Fact-checked by Grok 2 weeks ago

Radiosensitivity

Radiosensitivity refers to the relative susceptibility of cells, tissues, organs, or organisms to the damaging effects of , primarily due to the induction and repair of DNA damage. This property is fundamental in radiation biology, as it governs the biological response to , ranging from acute tissue reactions to long-term effects like . The concept of radiosensitivity was pioneered in the early , most notably through the law of Bergonié and Tribondeau established in 1906, which posits that the radiosensitivity of cells is directly proportional to their mitotic activity and inversely proportional to their degree of differentiation. This law, derived from experiments on rabbit testes, provided an early framework for understanding why rapidly dividing cells, such as those in or , are more vulnerable to than quiescent or highly differentiated cells like neurons. Over time, this principle has been refined by molecular insights, revealing that radiosensitivity is modulated by pathways, including double-strand break repair mechanisms. Several factors influence radiosensitivity, including intrinsic genetic variations such as mutations in genes like ATM, BRCA1/2, or those associated with syndromes like ataxia-telangiectasia, which impair DNA repair efficiency. Extrinsic elements, such as age (with higher sensitivity in fetuses and young individuals), hormonal status, and oxygenation levels, also play critical roles; the oxygen effect, for instance, demonstrates that oxygenated tissues are two to three times more radiosensitive than hypoxic ones due to oxygen's role in fixing radiation-induced free radical damage. Hypoxic tumor environments, common in solid cancers, thus confer radioresistance, complicating treatment outcomes. In clinical contexts, radiosensitivity is pivotal for radiotherapy, where exploiting differences between tumor and normal tissue sensitivity enables targeted cell killing while sparing healthy structures. Biomarkers of radiosensitivity, including genetic profiling and functional assays, are increasingly used to predict patient responses, personalize dosing, and integrate radiosensitizers like oxygen-mimetic compounds to enhance tumor control. As of 2025, advances in individual radiosensitivity testing, including the Radiation-Induced Lymphocyte Apoptosis (RILA) assay and AI-integrated multi-omics approaches, are improving predictions of treatment toxicity and enabling more precise personalization of radiotherapy regimens. This understanding also informs radiation protection standards, emphasizing the need to account for individual variability in occupational and environmental exposures.

Definition and Fundamentals

Definition

Radiosensitivity refers to the relative of cells, tissues, organs, , or other substances to the injurious action of , typically measured by the degree of damage or lethality induced per unit dose absorbed. This concept encompasses the varying responses of biological entities to , where the extent of harm depends on the inherent vulnerability of the target to events that disrupt molecular structures. A key distinction lies between radiosensitive and radioresistant entities: highly radiosensitive cells, such as lymphocytes in lymphoid organs and , exhibit rapid damage and even at low doses due to their high division rates, whereas radioresistant cells like mature muscle and cells show minimal effects owing to their low metabolic activity and non-dividing nature. This variation underscores that radiosensitivity is not uniform across cell types, with actively proliferating, undifferentiated cells generally more prone to radiation-induced . Basic principles of radiosensitivity involve dose-response relationships that describe how biological effects scale with . For low doses, the linear no-threshold (LNT) model posits a proportional increase in risks, such as cancer , with no safe exposure level, extrapolated from higher-dose data. In contrast, threshold models apply to high doses and deterministic effects, where damage manifests only above a certain dose level beyond which severity escalates. The primary unit for measuring , which quantifies radiosensitivity, is the (), defined as 1 joule of energy deposited per of matter (1 = 1 J/kg). Biological effectiveness relates to this through the in sieverts (), which adjusts the by a radiation weighting factor to account for the varying capacity of different types to cause biological damage.

