Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.531
Filter
1.
Nat Commun ; 15(1): 5053, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871684

ABSTRACT

Childhood radioactive iodine exposure from the Chornobyl accident increased papillary thyroid carcinoma (PTC) risk. While cervical lymph node metastases (cLNM) are well-recognized in pediatric PTC, the PTC metastatic process and potential radiation association are poorly understood. Here, we analyze cLNM occurrence among 428 PTC with genomic landscape analyses and known drivers (131I-exposed = 349, unexposed = 79; mean age = 27.9 years). We show that cLNM are more frequent in PTC with fusion (55%) versus mutation (30%) drivers, although the proportion varies by specific driver gene (RET-fusion = 71%, BRAF-mutation = 38%, RAS-mutation = 5%). cLNM frequency is not associated with other characteristics, including radiation dose. cLNM molecular profiling (N = 47) demonstrates 100% driver concordance with matched primary PTCs and highly concordant mutational spectra. Transcriptome analysis reveals 17 differentially expressed genes, particularly in the HOXC cluster and BRINP3; the strongest differentially expressed microRNA also is near HOXC10. Our findings underscore the critical role of driver alterations and provide promising candidates for elucidating the biological underpinnings of PTC cLNM.


Subject(s)
Chernobyl Nuclear Accident , Iodine Radioisotopes , Lymphatic Metastasis , Mutation , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Lymphatic Metastasis/genetics , Male , Adult , Female , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Adolescent , Proto-Oncogene Proteins B-raf/genetics , Young Adult , Lymph Nodes/pathology , Proto-Oncogene Proteins c-ret/genetics , Child , Genomics , Middle Aged , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Gene Expression Profiling , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Neck/pathology , Gene Expression Regulation, Neoplastic
2.
Int J Radiat Biol ; 100(6): 903-911, 2024.
Article in English | MEDLINE | ID: mdl-38652836

ABSTRACT

PURPOSE: The effect of chronic low dose-rate radiation exposure on cancers was investigated by analyzing the data of mice experiments conducted at the Institute for Environmental Sciences (IES). This analysis focuses on the differences between malignant lymphomas and solid cancers. MATERIALS AND METHODS: The analysis is conducted based on the mathematical model introduced in our previous work. The model is expanded to analyze malignant lymphomas and solid cancers separately. Using the expanded model, the effect of chronic low dose-rate radiation on malignant lymphomas and solid cancers are discussed based on their occurrences, progressions, and mortalities. RESULTS: Non-irradiated control group and 20 mGy/day × 400 days irradiated groups are analyzed. The analysis showed that radiation exposure shortened mean life expectancy for both malignant lymphomas and solid cancers (shorter by 89.6 days for malignant lymphomas and 149.3 days for solid cancers). For malignant lymphomas, both the occurrence and the progression are affected by radiation exposure. The mean age at which malignant lymphoma developed in mice was shortened by 32.7 days and the mean progression period was shortened by 57.3 days. The occurrence of solid cancer is also affected by radiation exposure, wherein the mean age at which solid cancer develops was shortened by 147.9 days. However, no significant change in progression period of solid cancers was seen in the analysis. CONCLUSIONS: The analysis showed that the occurrence and mean lifespan are affected in both malignant lymphomas and solid cancers. The shortening of the progression period is only seen in malignant lymphoma, no significant change was observed in solid cancers.


Subject(s)
Dose-Response Relationship, Radiation , Lymphoma , Neoplasms, Radiation-Induced , Animals , Mice , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Lymphoma/etiology , Lymphoma/pathology , Neoplasms/radiotherapy , Neoplasms/pathology , Neoplasms/etiology , Radiation Exposure/adverse effects , Radiation Dosage , Female , Male
3.
Life Sci Space Res (Amst) ; 41: 202-209, 2024 May.
Article in English | MEDLINE | ID: mdl-38670648

ABSTRACT

Explorations of the Moon and Mars are planned as future manned space missions, during which humans will be exposed to both radiation and microgravity. We do not, however, know the health effects for such combined exposures. In a ground-based experiment, we evaluated the combined effects of radiation and simulated microgravity on tumorigenesis by performing X-irradiation and tail suspension in C3B6F1 ApcMin/+ mice, a well-established model for intestinal tumorigenesis. Mice were irradiated at 2 weeks of age and underwent tail suspension for 3 or 11 weeks using a special device that avoids damage to the tail. The tail suspension treatment significantly reduced the thymus weight after 3 weeks but not 11 weeks, suggesting a transient stress response. The combination of irradiation and tail suspension significantly increased the number of small intestinal tumors less than 2 mm in diameter as compared with either treatment alone. The combined treatment also increased the fraction of malignant tumors among all small intestinal tumors as compared with the radiation-only treatment. Thus, the C3B6F1 ApcMin/+ mouse is a useful model for assessing cancer risk in a simulated space environment, in which simulated microgravity accelerates tumor progression when combined with radiation exposure.


