1
|
Kumari S, Mukherjee S, Sinha D, Abdisalaam S, Krishnan S, Asaithamby A. Immunomodulatory Effects of Radiotherapy. Int J Mol Sci 2020; 21:E8151. [PMID: 33142765 PMCID: PMC7663574 DOI: 10.3390/ijms21218151] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Radiation therapy (RT), an integral component of curative treatment for many malignancies, can be administered via an increasing array of techniques. In this review, we summarize the properties and application of different types of RT, specifically, conventional therapy with x-rays, stereotactic body RT, and proton and carbon particle therapies. We highlight how low-linear energy transfer (LET) radiation induces simple DNA lesions that are efficiently repaired by cells, whereas high-LET radiation causes complex DNA lesions that are difficult to repair and that ultimately enhance cancer cell killing. Additionally, we discuss the immunogenicity of radiation-induced tumor death, elucidate the molecular mechanisms by which radiation mounts innate and adaptive immune responses and explore strategies by which we can increase the efficacy of these mechanisms. Understanding the mechanisms by which RT modulates immune signaling and the key players involved in modulating the RT-mediated immune response will help to improve therapeutic efficacy and to identify novel immunomodulatory drugs that will benefit cancer patients undergoing targeted RT.
Collapse
Affiliation(s)
- Sharda Kumari
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Shibani Mukherjee
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Debapriya Sinha
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Salim Abdisalaam
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, FL 32224, USA;
| | - Aroumougame Asaithamby
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| |
Collapse
|
2
|
Li S, Chu X, Ye L, Ni J, Zhu Z. A narrative review of synergistic drug administration in unresectable locally advanced non-small cell lung cancer: current landscape and future prospects in the era of immunotherapy. Transl Lung Cancer Res 2020; 9:2082-2096. [PMID: 33209628 PMCID: PMC7653136 DOI: 10.21037/tlcr-20-512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/25/2020] [Indexed: 12/25/2022]
Abstract
Based on the PACIFIC study, the standard care of unresectable locally advanced non-small cell lung cancer (LA-NSCLC) shifted from concurrent chemo-radiotherapy (CCRT) alone to CCRT followed by durvalumab consolidation in 2017. In the era of immunotherapy, two kinds of therapeutic drugs are involved in the management of LA-NSCLC: chemotherapeutics and anti-PD-1/PD-L1 agents. However, the best choices of systematic chemotherapy, immunotherapy, and treatment schedule remain controversial. The immune modulation effects of chemotherapy, as well as the potential immunosuppressive impact of pretreatment medications, should be taken into consideration. Indeed, chemotherapeutics are double-edged swords to immunotherapy, with both stimulatory and suppressive effects on the immune system. Moreover, low-dose chemotherapy is reported to enhance anti-tumor immune responses with reduced toxicities. As for glucocorticoids, there is no consensus about its exact impact on the efficacy of immunotherapy. In addition, the timing of anti-PD-1/PD-L1 agent related to CCRT has three modes: induction, concurrent, and consolidation therapy. Although CCRT followed by durvalumab consolidation is the standard of care, the best sequence of immunotherapy and chemo-radiotherapy is still under debate. Furthermore, the efficacy and toxicity of various PD-1/PD-L1 inhibitors should be compared, especially in the background of CCRT. In this review, we will summarize the detailed knowledge about chemotherapeutics and anti-PD-1/PD-L1 axis agents, and discuss the potential implications in designing novel, effective treatment strategies for LA-NSCLC.
Collapse
Affiliation(s)
- Shuyan Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Luxi Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Cancer Cell Death-Inducing Radiotherapy: Impact on Local Tumour Control, Tumour Cell Proliferation and Induction of Systemic Anti-tumour Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 930:151-72. [PMID: 27558821 DOI: 10.1007/978-3-319-39406-0_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Radiotherapy (RT) predominantly is aimed to induce DNA damage in tumour cells that results in reduction of their clonogenicity and finally in tumour cell death. Adaptation of RT with higher single doses has become necessary and led to a more detailed view on what kind of tumour cell death is induced and which immunological consequences result from it. RT is capable of rendering tumour cells immunogenic by modifying the tumour cell phenotype and the microenvironment. Danger signals are released as well as the senescence-associated secretory phenotype. This results in maturation of dendritic cells and priming of cytotoxic T cells as well as in activation of natural killer cells. However, RT on the other hand can also result in immune suppressive events including apoptosis induction and foster tumour cell proliferation. That's why RT is nowadays increasingly combined with selected immunotherapies.
