M, Gilmour K, Thrasher AJ, Anderson J. Development of anti-PAX3 immune responses; a target for cancer immunotherapy. Cancer Immunol Immunother 2007; 56:1381-95. [PMID: 17318653 PMCID: PMC11030167 DOI: 10.1007/s00262-007-0294-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Accepted: 01/21/2007] [Indexed: 10/23/2022]
Abstract
PAX3 is overexpressed in several human cancers and is absent from normal adult human tissues. It is known to have an oncogenic function in human malignancy, and is therefore a promising target for cancer immunotherapy. We screened the murine and human PAX3 amino acid sequences for peptides that bind common MHC class I types, and identified murine GVFINGRPL and human KLTEARVQV sequences. Mice immunised with either a selected PAX3 peptide, or with a PAX3 expressing DNA vector, developed specific anti-PAX3 immune responses that inhibited tumour growth. The intensity of the immune response was significantly enhanced by pulsing of the peptide onto dendritic cells. Anti-PAX3 T cell lines were established from splenocytes of immunised mice. Intravenous administration of anti-PAX3 T cells caused regression of established tumours indicating a promising clinical application for anti-PAX3 immunotherapy. The human peptide stimulated growth of similar T cell lines from peripheral blood of three out of three normal human blood donors. These showed specific cytotoxicity against a range of human PAX3+ and HLA-A2+ cancer cell lines. Moreover, an anti-PAX3 response was detected as a component of the anti-tumour immune response in a patient treated with lysate pulsed dendritic cell vaccination. The ability to generate strong and specific anti PAX3 immune responses from the T cell repertoire in both mice and humans, provides evidence for PAX3 as a promising target for immunotherapy of cancer.
Collapse
Affiliation(s)
- Nourredine Himoudi
- Unit of Molecular Haematology and Cancer Biology, Institute of Child Health, London, WC1N 1EH UK
| | - Steven Nabarro
- Unit of Molecular Haematology and Cancer Biology, Institute of Child Health, London, WC1N 1EH UK
| | - Mengyong Yan
- Unit of Molecular Haematology and Cancer Biology, Institute of Child Health, London, WC1N 1EH UK
| | - Kimberly Gilmour
- Unit of Molecular Immunology, Institute of Child Health, London, WC1N 1EH UK
| | - Adrian J. Thrasher
- Unit of Molecular Immunology, Institute of Child Health, London, WC1N 1EH UK
| | - John Anderson
- Unit of Molecular Haematology and Cancer Biology, Institute of Child Health, London, WC1N 1EH UK
- Department of Paediatric Oncology, Great Ormond Street Hospital, London, WC1N 3JH UK
| |
Collapse
|
29
|
Loser K, Apelt J, Voskort M, Mohaupt M, Balkow S, Schwarz T, Grabbe S, Beissert S. IL-10 controls ultraviolet-induced carcinogenesis in mice. THE JOURNAL OF IMMUNOLOGY 2007; 179:365-71. [PMID: 17579057 DOI: 10.4049/jimmunol.179.1.365] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
UV radiation-induced immunosuppression contributes significantly to the development of UV-induced skin cancer by inhibiting protective immune responses. IL-10 has been shown to be a key mediator of UV-induced immunosuppression. To investigate the role of IL-10 during photocarcinogenesis, groups of IL-10(+/+), IL-10(+/-), and IL-10(-/-) mice were chronically irradiated with UV. IL-10(+/+) and IL-10(+/-) mice developed skin cancer to similar extents, whereas IL-10(-/-) mice were protected against the induction of skin malignancies by UV. Because UV is able to induce regulatory T cells, which play a role in the suppression of protective immunity, UV-induced regulatory T cell function was analyzed. Splenic regulatory T cells from UV-irradiated IL-10(-/-) mice were unable to confer immunosuppression upon transfer into naive recipients. UV-induced CD4+CD25+ T cells from IL-10(-/-) mice showed impaired suppressor function when cocultured with conventional CD4+CD25- T cells. CD4+CD25- T cells from IL-10(-/-) mice produced increased amounts of IFN-gamma and enhanced numbers of CD4+TIM-3+ T cells were detectable within UV-induced tumors in IL-10(-/-) mice, suggesting strong Th1-driven immunity. Mice treated with CD8+ T cells from UV-irradiated IL-10(-/-) mice rejected a UV tumor challenge significantly faster, and augmented numbers of granzyme A+ cells were detected within injected UV tumors in IL-10(-/-) animals, suggesting marked antitumoral CTL responses. Together, these findings indicate that IL-10 is critically involved in antitumoral immunity during photocarcinogenesis. Moreover, these results point out the crucial role of Th1 responses and UV-induced regulatory T cell function in the protection against UV-induced tumor development.