Subject(s)
Intestinal Neoplasms , Weightlessness Simulation , Animals , Mice , Intestinal Neoplasms/pathology , Intestinal Neoplasms/etiology , Carcinogenesis/radiation effects , Mice, Inbred C57BL , Hindlimb Suspension , Male , X-Rays , Disease Models, Animal , Female , Intestine, Small/radiation effects , Intestine, Small/pathology , Thymus Gland/radiation effects , Thymus Gland/pathology , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/etiology
4.
Life Sci Space Res (Amst) ; 41: 43-51, 2024 May.
Article in English | MEDLINE | ID: mdl-38670651

ABSTRACT

Prolonged manned space flight exposure risks to galactic comic radiation, has led to uncertainties in a variety of health risks. Our previous work, utilizing either single ion or multiple ion radiation exposure conducted at the NSRL (NASA Space Radiation Laboratory, Brookhaven, NY) demonstrated that HZE ion components of the GCR result in persistent inflammatory signaling, increased mutations, and higher rates of cancer initiation and progression. With the development of the 33-beam galactic cosmic radiation simulations (GCRsim) at the NSRL, we can more closely test on earth the radiation environment found in space. With a previously used lung cancer susceptible mouse model (K-rasLA-1), we performed acute exposure experiments lasting 1-2 h, and chronic exposure experiments lasting 2-6 weeks with a total dose of 50 cGy and 75 cGy. We obtained histological samples from a subset of mice 100 days post-irradiation, and the remaining mice were monitored for overall survival up to 1-year post-irradiation. When we compared acute exposures (1-2 hrs.) and chronic exposure (2-6 weeks), we found a trend in the increase of lung adenocarcinoma respectively for a total dose of 50 cGy and 75 cGy. Furthermore, when we added neutron exposure to the 75 cGy of GCRsim, we saw a further increase in the incidence of adenocarcinoma. We interpret these findings to suggest that the risks of carcinogenesis are heightened with doses anticipated during a round trip to Mars, and this risk is magnified when coupled with extra neutron exposure that are expected on the Martian surface. We also observed that risks are reduced when the NASA official 33-beam GCR simulations are provided at high dose rates compared to low dose rates.


Subject(s)
Cosmic Radiation , Disease Progression , Lung Neoplasms , Neoplasms, Radiation-Induced , Animals , Cosmic Radiation/adverse effects , Mice , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Space Flight , Female , Male
5.
Ann Pathol ; 44(3): 210-214, 2024 May.
Article in French | MEDLINE | ID: mdl-38448265

ABSTRACT

INTRODUCTION: First case of radiation-induced parotid leiomyosarcoma. ANATOMO-CLINICAL OBSERVATION: A 50-year-old woman with a history of cervical irradiation for Hodgkin's lymphoma presented with a right parotid tumefaction. Examination noted a deep adherent pretragal mass with peripheral facial palsy. A total parotidectomy with intra-operative examination and cervical curage was performed. Histopathological analysis concluded to a grade 3 parotid leiomyosarcoma according to the National Federation of Cancer Centers. Adjuvant radiotherapy was performed. After 24 months of follow-up, the patient presented bone and liver metastases without local recurrence. DISCUSSION: This is the first case of radiation-induced leiomyosarcoma and the 12th case of parotid leiomyosarcoma described in the literature. The management associates surgery with adjuvant radiotherapy. Follow-up is by clinical examination, parotid MRI, and annual thoracoabdominal CT scan to search for metastases. Recurrences occur during the first year in 40 to 64% of cases, and distant metastases in 40 to 60% of cases. The 5-year survival rate is between 10 and 30%.