Collapse
|
4
|
Takahashi J, Misawa M, Iwahashi H. Combined treatment with X-ray irradiation and 5-aminolevulinic acid elicits better transcriptomic response of cell cycle-related factors than X-ray irradiation alone. Int J Radiat Biol 2016; 92:774-789. [PMID: 27586078 DOI: 10.1080/09553002.2016.1230240] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE 5-Aminolevulinic acid (ALA) is a precursor of the photosensitizer protoporphyrin (PpIX) used in photodynamic therapy. In our previous work, PpIX enhanced the generation of reactive oxygen species by X-ray irradiation. In this study, we evaluated the potential of ALA as an endogenous sensitizer to X-ray irradiation. METHODOLOGY Tumor-bearing C57BL/6J mice implanted with B16-BL6 melanoma cells were subsequently treated with irradiation (3 Gy/day for 10 days; total, 30 Gy) plus local administration of 50 mg/kg ALA 24 hours prior to each irradiation (ALA-XT). Tumor-bearing mice without treatment (NT), those treated with ALA only (ALAT), and those treated with X-ray irradiation only (XT) were used as controls. RESULTS ALA potentiated tumor suppression by X-ray irradiation. In microarray analyses using tumor tissue collected after 10 sessions of fractional irradiation, functional analysis revealed that the majority of dysregulated genes in the XT and ALA-XT groups were related to cell-cycle arrest. Finally, the XT and ALA-XT groups differed in the strength of expression, but not in the pattern of expression. CONCLUSIONS mRNA analysis revealed that the combined use of ALA and X-ray irradiation sensitized tumors to X-ray treatment. Furthermore, the present results were consistent with ALA's tumor suppressive effects in vivo.
Collapse
Affiliation(s)
- Junko Takahashi
- a Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology , Tsukuba , Ibaraki , Japan
| | - Masaki Misawa
- b Human Technology Research Institute, National Institute of Advanced Industrial Science and Technology , Tsukuba , Ibaraki , Japan
| | - Hitoshi Iwahashi
- c Faculty of Applied Biological Sciences , Gifu University , Gifu , Japan
| |
Collapse
|
5
|
McPartlin A, Grimaldo C, Lyons J, Burke D, Mitra S, Choudhury A. Successful delivery of chemotherapy to treat small-cell prostate cancer in a patient undergoing haemodialysis. Clin Kidney J 2015; 7:593-4. [PMID: 25859378 PMCID: PMC4389136 DOI: 10.1093/ckj/sfu097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 08/21/2014] [Indexed: 11/17/2022] Open
Abstract
We report on the successful treatment of small-cell prostate cancer in a patient undergoing haemodialysis. The therapeutic regimen included 300 mg/m2 of carboplatin and 50 mg/m2 of etoposide coupled with radical radiotherapy. Adjustments to the patient's haemodialysis prescription included the use of high flux, a larger dialyser surface area and an increased dialysis time. The parameters used aided tolerance to the drug, allowing the delivery of safe, effective treatment. At an interval of over 12 months post-treatment the patient shows no clinical evidence of recurrent disease. This case provides evidence to encourage the use of chemotherapy in otherwise potentially undertreated haemodialysed patients.
Collapse
Affiliation(s)
| | - Claudia Grimaldo
- The Christie NHS Foundation Trust , Manchester , UK ; The University of Manchester , Manchester , UK
| | | | - Daniel Burke
- Central Manchester University Hospitals NHS Foundation Trust , Manchester , UK
| | - Sandip Mitra
- Central Manchester University Hospitals NHS Foundation Trust , Manchester , UK
| | - Ananya Choudhury
- The Christie NHS Foundation Trust , Manchester , UK ; The University of Manchester , Manchester , UK
| |
Collapse
|
6
|
Wang B, Li B, Dai Z, Ren S, Bai M, Wang Z, Li Z, Lin S, Wang Z, Huang N, Yang P, Liu M, Min W, Ma H. Low-dose splenic radiation inhibits liver tumor development of rats through functional changes in CD4+CD25+Treg cells. Int J Biochem Cell Biol 2014; 55:98-108. [PMID: 25168696 DOI: 10.1016/j.biocel.2014.08.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 07/27/2014] [Accepted: 08/17/2014] [Indexed: 02/07/2023]
Abstract
The increased number of CD4(+)CD25(+)Treg cells in tumor local and peripheral splenic tissues is related to the low immune function as well as to tumor recurrence and metastasis. Our pre-clinical studies showed that low-dose radiation (LDR) of the spleen in liver cancer patients significantly improves immune functions. However, the molecular mechanisms of such radiation remained ill defined. This study explores the role of CD4(+)CD25(+)Treg cells in radiation-induced immunomodulatory effects. Using the diethylnitrosamine (DEN)-induced rat liver tumor model and in vitro cell experiments, the percentage of CD4(+)CD25(+)Treg/CD4(+) cells in the blood and the expressions of Foxp3(+), IL-10, TGF-β, and cytotoxic T lymphocyte-associated antigen-4(CTLA-4) in spleen and liver tumors significantly decreased after LDR of the spleen in rats with liver cancer. The tumors became smaller than those in the non-radiated group, with both showing a parallel relation. Flow cytometry and MTT results revealed that LDR failed to inhibit CD4(+)CD25(+)Treg cell proliferation. Conversely, apoptosis was reduced and proliferation was stimulated. This process also changed CTLA-4 molecule expression on the surfaces of CD4(+)CD25(+)Treg cells and reduced their inhibitory function against CD4(+)CD25(-)T cell proliferation, and the suppression function of CD4(+)CD25(+)Treg cells was further weakened with the introduction of the CTLA-4 inhibitor. Findings demonstrate that the reduction of CTLA-4 expression on the CD4(+)CD25(+)Treg cell surface and the further inhibition of cell function may be considered as important regulators of LDR-induced immunomodulatory effects. This study provides experimental evidence to elucidate the immune enhancement induced by this process and presents a novel method for liver cancer immunotherapy.