Historical Development

The discovery of X-rays by Wilhelm Conrad Röntgen in 1895 marked the inception of observations related to radiosensitivity, as early experimenters soon reported cases of skin burns and other injuries from prolonged exposure during radiographic studies. These initial findings highlighted the biological effects of , prompting awareness of tissue damage thresholds, though the underlying mechanisms remained unexplored for decades. In 1906, French radiologists Jean Bergonié and Louis Tribondeau formulated a foundational principle linking radiosensitivity to cellular characteristics, stating that radiosensitivity is directly proportional to the reproductive activity of and inversely proportional to their degree of differentiation. This law provided an early framework for predicting tissue responses to , emphasizing that rapidly dividing, undifferentiated —such as those in germinal epithelium or —are more vulnerable than quiescent, mature ones. By the mid-20th century, quantitative approaches advanced this understanding through the development of cell survival curves, notably by Titia Alper and Norma E. Gillies in 1960, who analyzed sigmoid-shaped curves in irradiated to model dose-dependent lethality and recovery processes. Concurrently, the linear-quadratic (LQ) model emerged as a key tool for describing cell killing, combining linear (irreparable) and quadratic (repairable) components of damage, with seminal contributions tracing back to studies in the and formalization in by the 1970s. From the 1950s to 1960s, research solidified DNA as the primary target of radiation-induced damage, as correlations between chromosomal aberrations and cell death underscored the genetic basis of radiosensitivity, shifting focus from vague tissue effects to molecular lesions like double-strand breaks. This era also saw the establishment of international standards, with the International Commission on Radiological Protection (ICRP) issuing Publication 26 in 1977, which updated dose limits and protection principles based on evolving evidence of stochastic and deterministic risks. Subsequent ICRP revisions, such as Publication 60 in 1990, refined these guidelines to incorporate linear no-threshold assumptions for low-dose exposures. By the 2020s, advancements in have enabled personalized assessments of radiosensitivity, integrating multi-omics data—such as signatures—to predict individual responses and tailor radiotherapy, with tools like 10-gene predictors showing promise for identifying high-risk patients. These developments, highlighted in reviews up to 2025, build on historical milestones by leveraging high-throughput sequencing to address variability in pathways and tumor responses.

Factors Affecting Radiosensitivity

Intrinsic Cellular Factors

Intrinsic cellular factors refer to the inherent biological properties within that dictate their baseline response to , independent of external influences. These factors include the cell's position in the , its degree of , genetic makeup affecting repair mechanisms, and tissue-specific characteristics. Variations in these intrinsic elements lead to differences in radiosensitivity across cell types and organisms, with rapidly proliferating or repair-deficient cells generally exhibiting heightened vulnerability. Cell cycle position profoundly influences radiosensitivity, as and condensation states vary across phases, affecting damage susceptibility and repair efficiency. Cells in the mitotic () phase and are the most sensitive, showing steeper survival curves with lower surviving fractions at equivalent doses compared to other phases, due to condensed chromosomes that hinder repair and increase lethality from double-strand breaks. In contrast, late S-phase cells display the lowest sensitivity, with shallower survival curves reflecting enhanced repair during active , while G1-phase cells exhibit intermediate resistance. For instance, in cells exposed to X-rays, M-phase cells yielded the lowest survival, underscoring this phase-dependent pattern. The degree of cellular differentiation and specialization also determines radiosensitivity, with undifferentiated, rapidly dividing cells proving more vulnerable than mature, quiescent ones. Stem cells and progenitors in tissues like undergo frequent , rendering them highly susceptible to radiation-induced or , as their active proliferation amplifies unrepaired damage propagation. Conversely, highly differentiated, non-dividing cells such as neurons possess lower metabolic rates and robust repair pathways, conferring greater resistance despite potential vulnerability in supportive . This gradient aligns with the law of Bergonié and Tribondeau, which posits that radiosensitivity inversely correlates with and directly with division rate. Genetic and molecular factors, particularly mutations in genes, significantly modulate intrinsic by impairing the cell's ability to mend radiation-induced lesions. Defects in ataxia-telangiectasia mutated () kinase, crucial for detecting double-strand breaks and activating checkpoints, lead to heightened , as seen in ATM-deficient tumors that respond better to radiotherapy due to persistent damage. Similarly, mutations compromise repair, increasing lethality from unrepaired breaks and elevating overall cellular vulnerability. Tissue-specific examples illustrate these intrinsic differences in practice. Lymphocytes, as rapidly renewing immune cells, exhibit high , undergoing rapid post-irradiation due to limited repair capacity and high division rates, which contributes to lymphopenia in exposed individuals. Spermatogonia in the testes represent another highly sensitive population, with radiation targeting these stem cells to cause sterility at doses as low as 1-6 , reflecting their undifferentiated state and active proliferation. Hepatocytes, in contrast, show moderate , tolerating higher doses (up to 30-50 in partial liver volumes) owing to their differentiated, relatively quiescent nature and efficient metabolic repair, though chronic exposure can still induce . These examples highlight how intrinsic factors translate to organ-level responses, with DNA as a primary target whose repair underpins variations.