Subject(s)
Leiomyosarcoma , Neoplasms, Radiation-Induced , Parotid Neoplasms , Humans , Leiomyosarcoma/pathology , Leiomyosarcoma/secondary , Parotid Neoplasms/pathology , Parotid Neoplasms/secondary , Parotid Neoplasms/radiotherapy , Middle Aged , Female , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/etiology , Hodgkin Disease/radiotherapy , Hodgkin Disease/pathology , Radiotherapy, Adjuvant
6.
Laryngoscope ; 134(7): 3143-3145, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38459959

ABSTRACT

Keloid scars can cause significant morbidity to the patient including substantial cosmetic disfigurement, particularly in the head and neck region. Surgical excision followed by immediate postoperative radiation therapy has been shown to be more effective than single modality treatment. Radiation therapy increases risk for radiation-induced malignancy, though very few cases have been reported in the treatment of keloids. We report the case of a patient with a large postauricular keloid treated with excision and immediate post-operative radiation who developed a high-grade pleomorphic sarcoma in the scar bed. Laryngoscope, 134:3143-3145, 2024.


Subject(s)
Keloid , Humans , Keloid/etiology , Keloid/surgery , Keloid/radiotherapy , Male , Neoplasms, Radiation-Induced/surgery , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Sarcoma/radiotherapy , Sarcoma/surgery , Sarcoma/etiology , Female , Middle Aged
7.
Int J Radiat Biol ; 100(2): 176-182, 2024.
Article in English | MEDLINE | ID: mdl-37755376

ABSTRACT

PURPOSE: In this paper, we described our mathematical model for radiation-induced life shortening in detail and applied the model to the experimental data on mice to investigate the effect of radiation on cancer-related life-shortening. MATERIALS AND METHODS: Our mathematical model incorporates the following components: (i) occurrence of cancer, (ii) progression of cancer over time, and (iii) death from cancer. We evaluated the progression of cancer over time by analyzing the cancer incidence data and cumulative mortalities data obtained from mice experiments conducted at the Institute for Environmental Sciences (IES). RESULTS: We analyzed non-irradiated control and 20 mGy/day × 400 days irradiated groups. In the analysis, all malignant neoplasms were lumped together and referred to as 'cancer'. Our analysis showed that the reduction in lifespan (104 days in median) was the result of the early onset of cancer (68 days in median) and the shortening of the cancer progression period (48 days in median). CONCLUSIONS: We described in detail our mathematical model for radiation-induced life-shortening attributed to cancer. We analyzed the mice data obtained from the experiment conducted at the IES using our model. We decomposed radiation-induced life-shortening into the early onset of cancer and the shortening of the cancer progression period.


Subject(s)
Neoplasms, Radiation-Induced , Mice , Animals , Radiation Dosage , Dose-Response Relationship, Radiation , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Gamma Rays , Models, Theoretical
8.
Nat Med ; 29(12): 3111-3119, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37946058

ABSTRACT

Over one million European children undergo computed tomography (CT) scans annually. Although moderate- to high-dose ionizing radiation exposure is an established risk factor for hematological malignancies, risks at CT examination dose levels remain uncertain. Here we followed up a multinational cohort (EPI-CT) of 948,174 individuals who underwent CT examinations before age 22 years in nine European countries. Radiation doses to the active bone marrow were estimated on the basis of body part scanned, patient characteristics, time period and inferred CT technical parameters. We found an association between cumulative dose and risk of all hematological malignancies, with an excess relative risk of 1.96 (95% confidence interval 1.10 to 3.12) per 100 mGy (790 cases). Similar estimates were obtained for lymphoid and myeloid malignancies. Results suggest that for every 10,000 children examined today (mean dose 8 mGy), 1-2 persons are expected to develop a hematological malignancy attributable to radiation exposure in the subsequent 12 years. Our results strengthen the body of evidence of increased cancer risk at low radiation doses and highlight the need for continued justification of pediatric CT examinations and optimization of doses.


Subject(s)
Hematologic Neoplasms , Neoplasms, Radiation-Induced , Radiation Exposure , Humans , Child , Adolescent , Young Adult , Adult , Radiation Dosage , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Hematologic Neoplasms/epidemiology , Hematologic Neoplasms/etiology , Radiation Exposure/adverse effects , Tomography, X-Ray Computed/adverse effects
9.
Radiat Res ; 200(1): 48-64, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37141110