Collapse
Affiliation(s)
- Baofeng Wang
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Baohua Li
- Department of Surgery, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Song Ren
- Department of Surgery, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Minghua Bai
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Zhongwei Wang
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Zongfang Li
- Department of Surgery, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Zhidong Wang
- Department of Surgery, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Na Huang
- Department of Surgery, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Pengtao Yang
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Mengjie Liu
- Department of Oncology, The First Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Weili Min
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| | - Hongbing Ma
- Department of Oncology, The Second Affiliated Hospital of School of Medicine, Xi'an Jiaotong University, Xi'an 710004, PR China.
| |
Collapse
|
7
|
Aryankalayil MJ, Makinde AY, Gameiro SR, Hodge JW, Rivera-Solis PP, Palayoor ST, Ahmed MM, Coleman CN. Defining molecular signature of pro-immunogenic radiotherapy targets in human prostate cancer cells. Radiat Res 2014; 182:139-48. [PMID: 25003313 DOI: 10.1667/rr13731.1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
To understand the impact of clinically relevant radiation therapy (RT) on tumor immune gene expression and to utilize the changes that occur during treatment to improve cancer treatment outcome, we examined how immune response genes are modulated in prostate cancer cells of varying p53 status. LNCaP (p53 wild-type), PC3 (p53 null) and DU145 (p53 mutant) cells received a 10 Gy single dose or 1 Gy × 10 multifractionated radiation dose to simulate hypofractionated and conventionally fractionated prostate radiotherapy. Total RNA was isolated 24 h after multifractionated radiation treatment and single-dose treatments and subjected to microarray analysis and later validated by RT-PCR. RT-PCR was utilized to identify total-dose inflection points for significantly upregulated genes in response to multifractionated radiation therapy. Radiation-induced damage-associated molecular pattern molecules (DAMPs) and cytokine analyses were performed using bioluminescence and ELISA. Multifractionated doses activated immune response genes more robustly than single-dose treatment, with a relatively larger number of immune genes upregulated in PC3 compared to DU145 and LNCaP cells. The inflection point of multifractionated radiation-induced immune genes in PC3 cells was observed in the range of 8-10 Gy total radiation dose. Although both multifractionated and single-dose radiation-induced proinflammatory DAMPs and positively modulated the cytokine environment, the changes were of higher magnitude with multifractionated therapy. The findings of this study together with the gene expression data suggest that cells subjected to multifractionated radiation treatment would promote productive immune cell-tumor cell interactions.