Collapse
MESH Headings
- Animals
- Cytokines/biosynthesis
- Immune Tolerance/genetics
- Immune Tolerance/radiation effects
- Interleukin-10/deficiency
- Interleukin-10/genetics
- Interleukin-10/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Neoplasms, Radiation-Induced/genetics
- Neoplasms, Radiation-Induced/immunology
- Neoplasms, Radiation-Induced/pathology
- Neoplasms, Radiation-Induced/prevention & control
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Skin Neoplasms/prevention & control
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- T-Lymphocytes, Regulatory/radiation effects
- Ultraviolet Rays
Collapse
Affiliation(s)
- Karin Loser
- Department of Dermatology and Interdisciplinary Center of Clinical Research, Interdisziplinäres Zentrum für Klinische Forschung, University of Münster, Von-Esmarch-Strasse 58, Münster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The ability of the immune system to identify and destroy nascent tumors, and to thereby function as a primary defense against cancer, has been debated for many decades. Recent findings by a number of investigators in both mouse models of cancer and humans with cancer now offer compelling evidence that particular immune cell types, effector molecules, and pathways can sometimes collectively function as extrinsic tumor suppressor mechanisms. This work provides the basis for further study of natural immunity to cancer and for rational use of this information in the design of immunotherapies in combination with other conventional cancer treatments.
Collapse
Affiliation(s)
- Jeremy B Swann
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne 8006, Victoria, Australia
| | | |
Collapse
|
31
|
Gorman S, Tan JWY, Yerkovich ST, Finlay-Jones JJ, Hart PH. CD4+ T Cells in Lymph Nodes of UVB-Irradiated Mice Suppress Immune Responses to New Antigens Both In Vitro and In Vivo. J Invest Dermatol 2007; 127:915-24. [PMID: 17068476 DOI: 10.1038/sj.jid.5700600] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mechanisms by which erythemal UVB irradiation modulates systemic immune responses to antigens applied to non-irradiated sites are poorly understood. In this study, regulatory CD4+ T cells were identified in the skin-draining lymph nodes (SDLNs) of UVB-irradiated, but otherwise naive mice. A transgenic mouse strain (DO11.10) was utilized in which the majority of CD4+ T cells expressed the ovalbumin (OVA(323-339)) T-cell receptor. Thus, T-cell responses could be examined following erythemal UVB irradiation without further antigen sensitization. CD4+ T cells from the SDLNs of UVB-irradiated mice had significantly reduced capacity to respond to presentation of the OVA(323-339) peptide in vitro. Transfer of CD4+ T cells from the SDLNs of UVB-irradiated antigen-naive mice significantly reduced both OVA sensitization and contact hypersensitivity responses to an experimental hapten in the recipient mice. Depletion of CD4+CD25+ cells abrogated this UVB-suppressive effect in the in vitro proliferation assay. There was also a significant increase in the proportion of CD4+CD25+Foxp3+ cells in the SDLNs of UVB-irradiated mice. The potential of these regulatory cells poised to regulate responses to incoming antigens at distant non-irradiated sites broadens the biological impact of UVB irradiation of skin on immunity.