ABSTRACT

The CGL1 human hybrid cell system has been utilized for many decades as an excellent cellular tool for investigating neoplastic transformation. Substantial work has been done previously implicating genetic factors related to chromosome 11 to the alteration of tumorigenic phenotype in CGL1 cells. This includes candidate tumor suppressor gene FOSL1, a member of the AP-1 transcription factor complex which encodes for protein FRA1. Here we present novel evidence supporting the role of FOSL1 in the suppression of tumorigenicity in segregants of the CGL1 system. Gamma-induced mutant (GIM) and control (CON) cells were isolated from 7 Gy gamma-irradiated CGL1s. Western, Southern and Northern blot analysis were utilized to assess FOSL1/FRA1 expression as well as methylation studies. GIMs were transfected to re-express FRA1 and in vivo tumorigenicity studies were conducted. Global transcriptomic microarray and RT-qPCR analysis were used to further characterize these unique cell segregants. GIMs were found to be tumorigenic in vivo when injected into nude mice whereas CON cells were not. GIMs show loss of Fosl/FRA1 expression as confirmed by Western blot. Southern and Northern blot analysis further reveals that FRA1 reduction in tumorigenic CGL1 segregants is likely due to transcriptional suppression. Results suggest that radiation-induced neoplastic transformation of CGL1 is in part due to silencing of the FOSL1 tumor suppressor gene promoter by methylation. The radiation-induced tumorigenic GIMs transfected to re-express FRA1 resulted in suppression of subcutaneous tumor growth in nude mice in vivo. Global microarray analysis and RT-qPCR validation elucidated several hundred differentially expressed genes. Downstream analysis reveals a significant number of altered pathways and enriched Gene Ontology terms genes related to cellular adhesion, proliferation, and migration. Together these findings provide strong evidence that FRA1 is a tumor suppressor gene deleted and epigenetically silenced after ionizing radiation-induced neoplastic transformation in the CGL1 human hybrid cell system.


Subject(s)
Cell Transformation, Neoplastic , Neoplasms, Radiation-Induced , Animals , Mice , Humans , Mice, Nude , Cell Transformation, Neoplastic/genetics , HeLa Cells , Genes, Tumor Suppressor , Carcinogenesis/genetics , Neoplasms, Radiation-Induced/pathology , Phenotype , Genomics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic
10.
CMAJ ; 195(16): E575-E583, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37094867

ABSTRACT

BACKGROUND: Children are more susceptible to radiation-induced damage than adults, but little research has compared the risk of cancer after exposure to radiation during computed tomography (CT) among children at different ages. We aimed to explore the risk of intracranial tumours, leukemia or lymphoma among children, adolescents and young adults (aged < 25 yr) after radiation exposure from CT at or before the age of 18 years. METHODS: We conducted a nested, population-based case-control study using data from Taiwan's publicly funded health care system. We identified participants younger than 25 years with newly diagnosed intracranial tumours, leukemia or lymphoma, from Jan. 1, 2000, to Dec. 31, 2013. We assigned 10 non-cancer controls for each case, matching by sex, date of birth and day of entry to the cohort. We considered CT scans received at or before the age of 18 years and 3 or more years before the index date (the date of cancer diagnosis for cases) as exposure. We used conditional logistic regression models and incidence rate ratios (IRRs) to estimate the relationship between risk of these cancers and CT radiation exposure. RESULTS: We identified 7807 cases and matched to 78 057 controls. Compared with no exposure, exposure to a single pediatric CT scan did not increase risk of intracranial tumours, leukemia or lymphoma. However, participants exposed to 4 or more CT scans had an elevated incidence (IRR 2.30, 95% confidence interval 1.43-3.71) of one of the cancer outcomes of interest. Receiving 4 or more CT scans at or before 6 years of age was associated with the highest risks of cancer, followed by ages 7-12 years and 13-18 years (p for trend < 0.001). INTERPRETATION: Exposure to a single CT scan was not associated with increased risks of subsequent intracranial tumours, leukemia or lymphoma among children; however, we observed increased cancer risks among those with 4 or more CT scans, especially among younger children. Although these cancers are uncommon, the findings of this study underscore the importance of prudent use of CT in the pediatric population.


Subject(s)
Brain Neoplasms , Leukemia , Lymphoma , Neoplasms, Radiation-Induced , Radiation Exposure , Adolescent , Young Adult , Child , Humans , Adult , Case-Control Studies , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Tomography, X-Ray Computed/methods , Lymphoma/complications
11.
Lancet Oncol ; 24(1): 45-53, 2023 01.
Article in English | MEDLINE | ID: mdl-36493793