Collapse
Affiliation(s)
- Molykutty J Aryankalayil
- a Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Kulzer L, Rubner Y, Deloch L, Allgäuer A, Frey B, Fietkau R, Dörrie J, Schaft N, Gaipl US. Norm- and hypo-fractionated radiotherapy is capable of activating human dendritic cells. J Immunotoxicol 2014; 11:328-36. [PMID: 24512329 DOI: 10.3109/1547691x.2014.880533] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite the transient immunosuppressive properties of local radiotherapy (RT), this classical treatment modality of solid tumors is capable of inducing immunostimulatory forms of tumor-cell death. The resulting 'immunotoxicity' in the tumor, but not in healthy tissues, may finally lead to immune-mediated destruction of the tumor. However, little is known about the best irradiation scheme in this setting. This study examines the immunological effects of differently irradiated human colorectal tumor cells on human monocyte-derived dendritic cells (DC). Human SW480 tumor cells were irradiated with a norm-fractionation scheme (5 × 2 Gy), a hypo-fractionated protocol (3 × 5 Gy), and with a high single irradiation dose (radiosurgery; 1 × 15 Gy). Subsequently, human immature DC (iDC) were co-incubated with supernatants (SN) of these differently treated tumor cells. Afterwards, DC were analyzed regarding the expression of maturation markers, the release of cytokines, and the potential to stimulate CD4(+) T-cells. The co-incubation of iDC with SN of tumor cells exposed to norm- or hypo-fractionated RT resulted in a significantly increased secretion of the immune activating cytokines IL-12p70, IL-8, IL-6, and TNFα, compared to iDC co-incubated with SN of tumor cells that received a high single irradiation dose or were not irradiated. In addition, DC-maturation markers CD80, CD83, and CD25 were also exclusively elevated after co-incubation with the SN of fractionated irradiated tumor cells. Furthermore, the SN of tumor cells that were irradiated with norm- or hypo-fractionated RT triggered iDC to stimulate CD4(+) T-cells not only in an allogenic, but also in an antigen-specific manner like mature DC. Collectively, these results demonstrate that norm- and hypo-fractionated RT induces a fast human colorectal tumor-cell death with immunogenic potential that can trigger DC maturation and activation in vitro. Such findings may contribute to the improvement of irradiation protocols for the most beneficial induction of anti-tumor immunity.
Collapse
|
9
|
Venkataramanaa NK, Venkatesh PK, Dwarakanath BS, Vani S. Protective effect on normal brain tissue during a combinational therapy of 2-deoxy-d-glucose and hypofractionated irradiation in malignant gliomas. Asian J Neurosurg 2013; 8:9-14. [PMID: 23741257 PMCID: PMC3667464 DOI: 10.4103/1793-5482.110274] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Purpose: To investigate the effect of 2-deoxy–D-glucose (2-DG), an inhibitor of glucose transport and glycolysis, on glioblastoma and the normal brain tissue during combined treatment with hypofractionated radiotherapy. Materials and Methods: Twenty patients with malignant gliomas (18 Glioblastoma Multiformae, 2 Anasplastic Astrocytoma grade III) following surgery were treated weekly (once) with 2-DG, (250 mg/kg body weight), followed by 5 Gy of radiation to the tumor bed per fraction for 7 weeks. Clinical evaluation, complete hemogram, and random blood sugar levels were carried out in each cycle. Follow-up computed tomography (CT)/magnetic resonance imaging (MRI) was done to evaluate radiation-induced changes. Kernofsky Performance scale (KPS) was recorded preoperatively; postoperatively, and post-therapy till the last follow-up. Results: Twenty patients were recruited for this trail; 19 of them completed the treatment and 1 discontinued. The survival period ranged between 6 and 36 months after the treatment, with a median survival of 14 months. CT and MRI revealed significant tumor necrosis. Histological evidence from the tissue during reexploration confirms the hypothesis of protective effect of 2-DG on normal brain. KPS was above 80% in majority of the patients, 6 months after the surgery. Conclusion: Radiotherapy coupled with 2-DG enhances tumor necrosis selectively and significantly while the normal brain gets relatively protected. This has been reflected in our study both clinically by preservation of quality-of-life and pathologically by retaining the integrity of normal brain architecture.
Collapse
|
10
|
Schoenfeld JD, Margalit DN, Kasperzyk JL, Shui IM, Rider JR, Epstein MM, Meisner A, Kenfield SA, Martin NE, Nguyen PL, Kantoff PW, Giovannucci EL, Stampfer MJ, Mucci LA. A single nucleotide polymorphism in inflammatory gene RNASEL predicts outcome after radiation therapy for localized prostate cancer. Clin Cancer Res 2013; 19:1612-9. [PMID: 23382116 PMCID: PMC3602407 DOI: 10.1158/1078-0432.ccr-12-2718] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE To study associations between single nucleotide polymorphisms (SNP) in Ribonuclease L (RNASEL), a gene implicated in inflammation and prostate cancer risk, and outcomes after radiation therapy. EXPERIMENTAL DESIGN We followed participants in the prospective US Health Professionals Follow-Up Study treated with radiation therapy for early-stage prostate cancer. Three SNPs were genotyped based on previously determined functional and biological significance. We used multivariable Cox proportional hazards models to assess per-allele associations with the primary outcome defined as time to a composite endpoint including development of lethal prostate cancer or biochemical recurrence. RESULTS We followed 434 patients treated with radiation therapy for a median of 9 years. On multivariate analysis, the rs12757998 variant allele was associated with significantly decreased risk of the composite endpoint [HR: 0.65; 95% confidence interval (CI), 0.45-0.94%; P = 0.02] driven by decreased biochemical recurrence (HR: 0.60; 95% CI, 0.40-0.89%; P = 0.01) and men treated with external beam (HR: 0.58; 95% CI, 0.36-0.93%; P = 0.02). In contrast, in 516 men from the same cohort treated with radical prostatectomy, we found no significant impact of this variant on outcome. Furthermore, the rs12757998 variant allele significantly modified the association between androgen deprivation therapy and outcomes after radiation therapy (p-interaction = 0.02). CONCLUSION We show an association between RNASEL SNP rs12757998 and outcome after radiation therapy for prostate cancer. This SNP is associated with increased circulating C-reactive protein and interleukin-6, suggesting a potential role for inflammation in the response to radiation. If validated, genetic predictors of outcome may help inform prostate cancer management.