Collapse
Affiliation(s)
- Shelley Gorman
- Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, Perth, Western Australia, Australia
| | | | | | | | | |
Collapse
|
32
|
|
33
|
Paus R, Loser K, Beissert S. Commentary 1. Exp Dermatol 2006. [DOI: 10.1111/j.1600-0625.2006.00506_8.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
34
|
Paus R, Schröder JM, Reich K, Kabashima K, Liu FT, Romani N, Metz M, Kerstan A, Lee PHA, Loser K, Schön MP, Maurer M, Stoitzner P, Beissert S, Tokura Y, Gallo RL, Reich K. Who is really in control of skin immunity underphysiologicalcircumstances - lymphocytes, dendritic cells or keratinocytes? Exp Dermatol 2006. [DOI: 10.1111/j.1600-0625.2006.00506.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
35
|
Sansom DM, Walker LSK. The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol Rev 2006; 212:131-48. [PMID: 16903911 DOI: 10.1111/j.0105-2896.2006.00419.x] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The profound influence of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) on T-cell immunity has been known for over a decade, yet the precise roles played by these molecules still continue to emerge. Initially viewed as molecules that provide cell-intrinsic costimulatory and inhibitory signals, recent evidence suggests that both CD28 and CTLA-4 are also important in the homeostasis and function of a population of suppressive cells, termed regulatory T cells (Tregs). Here we review the main features of the CD28 and CTLA-4 system and examine how these impact upon Treg biology.
Collapse
Affiliation(s)
- David M Sansom
- MRC Centre for Immune Regulation, University of Birmingham Medical School, Birmingham, UK.
| | | |
Collapse
|
36
|
Abstract
Immunologic self-tolerance is critically dependent on the induction but also on the downregulation of immune responses. Though ignored and neglected for many years, suppressor T cells, now renamed regulatory T cells (Tregs), play an important role in the negative regulation of immune responses. Several subsets of Tregs have been described. Naturally occurring CD4(+)CD25(+) Tregs are important in the prevention of autoimmune diseases. Type 1 Tregs, another subtype of Treg that is inducible, exert their suppressive activity primarily via the release of IL-10. Detailed knowledge about the phenotype and mode of action of these cells will significantly increase our understanding of the pathogenesis of autoimmune diseases and will also help to identify new therapeutic strategies.
Collapse
Affiliation(s)
- Stefan Beissert
- Department of Dermatology, University of Münster, Münster, Germany
| | | | | |
Collapse
|
37
|
Abstract
Lupus erythematosus (LE) is an autoimmune disease which can be triggered by environmental factors such as solar irradiation. It has long been observed that especially ultraviolet (UV) exposure can induce and exacerbate skin lesions in patients with this disease. However, despite the frequency of photosensitivity in LE, the mechanisms by which UV irradiation activates autoimmune responses is only now becoming increasingly unfolded by advanced molecular and cellular biological investigations. Phototesting, according to a standardized protocol with UVA and UVB irradiation has proven to be a valid model to study photosensitivity in various subtypes of LE and to evaluate the underlying pathomechanisms of this disease. Detailed analysis of the molecular events that govern lesion formation in experimentally photoprovoced LE showed increased accumulation of apoptotic keratinocytes and impaired expression of the inducible nitric oxide synthase (iNOS). In the near future, gene expression profiling and proteomics will further increase our knowledge on the complexity of the "UV response" in LE. This review summarizes the current understanding of the clinical and molecular mechanisms that initiate photosensitivity in this disease.
Collapse
Affiliation(s)
- Annegret Kuhn
- Department of Dermatology, University of Düsseldorf, Düsseldorf, Germany.