ABSTRACT

BACKGROUND: The European EPI-CT study aims to quantify cancer risks from CT examinations of children and young adults. Here, we assess the risk of brain cancer. METHODS: We pooled data from nine European countries for this cohort study. Eligible participants had at least one CT examination before age 22 years documented between 1977 and 2014, had no previous diagnosis of cancer or benign brain tumour, and were alive and cancer-free at least 5 years after the first CT. Participants were identified through the Radiology Information System in 276 hospitals. Participants were linked with national or regional registries of cancer and vital status, and eligible cases were patients with brain cancers according to WHO International Classification of Diseases for Oncology. Gliomas were analysed separately to all brain cancers. Organ doses were reconstructed using historical machine settings and a large sample of CT images. Excess relative risks (ERRs) of brain cancer per 100 mGy of cumulative brain dose were calculated with linear dose-response modelling. The outcome was the first reported diagnosis of brain cancer after an exclusion period of 5 years after the first electronically recorded CT examination. FINDINGS: We identified 948 174 individuals, of whom 658 752 (69%) were eligible for our study. 368 721 (56%) of 658 752 participants were male and 290 031 (44%) were female. During a median follow-up of 5·6 years (IQR 2·4-10·1), 165 brain cancers occurred, including 121 (73%) gliomas. Mean cumulative brain dose, lagged by 5 years, was 47·4 mGy (SD 60·9) among all individuals and 76·0 mGy (100·1) among people with brain cancer. A significant linear dose-response relationship was observed for all brain cancers (ERR per 100 mGy 1·27 [95% CI 0·51-2·69]) and for gliomas separately (ERR per 100 mGy 1·11 [0·36-2·59]). Results were robust when the start of follow-up was delayed beyond 5 years and when participants with possibly previously unreported cancers were excluded. INTERPRETATION: The observed significant dose-response relationship between CT-related radiation exposure and brain cancer in this large, multicentre study with individual dose evaluation emphasises careful justification of paediatric CTs and use of doses as low as reasonably possible. FUNDING: EU FP7; Belgian Cancer Registry; La Ligue contre le Cancer, L'Institut National du Cancer, France; Ministry of Health, Labour and Welfare of Japan; German Federal Ministry of Education and Research; Worldwide Cancer Research; Dutch Cancer Society; Research Council of Norway; Consejo de Seguridad Nuclear, Generalitat de Catalunya, Spain; US National Cancer Institute; UK National Institute for Health Research; Public Health England.


Subject(s)
Brain Neoplasms , Glioma , Neoplasms, Radiation-Induced , Radiation Exposure , Child , Humans , Male , Female , Young Adult , Adult , Cohort Studies , Radiation Dosage , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/epidemiology , Brain Neoplasms/etiology , Glioma/diagnostic imaging , Glioma/epidemiology , Glioma/etiology , Radiation Exposure/adverse effects , Tomography, X-Ray Computed/adverse effects , Tomography, X-Ray Computed/methods
12.
Australas J Dermatol ; 64(1): 100-107, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36305612

ABSTRACT

BACKGROUND/OBJECTIVES: Low-dose X-ray radiotherapy to treat tinea capitis during childhood is a well-known risk factor for scalp basal cell carcinomas (BCCs). Post-radiotherapy BCCs are often multiple, and it has been suggested that they display more aggressive features. Our main objective was to study the clinicopathological aspects of post-radiotherapy BCCs to evaluate their biological behaviour and identify features that may differ from other BCCs. METHODS: We performed an observational, retrospective study assessing multiple clinical and pathological characteristics of patients with post-radiotherapy BCCs. RESULTS: We studied 96 patients with 427 post-radiotherapy scalp BCCs. Post-radiotherapy BCCs were often multiple (median of 4 lesions/patient, ranging from 1 to 54). Significant comorbidities included a high incidence of thyroid disease and meningiomas. Recurrences were observed in 23% of patients, but there may be confounding factors, such as referral bias, heterogenous treatment modalities and occurrence of new tumours due to field effect. We found a high incidence of infundibulocystic BCCs (in 14.6% of patients and corresponding to 5.4% of the total number of tumours), trichoblastomas (5.2%) and neurofibromas of the scalp (10%). CONCLUSIONS: This study is consistent with the occurrence of multiple lesions (sometimes numerous) and a relatively high tendency for recurrence in post-radiotherapy BCCs, as suggested by previous studies. We also found a high incidence of the infundibulocystic variant and a higher risk of follicular tumours and neurofibromas, which suggests that radiotherapy may influence the type of differentiation of BCCs and contribute to induce neoplasms of different cell lines.