Collapse
|
11
|
Lourenço J, Pereira R, Pinto F, Caetano T, Silva A, Carvalheiro T, Guimarães A, Gonçalves F, Paiva A, Mendo S. Biomonitoring a human population inhabiting nearby a deactivated uranium mine. Toxicology 2013; 305:89-98. [PMID: 23370006 DOI: 10.1016/j.tox.2013.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 01/21/2013] [Accepted: 01/21/2013] [Indexed: 12/21/2022]
Abstract
Environmental exposure to uranium and its daughter radionuclides, has been linked to several negative effects such as those related with important physiological processes, like hematopoiesis, and may also be associated with genotoxicity effects. Herein, genotoxic effects, immunotoxicity, trace elements and C reactive protein (CRP) analyses, were performed in peripheral blood samples collected from individuals of a population living near a deactivated uranium mine. C reactive protein analysis was performed to exclude candidates with active inflammatory processes from further evaluations. DNA damage and immunotoxicity (immunophenotyping and immune cell counts) were evaluated by comet assay and flow cytometry, respectively. Significant DNA damage was observed in the peripheral blood samples from volunteers living in the Cunha Baixa village. A significant decrease of NK and T lymphocytes counts were observed in the individuals from the Cunha Baixa village, when compared with individuals from the reference site. Uranium and manganese levels were significantly higher in the Cunha Baixa village inhabitants. On the other hand, zinc levels were significantly lower in those individuals when compared with the volunteers from the control village. Results suggest that inhabitants from Cunha Baixa have a higher risk of suffering from serious diseases such as cancer, since high DNA damages were observed in peripheral blood leukocytes and also decreased levels of NK and T cells, which play an essential role in the defense against tumor growth.
Collapse
Affiliation(s)
- J Lourenço
- Departamento de Biologia, Universidade de Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Farooque A, Mathur R, Verma A, Kaul V, Bhatt AN, Adhikari JS, Afrin F, Singh S, Dwarakanath BS. Low-dose radiation therapy of cancer: role of immune enhancement. Expert Rev Anticancer Ther 2011; 11:791-802. [PMID: 21554054 DOI: 10.1586/era.10.217] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The efficacy of conventional radiation therapy, one of the most widely used treatment modalities of cancer, is limited by resistance of tumors as well as normal tissue toxicity. In the last decade, several studies have shown that protocols using low-dose radiation (LDR) are more effective in providing local tumor control with negligible normal tissue toxicity. LDR stimulates antioxidant capacity, repair of DNA damage, apoptosis and induction of immune responses, which might be collectively responsible for providing effective local tumor control. This article focuses on the immunostimulatory effects of LDR in in vivo models and its clinical efficacy, supporting the use of LDR regimens (alone or as adjuvant) as an anticancer treatment.
Collapse
Affiliation(s)
- Abdullah Farooque
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Road, Delhi 110 0054, India
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
|
14
|
Ad.Egr-TNF and local ionizing radiation suppress metastases by interferon-beta-dependent activation of antigen-specific CD8+ T cells. Mol Ther 2010; 18:912-20. [PMID: 20197756 DOI: 10.1038/mt.2010.18] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Ad.Egr-TNF is a radioinducible adenovector currently in phase 3 trials for inoperable pancreatic cancer. The combination of Ad.Egr-TNF and ionizing radiation (IR) contributes to local tumor control through the production of tumor necrosis factor-alpha (TNFalpha) in the tumor microenvironment. Moreover, clinical and preclinical studies with Ad.Egr-TNF/IR have suggested that this local approach suppresses the growth of distant metastatic disease; however, the mechanisms responsible for this effect remain unclear. These studies have been performed in wild-type (WT) and TNFR1,2(-/-) mice to assess the role of TNFalpha-induced signaling in the suppression of draining lymph node (DLN) metastases. The results demonstrate that production of TNFalpha in the tumor microenvironment induces expression of interferon (IFNbeta). In turn, IFNbeta stimulates the production of chemokines that recruit CD8(+) T cells to the tumor. The results further demonstrate that activation of tumor antigen-specific CD8(+) CTLs contributes to local antitumor activity and suppression of DLN metastases. These findings support a model in which treatment of tumors with Ad.Egr-TNF and IR is mediated by local and distant immune-mediated antitumor effects that suppress the development of metastases.