| | | |
Collapse
|
38
|
Zhong RK, Loken M, Lane TA, Ball ED. CTLA-4 blockade by a human MAb enhances the capacity of AML-derived DC to induce T-cell responses against AML cells in an autologous culture system. Cytotherapy 2006; 8:3-12. [PMID: 16627340 DOI: 10.1080/14653240500499507] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Cells from AML patients can differentiate into the phenotype of DC when cultured with GM-CSF and IL-4. Such cytokine-treated AML-derived DC (AML-DC) can stimulate autologous T cells. In this study we examined whether an anti-CTLA-4 MAb (MDX-010) could enhance the generation of autologous anti-AML T cells. METHODS MAb MDX-010 was added to AML PBMC cultures in the presence of GM-CSF and IL-4, a previously reported AML-DC culture method of generating anti-AML T cells. T-cell activation and proliferation were examined thereafter. RESULTS Addition of MDX-010 to GM-CSF/IL-4-conditioned AML-DC cultures induced a mean seven-fold increase in the numbers of autologous T cells compared with cultures without MDX-010 (P < 0.007). T cells stimulated by AML-DC with CTLA-4 blockade were significantly more cytotoxic towards autologous AML cells than those without MDX-010 (42 +/- 23% vs. 26 +/- 15%, E:T ratio of 20). T cells stimulated by AML-DC with CTLA-4 blockade had significantly greater proportions of T cells producing IFN-gamma in response to autologous AML cells than those cultured with AML-DC alone (10.7 +/- 4.7% vs. 4.5 +/- 2.4% for CD4+ IFN-gamma+ CD69+ and 9.8 +/- 4.1% vs. 4 +/- 2.1% for CD8+ IFN-gamma+ CD69+ with or without MDX-010; n = 7; P < 0.007, P < 0.003, respectively). DISCUSSION CTLA-4 blockade enhances the activity and numbers of AML-reactive T cells that can be stimulated by autologous AML-DC and may enhance the efficacy of adoptive immunotherapy of AML.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antigens, CD
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CTLA-4 Antigen
- Cell Culture Techniques
- Cell Proliferation
- Culture Media, Conditioned
- Cytotoxicity, Immunologic/drug effects
- Dendritic Cells/cytology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Flow Cytometry
- Humans
- Interferon-gamma/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukocytes, Mononuclear/metabolism
- Lymphocyte Activation
- Receptors, Interleukin-2/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- R K Zhong
- Department of Medicine and the Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093-0960, USA
| | | | | | | |
Collapse
|
39
|
McLoone P, Norval M. Adaptation to the UV-induced suppression of phagocytic activity in murine peritoneal macrophages following chronic exposure to solar simulated radiation. Photochem Photobiol Sci 2005; 4:792-7. [PMID: 16189553 DOI: 10.1039/b503094c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Exposure of certain strains of mice to ultraviolet radiation (UVR) is known to suppress both local and systemic immune responses, including a reduction in the phagocytic activity of peritoneal macrophages. However, in many instances, the immunological effects have been observed following a single or a limited number of doses of UVR from sources containing a higher proportion of UVB than that emitted by the sun. The first aim of the present study was to establish whether a single exposure of C3H/HeN mice to solar simulated radiation (SSR) suppressed the ability of the peritoneal macrophages to phagocytose opsonised sheep red blood cells. The mice were irradiated with SSR from Cleo Natural lamps and a single dose of 31.9 J cm(-2) was found to be the minimal dose for significant suppression of macrophage phagocytic activity. Such a dose did not modulate the surface expression of I-A(k), CD11b, CD86 or FcgammaRII/III (CD32/16) on the macrophages. The second aim was to assess whether repeated SSR exposures with a dose below the minimal immunosuppressive dose affected macrophage activity and, if so, to test for photoadaptation by repeated exposures followed by a single, normally immunosuppressive dose of SSR, and then assaying the macrophage activity. Groups of mice were irradiated on each of 2, 10 and 30 days with 14.9 J cm(-2) SSR, followed in some instances by a single additional exposure of 31.9 J cm(-2) on the same day as the last irradiation. The phagocytic activity of the peritoneal macrophages was tested 24 h later. It was reduced by 32%, 18% and 4% respectively after 2, 10 and 30 repeated exposures to SSR, and by 39%, 21% and 7% respectively after 2, 10 and 30 repeated exposures plus the additional higher dose at the end. Thus, although the macrophage activity was initially suppressed by the SSR, photoadaptation of this immune parameter occurred following repeated exposures.
Collapse
Affiliation(s)
- Pauline McLoone
- Medical Microbiology, University of Edinburgh Medical School, Teviot Place, Edinburgh, ScotlandEH8 9AG
| | | |
Collapse
|