Subject(s)
Carcinoma, Basal Cell , Meningeal Neoplasms , Neoplasms, Radiation-Induced , Neurofibroma , Skin Neoplasms , Tinea Capitis , Humans , Scalp/pathology , Retrospective Studies , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Carcinoma, Basal Cell/etiology , Carcinoma, Basal Cell/radiotherapy , Carcinoma, Basal Cell/epidemiology , Skin Neoplasms/etiology , Skin Neoplasms/radiotherapy , Skin Neoplasms/epidemiology , Tinea Capitis/radiotherapy , Tinea Capitis/complications , Neurofibroma/pathology
13.
Exp Dermatol ; 32(4): 341-347, 2023 04.
Article in English | MEDLINE | ID: mdl-36333872

ABSTRACT

Hydrochlorothiazide (HCTZ) is a frequently prescribed diuretic that exhibits photosensitizing properties. It is used to treat hypertension and edema. Dermato-epidemiological studies in various populations have linked HCTZ treatment with increased risk of particular types of skin cancer, including malignant melanoma (lentigo subtype), and both basal cell carcinoma and squamous cell carcinoma (SCC). This study investigated whether either of two different doses of HCTZ increased the risk of SCC development in mice exposed to ultraviolet radiation (UVR). A total of three groups of hairless mice were used in this study (total, N = 71). One group received a low dose (0.26 mg/mouse/day) and another group received a high dose (0.52 mg/mouse/day) of HCTZ in their drinking water; a third UVR control group received only tap water. All three groups were irradiated with UVR until the mice developed three tumours that were 4 mm in size. The times to SCC tumour development were recorded. In the low-dose group, the median time to develop an SCC tumour was 170 days; in both the high-dose group and the control group, the median time to develop anexd SCC tumour was 163 days (p ≥ 0.331). In our hairless mouse model, we found that mice treated with UVR plus HCTZ did not develop SCCs more rapidly than mice treated with UVR but not HCTZ.


Subject(s)
Carcinoma, Squamous Cell , Neoplasms, Radiation-Induced , Skin Neoplasms , Animals , Mice , Ultraviolet Rays/adverse effects , Mice, Hairless , Hydrochlorothiazide/adverse effects , Skin Neoplasms/pathology , Skin/pathology , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology
14.
Radiat Environ Biophys ; 61(4): 631-637, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36167896

ABSTRACT

Gastrointestinal (GI) cancer risk among astronauts after encountering galactic cosmic radiation (GCR) is predicted to exceed safe permissible limits in long duration deep-space missions. Current predictions are based on relative biological effectiveness (RBE) values derived from in-vivo studies using single-ion beams, while GCR is essentially a mixed radiation field composed of protons (H), helium (He), and heavy ions. Therefore, a sequentially delivered proton (H) → Helium (He) → Oxygen (O) → Silicon (Si) beam was designed to simulate simplified-mixed-field GCR (Smf-GCR), and Apc1638N/+ mice were total-body irradiated to sham or γ (157Cs) or Smf-GCR followed by assessment of GI-tumorigenesis at 150 days post-exposure. Further, GI-tumor data from equivalent doses of heavy-ions (i.e., 0.05 Gy of O and Si) in 0.5 Gy of Smf-GCR were compared to understand the contributions of heavy-ions in GI-tumorigenesis. The Smf-GCR-induced tumor and carcinoma count were significantly greater than γ-rays, and male preponderance for GI-tumorigenesis was consistent with our earlier findings. Comparison of tumor data from Smf-GCR and equivalent doses of heavy ions revealed an association between higher GI-tumorigenesis where dose received from heavy-ions contributed to > 95% of the total GI-tumorigenic effect observed after Smf-GCR. This study provides the first experimental evidence that cancer risk after GCR exposure could largely depend on doses received from constituent heavy-ions.


Subject(s)
Cosmic Radiation , Heavy Ions , Neoplasms, Radiation-Induced , Radiation Exposure , Space Flight , Mice , Male , Animals , Heavy Ions/adverse effects , Helium , Cosmic Radiation/adverse effects , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Carcinogenesis , Protons
15.
Radiat Res ; 198(5): 475-487, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36048240