Collapse
|
15
|
Mann DL, Celluzzi CM, Hankey KG, Harris KM, Watanabe R, Hasumi K. Combining conventional therapies with intratumoral injection of autologous dendritic cells and activated T cells to treat patients with advanced cancers. Ann N Y Acad Sci 2009; 1174:41-50. [PMID: 19769735 DOI: 10.1111/j.1749-6632.2009.04934.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that have been used in cancer immunotherapy. To take advantage of the ability of DCs to acquire antigenic materials from their environment and generate primary as well as recall immune responses, 37 patients with advanced cancers were enrolled in a series of protocols based on direct intratumoral injection of immature DCs. To augment antigen uptake and antitumor immune response, DC injection was combined with radiotherapy or chemotherapy and/or injection of activated T cells. Treatments were well tolerated with no adverse reactions. Clinical responses were based on Response Evaluation Criteria in Solid Tumors, with the majority of patients showing stable disease. One of two patients who also received local radiation achieved a sustained complete response at injected and metastatic sites. The clinical responses observed in cancer patients with advanced disease suggest potential effectiveness of combination strategies and establish the basis for the current treatment protocol that is underway.
Collapse
Affiliation(s)
- Dean L Mann
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Optimization of dendritic cell loading with tumor cell lysates for cancer immunotherapy. J Immunother 2009; 31:620-32. [PMID: 18600182 DOI: 10.1097/cji.0b013e31818213df] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The immune response to cancer is critically determined by the way in which tumor cells die. As necrotic, stress-associated death can be associated with activation of antitumor immunity, whole tumor cell antigen loading strategies for dendritic cell (DC)-based vaccination have commonly used freeze-thaw "necrotic" lysates as an immunogenic source of tumor-associated antigens. In this study, the effect of such lysates on the ability of DCs to mature in response to well-established maturation stimuli was examined, and methods to enhance lysate-induced DC activation explored. Freeze-thaw lysates were prepared from murine tumor cell lines and their effects on bone marrow-derived DC maturation and function examined. Unmodified freeze-thaw tumor cell lysates inhibited the toll-like receptor-induced maturation and function of bone marrow-derived DCs, preventing up-regulation of CD40, CD86, and major histocompatibility complex class II, and reducing secretion of inflammatory cytokines [interleukin (IL)-12 p70, tumor necrosis factor-alpha, and IL-6]. Although IL-10 secretion was increased by lysate-pulsed DCs, this was not responsible for the observed suppression of IL-12. Although activation of the nuclear factor-kappaB pathway remained intact, the kinase activity of phosphorylated p38 mitogen-activated protein kinase was inhibited in lysate-pulsed DCs. Lysate-induced DC suppression was partially reversed in vitro by induction of tumor cell stress before lysis, and only DCs loaded with stressed lysates afforded protection against tumor challenge in vivo. These data suggest that ex vivo freeze-thaw of tumor cells does not effectively mimic in vivo immunogenic necrosis, and advocates careful characterization and optimization of tumor cell-derived vaccine sources for cancer immunotherapy.
Collapse
|
17
|
Optimization of dendritic cell loading with tumor cell lysates for cancer immunotherapy. JOURNAL OF IMMUNOTHERAPY (HAGERSTOWN, MD. : 1997) 2009. [PMID: 18600182 DOI: 10.1097/cji.0b013e31818213df00002371-200809000-00003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The immune response to cancer is critically determined by the way in which tumor cells die. As necrotic, stress-associated death can be associated with activation of antitumor immunity, whole tumor cell antigen loading strategies for dendritic cell (DC)-based vaccination have commonly used freeze-thaw "necrotic" lysates as an immunogenic source of tumor-associated antigens. In this study, the effect of such lysates on the ability of DCs to mature in response to well-established maturation stimuli was examined, and methods to enhance lysate-induced DC activation explored. Freeze-thaw lysates were prepared from murine tumor cell lines and their effects on bone marrow-derived DC maturation and function examined. Unmodified freeze-thaw tumor cell lysates inhibited the toll-like receptor-induced maturation and function of bone marrow-derived DCs, preventing up-regulation of CD40, CD86, and major histocompatibility complex class II, and reducing secretion of inflammatory cytokines [interleukin (IL)-12 p70, tumor necrosis factor-alpha, and IL-6]. Although IL-10 secretion was increased by lysate-pulsed DCs, this was not responsible for the observed suppression of IL-12. Although activation of the nuclear factor-kappaB pathway remained intact, the kinase activity of phosphorylated p38 mitogen-activated protein kinase was inhibited in lysate-pulsed DCs. Lysate-induced DC suppression was partially reversed in vitro by induction of tumor cell stress before lysis, and only DCs loaded with stressed lysates afforded protection against tumor challenge in vivo. These data suggest that ex vivo freeze-thaw of tumor cells does not effectively mimic in vivo immunogenic necrosis, and advocates careful characterization and optimization of tumor cell-derived vaccine sources for cancer immunotherapy.
Collapse
|
18
|
Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood 2009; 114:589-95. [PMID: 19349616 DOI: 10.1182/blood-2009-02-206870] [Citation(s) in RCA: 1030] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patients with locally advanced cancer or distant metastasis frequently receive prolonged treatment with chemotherapy and/or fractionated radiotherapy (RT). Despite the initial clinical response, treatment resistance frequently develops and cure in these patients is uncommon. Developments in RT technology allow for the use of high-dose (or ablative) RT to target local tumors, with limited damage to the surrounding normal tissue. We report that reduction of tumor burden after ablative RT depends largely on T-cell responses. Ablative RT dramatically increases T-cell priming in draining lymphoid tissues, leading to reduction/eradication of the primary tumor or distant metastasis in a CD8(+) T cell-dependent fashion. We further demonstrate that ablative RT-initiated immune responses and tumor reduction are abrogated by conventional fractionated RT or adjuvant chemotherapy but greatly amplified by local immunotherapy. Our study challenges the rationale for current RT/chemotherapy strategies and highlights the importance of immune activation in preventing tumor relapse. Our findings emphasize the need for new strategies that not only reduce tumor burden but also enhance the role of antitumor immunity.
Collapse
|
19
|
Deacon DH, Hogan KT, Swanson EM, Chianese-Bullock KA, Denlinger CE, Czarkowski AR, Schrecengost RS, Patterson JW, Teague MW, Slingluff CL. The use of gamma-irradiation and ultraviolet-irradiation in the preparation of human melanoma cells for use in autologous whole-cell vaccines. BMC Cancer 2008; 8:360. [PMID: 19055839 PMCID: PMC2612687 DOI: 10.1186/1471-2407-8-360] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Accepted: 12/04/2008] [Indexed: 11/10/2022] Open
Abstract
Background Human cancer vaccines incorporating autologous tumor cells carry a risk of implantation and subsequent metastasis of viable tumor cells into the patient who is being treated. Despite the fact that the melanoma cell preparations used in a recent vaccine trial (Mel37) were gamma-irradiated (200 Gy), approximately 25% of the preparations failed quality control release criteria which required that the irradiated cells incorporate 3H-thymidine at no more than 5% the level seen in the non-irradiated cells. We have, therefore, investigated ultraviolet (UV)-irradiation as a possible adjunct to, or replacement for gamma-irradiation. Methods Melanoma cells were gamma- and/or UV-irradiated. 3H-thymidine uptake was used to assess proliferation of the treated and untreated cells. Caspase-3 activity and DNA fragmentation were measured as indicators of apoptosis. Immunohistochemistry and Western blot analysis was used to assess antigen expression. Results UV-irradiation, either alone or in combination with gamma-irradiation, proved to be extremely effective in controlling the proliferation of melanoma cells. In contrast to gamma-irradiation, UV-irradiation was also capable of inducing significant levels of apoptosis. UV-irradiation, but not gamma-irradiation, was associated with the loss of tyrosinase expression. Neither form of radiation affected the expression of gp100, MART-1/MelanA, or S100. Conclusion These results indicate that UV-irradiation may increase the safety of autologous melanoma vaccines, although it may do so at the expense of altering the antigenic profile of the irradiated tumor cells.
Collapse
Affiliation(s)
- Donna H Deacon
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Melanoma and lymphoma rejection associated with eosinophil infiltration upon intratumoral injection of dendritic and NK/LAK cells. J Immunother 2008; 31:458-65. [PMID: 18463539 DOI: 10.1097/cji.0b013e318174a512] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dendritic cells (DCs) are promising tools for tumor immunotherapy. Their efficacy in the tumor environment increases when tumor cells die as a consequence of chemo/radiotherapy or when local stimuli promoting DC maturation and function are available. Dying tumor cells could represent a source of tumor antigens, which DCs cross-present to tumor-specific T cells. The outcome of cross presentation is in turn determined by the maturation state of DCs. Natural killer (NK)/lymphokine-activated killer (LAK) cells injected into growing tumors could both provide a source of dying cells for cross-presentation and deliver stimuli for DC maturation. Here, we report that NK/LAK cells recognized and killed in vivo major histocompatibility complex class I(low) highly tumorigenic, nonimmunogenic B16F1 melanoma cells when injected into exponentially growing neoplastic lesions. The simultaneous injection of immature DCs was required to heal animals. Similar results were obtained injecting NK/LAK cells and DC into growing Raucher leukaemia virus induced cell line lymphomas. Cured mice failed to reject other implantable tumors, and developed a specific cytotoxic response against the original neoplasm; moreover, they developed a long-lasting memory, and were protected against further challenges with living tumor cells only when both cell populations were introduced. The response associated to the preferential recruitment within tumors of eosinophils. The simultaneous injection in solid tumors of DCs and NK/LAK cells represents an attractive approach for antineoplastic immunotherapeutic strategies.