ABSTRACT

Lung is one of the high-risk organs for radiation-induced carcinogenesis, but the risk of secondary lung-cancer development after particle-beam therapy and the underlying mechanism(s) remain to be elucidated. To investigate the effects of particle-beam radiation on adjacent normal tissues during cancer therapy, 7-week-old male and female B6C3F1 mice were irradiated with 0.2-4 Gy of gamma rays (for comparison), carbon ions (290 MeV/u, linear energy transfer 13 keV/µm), or fast neutrons (0.05-1 Gy, mean energy, ∼2 MeV), and lung-tumor development was assessed by histopathology. Mice irradiated with ≥2 Gy of carbon ions or ≥0.2 Gy of neutrons developed lung adenocarcinoma (AC) significantly sooner than did non-irradiated mice. The relative biological effectiveness values for carbon ions for lung AC development were 1.07 for male mice and 2.59 for females, and the corresponding values for neutrons were 4.63 and 4.57. Genomic analysis of lung ACs revealed alterations in genes involved in Egfr signaling. Hyperphosphorylation of Erk and a frequent nuclear abnormality (i.e., nuclear groove) were observed in lung ACs of mice irradiated with carbon ions or neutrons compared with ACs from non-irradiated or gamma-ray-irradiated groups. Our data indicate that the induction of lung AC by carbon ions occurred at a rate similar to that for gamma rays in males and approximately 2-to 3-fold greater than that for gamma rays in females. In contrast, the effect of neutrons on lung AC development was approximately 4- to 5-fold greater than that of gamma rays. Our results provide valuable information concerning risk assessment of radiation-induced lung tumors after particle-beam therapy and increase our understanding of the molecular basis of tumor development.


Subject(s)
Lung Neoplasms , Neoplasms, Radiation-Induced , Male , Female , Mice , Animals , Gamma Rays/adverse effects , Carbon/adverse effects , Relative Biological Effectiveness , Neutrons , Fast Neutrons , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Lung Neoplasms/etiology , Ions , Lung/pathology , Dose-Response Relationship, Radiation
16.
Int J Radiat Oncol Biol Phys ; 114(3): 502-515, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35840114

ABSTRACT

PURPOSE: The majority of cancer-related deaths are attributed to metastasis rather than localized primary tumor progression. However, the factors that regulate the premetastatic niche (PMN) and metastasis have not yet been clearly elucidated. We investigated the antimetastatic effects of irradiated tumor cell-derived microparticles (RT-MPs) and highlighted the role of innate immune cells in PMN formation. METHODS AND MATERIALS: Mice were treated 3 times with isolated RT-MPs, followed by tumor cell injection via the tail vein. The hematoxylin and eosin staining was performed to assess the number of tumor nodules in the lungs, and in vivo luciferase-based noninvasive bioluminescence imaging was conducted to detected tumor burden. The mechanisms of RT-MPs mediated PMN formation was evaluated using flow cytometry, transwell assay, and reverse transcription-polymerase chain reaction. RESULTS: RT-MPs inhibited tumor cell colonization in the lungs. Neutrophils phagocytosed RT-MPs and secreted CCL3 and CCL4, which induced monocytes chemotaxis and maturation into macrophages. RT-MPs promoted the transition of neutrophils and macrophages into antitumor phenotypes, hence inhibiting cancer cell colonization and proliferation. CONCLUSIONS: RT-MPs inhibited PMN formation and lung metastasis in a neutrophil- and macrophage-dependent but T cell-independent manner.


Subject(s)
Cell-Derived Microparticles , Lung Neoplasms , Neoplasms, Radiation-Induced , Animals , Cell-Derived Microparticles/pathology , Eosine Yellowish-(YS) , Hematoxylin , Lung/pathology , Lung Neoplasms/pathology , Mice , Neoplasms, Radiation-Induced/pathology , Tumor Microenvironment
18.
Future Oncol ; 18(4): 445-455, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35018785

ABSTRACT

Background: The effect of radiotherapy (RT) for second primary malignancies (SPMs) among prostate cancer survivors is controversial. Methods: Applying logistic regression, competing risk analysis and propensity score matching method, this study analyzed clinical data from the Surveillance, Epidemiology, and End Results program to compare the risk for SPMs between patients receiving RT and non-RT. Results: In this study, prostate cancer patients treated with RT developed more SPMs in the anus, bladder, rectum, liver, lung and bronchus and lymphoma than non-RT groups. Conclusion: More intensive surveillance should be adopted for these cancers among prostate cancer survivors.


Plain language summary Patients with prostate cancer have the highest 5-year survival rate, which increases the risk for developing second primary malignancies (SPMs). The effect of radiotherapy (RT) for SPMs is controversial among prostate cancer survivors. This study analyzed a mass of prostate cancer patients from a public database to compare risk for SPMs between RT and non-RT groups. RT indeed increased certain categories of SPM and intensive surveillance should be considered.


Subject(s)
Neoplasms, Radiation-Induced/etiology , Neoplasms, Second Primary/etiology , Prostatic Neoplasms/radiotherapy , Aged , Cancer Survivors/statistics & numerical data , Humans , Incidence , Male , Middle Aged , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/pathology , Neoplasms, Second Primary/epidemiology , Neoplasms, Second Primary/pathology , Propensity Score , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/pathology , Radiotherapy/adverse effects , Radiotherapy/statistics & numerical data , Risk Assessment , SEER Program/statistics & numerical data , United States/epidemiology
19.
Blood ; 139(2): 217-227, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34428282

ABSTRACT

Epidemiological data have provided limited and inconsistent evidence on the relationship between radiation exposure and lymphoid neoplasms. We classified 553 lymphoid neoplasm cases diagnosed between 1950 and 1994 in the Life Span Study cohort of atomic bomb survivors into World Health Organization subtypes. Mature B-cell neoplasms represented 58%, mature T-cell and natural killer (NK)-cell neoplasms 20%, precursor cell neoplasms 5%, and Hodgkin lymphoma (HL) 3%, with the remaining 15% classified as non-Hodgkin lymphoid (NHL) neoplasms or lymphoid neoplasms not otherwise specified. We used Poisson regression methods to assess the relationship between radiation exposure and the more common subtypes. As in earlier reports, a significant dose response for NHL neoplasms as a group was seen for males but not females. However, subtype analyses showed that radiation dose was strongly associated with increased precursor cell neoplasms rates, with an estimated excess relative risk per Gy of 16 (95% Confidence interval: 7.0, >533) at age 50. The current data based primarily of tissue-based diagnoses suggest that the association between radiation dose and lymphoid neoplasms as a group is largely driven by the radiation effect on precursor cell neoplasms while presenting no evidence of a radiation dose response for major categories of mature cell neoplasms, either B- or T-/NK-cell, or more specific disease entities (diffuse large B-cell lymphoma, plasma cell myeloma, adult T-cell leukemia/lymphoma) or HL.


Subject(s)
Atomic Bomb Survivors , Leukemia, Lymphoid/etiology , Lymphoma/etiology , Neoplasms, Radiation-Induced/etiology , Adolescent , Adult , Female , Humans , Incidence , Leukemia, Lymphoid/pathology , Lymphoma/pathology , Male , Middle Aged , Neoplasms, Radiation-Induced/pathology , Radioactive Fallout/adverse effects , Risk , World Health Organization , Young Adult
20.
J Clin Endocrinol Metab ; 107(2): e448-e457, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34555150

ABSTRACT

Radioactive iodine has been considered a safe and effective therapeutic option for hyperthyroidism secondary to Graves disease and autonomously functioning thyroid nodules since the mid-20th century. The question of whether I-131 at the doses used for hyperthyroidism might increase the risk of cancer has been investigated in a number of observational cohort studies over the years, with the preponderance of evidence being reassuring as to its safety. In particular, the 1998 Cooperative Thyrotoxicosis Therapy Follow-up Study (CTTFUS) has been widely cited as compelling evidence that I-131 is safe in hyperthyroidism therapy with respect to carcinogenesis. However, in 2019, a study by Kitahara and colleagues re-analyzed the CTTFUS cohort, extending the follow-up time and applying a novel dosimetric model for estimating tissue absorbed doses of radiation. This new analysis concluded that radioactive iodine was associated with an increased risk for mortality from overall cancer, breast cancer, and non-breast solid cancers. Reaction to this study was vociferous and particularly negative in the nuclear medicine literature. This mini-review was inspired by the 2019 CTTFUS controversy, and it is intended to provide the necessary context for clinicians to provide nuanced advice to their patients on the subject. To that end, the pre-2019 literature is surveyed, the 2019 CTTFUS study and a 2020 follow-up are discussed, and lessons from the literature and critical commentaries are considered.


Subject(s)
Hyperthyroidism/therapy , Iodine Radioisotopes/adverse effects , Neoplasms, Radiation-Induced/epidemiology , Thyroid Gland/radiation effects , Thyroid Neoplasms/epidemiology , Dose-Response Relationship, Radiation , Humans , Incidence , Neoplasms, Radiation-Induced/pathology , Observational Studies as Topic , Radiometry , Risk Assessment/statistics & numerical data , Thyroid Gland/pathology , Thyroid Neoplasms/etiology , Thyroid Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...