Collapse
|
21
|
|
22
|
Baluna RG, Eng TY, Thomas CR. Adhesion molecules in radiotherapy. Radiat Res 2007; 166:819-31. [PMID: 17149971 DOI: 10.1667/rr0380.1] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2005] [Accepted: 06/27/2006] [Indexed: 11/03/2022]
Abstract
Recent studies have documented changes in adhesion molecule expression and function after exposure to ionizing radiation. Adhesion molecules mediate cell-cell and cell-matrix interactions and are essential for a variety of physiological and pathological processes including maintenance of normal tissue integrity as well as tumor development and progression. Consequently, modulation of adhesion molecules by radiation may have a role in radiation-induced tumor control and normal tissue damage by interfering with cell signaling, radioresistance, metastasis, angiogenesis, carcinogenesis, immune response, inflammation and fibrosis. In addition, the interactions of radiation with adhesion molecules could have a major impact in developing new strategies to increase the efficacy of radiation therapy. Remarkable progress has been made in recent years to design targeted drug delivery to radiation-up-regulated adhesion molecules. Furthermore, the inhibition of adhesion, migration, invasion and angiogenesis by blocking adhesion receptors may represent a new therapeutic approach to improve tumor control and decrease radiation toxicity. This review is focused on current data concerning the mechanistic interactions of radiation with adhesion molecules and the possible clinical-pathological implications in radiotherapy.
Collapse
Affiliation(s)
- Roxana G Baluna
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | |
Collapse
|
23
|
Thamm DH. Interactions between radiation therapy and immunotherapy: the best of two worlds? Vet Comp Oncol 2006; 4:189-97. [DOI: 10.1111/j.1476-5829.2006.00110.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
24
|
Capobianco A, Rovere-Querini P, Rugarli C, Manfredi AA. Melanoma cells interfere with the interaction of dendritic cells with NK/LAK cells. Int J Cancer 2006; 119:2861-9. [PMID: 16998790 DOI: 10.1002/ijc.22279] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Dendritic cells (DCs) and natural killer (NK) cells are key players at the interface between innate resistance and acquired immunity. NK cells can induce DC maturation, a differentiation process whereby DCs respond to a environmental stimulus and acquire the ability of eliciting adaptive immunity. Conversely, maturing DCs promote NK functions in vivo and in vitro. This interplay has important consequences on the immune response to pathogens and possibly to neoplastic cells. Here, we show that B16 melanoma cells actively modulate the interaction between DCs derived from bone marrow precursors and NK/LAK cells propagated from the spleen of C57BL/6 mice. DCs increased in a dose-dependent manner the ability of NK/LAK cells to kill melanoma cells and to produce cytokines. This activatory cross-talk entailed the production of IL-18 by DCs and of IFN-gamma by NK/LAK cells. Melanoma cells were not a passive target of NK activity; they regulated the outcome of the interaction between DCs and NK/LAK cells, inhibiting the in vitro production of cytokines as effectively as the genetic deletion of IL-18 or IFN-gamma. Interference with the NK/DC interaction possibly represents a mechanism used by growing tumors to evade the immune response.
Collapse
MESH Headings
- Animals
- Cell Communication
- Cell Line, Tumor
- Coculture Techniques
- Cytotoxicity Tests, Immunologic
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Female
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interleukin-18/genetics
- Interleukin-18/metabolism
- Killer Cells, Lymphokine-Activated/cytology
- Killer Cells, Lymphokine-Activated/immunology
- Killer Cells, Lymphokine-Activated/metabolism
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Male
- Melanoma/immunology
- Melanoma/metabolism
- Melanoma/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Time Factors
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Annalisa Capobianco
- Department of Oncology, Cancer Immunotherapy and Gene Therapy Program, Clinical Immunology Unit, H San Raffaele Scientific Institute & Vita-Salute San Raffaele University, Milano, Italy
| | | | | | | |
Collapse
|