Extrinsic Environmental Factors

Extrinsic environmental factors play a crucial role in modulating radiosensitivity by altering the cellular response to ionizing radiation exposure. These factors include the availability of oxygen, temperature, metabolic conditions, the quality of radiation, and the presence of chemical agents, each interacting with radiation-induced damage pathways to either enhance or diminish cell lethality. The oxygen effect is one of the most significant extrinsic modifiers of radiosensitivity, where the presence of molecular oxygen during irradiation dramatically increases cellular vulnerability. Under normoxic conditions, oxygen acts as a radiosensitizer by fixing radiation-induced free radical damage to DNA, preventing repair and leading to permanent lesions. In contrast, hypoxic cells, which lack sufficient oxygen (typically pO2 below 10 mmHg), exhibit 2-3 times greater resistance to radiation, quantified by the oxygen enhancement ratio (OER) of approximately 2-3 for low-linear energy transfer (LET) radiation such as X-rays. This resistance arises primarily from the indirect action of radiation, where ionizing events produce reactive species like hydroxyl radicals from water radiolysis; without oxygen, these radicals cause reversible damage that cells can repair more effectively. Hypoxia commonly occurs in solid tumors due to poor vascularization, contributing to therapeutic resistance in radiotherapy. Temperature influences radiosensitivity by affecting enzymatic repair processes and membrane integrity, with higher temperatures generally increasing sensitivity. Hyperthermia, typically at 40-43°C, radiosensitizes cells by inhibiting DNA repair mechanisms, such as and , and by enhancing the indirect damage from free radicals. This effect is more pronounced in tumor microenvironments, where elevated temperatures exacerbate stress in already compromised cells. Metabolic states, including nutrient deprivation and alterations, further modulate sensitivity; nutrient-poor conditions, often coupled with , can slow metabolic rates and enhance repair capacity, thereby increasing resistance, while acidic (below 6.5) in tumor regions may radiosensitize by disrupting protein function and repair enzymes, though the net effect varies by . The quality of radiation, characterized by its , determines the dependence on extrinsic factors like oxygen. Low-LET radiation, such as X-rays or gamma rays, relies heavily on indirect damage through free radicals, making it highly oxygen-dependent with an OER of 2-3. In contrast, high-LET radiation, like neutrons or heavy ions, deposits densely along tracks, causing complex DNA lesions primarily through direct , which reduces oxygen dependence and results in a lower OER (often 1.5-2). Neutrons, for example, exhibit reduced hypoxic protection compared to X-rays, offering advantages in treating oxygen-deficient tumors. Chemical modifiers provide targeted control over radiosensitivity, acting as sensitizers or protectors to optimize therapeutic ratios. Radiosensitizers, such as halogenated pyrimidines (e.g., bromodeoxyuridine or iododeoxyuridine), incorporate into DNA and increase damage from low-LET by facilitating strand breaks upon irradiation, with dose-modifying factors (DMF) around 1.2-1.5. Conversely, radioprotectors like thiols, including WR-2721 (), scavenge free radicals and protect normal tissues, achieving DMFs of 2-3 by preferentially accumulating in non-tumor cells due to their . These agents allow modulation of the radiation response without altering intrinsic cellular properties, with clinical applications focused on enhancing tumor kill while sparing healthy tissue.

Mechanisms of Radiation Damage

Target Molecules and Structures

Ionizing radiation primarily targets DNA within the cell nucleus, where it induces damage through both direct and indirect mechanisms. Direct damage occurs when radiation energy ionizes DNA molecules, breaking chemical bonds and leading to strand breaks, while indirect damage is mediated by reactive oxygen species (ROS), such as hydroxyl radicals (•OH) generated from water radiolysis, which oxidize DNA bases and sugars. Among these lesions, double-strand breaks (DSBs) are the most lethal form of DNA damage, as they sever both strands of the DNA double helix and are particularly difficult to repair accurately, often resulting in cell death, genomic instability, or mutations if unrepaired. Beyond DNA, ionizing radiation affects other essential macromolecules, contributing to overall cellular dysfunction and radiosensitivity. Proteins, including critical enzymes, undergo oxidation by •OH radicals, forming carbonyl groups on amino acid residues like lysine, cysteine, and histidine, or reductive modifications at sulfur-containing sites, which impair enzymatic activity and disrupt signaling pathways. Lipids in cell membranes experience peroxidation of polyunsaturated fatty acids, producing reactive aldehydes such as malondialdehyde and 4-hydroxynonenal, which increase membrane permeability and trigger inflammatory responses or apoptosis. RNA molecules are also susceptible to strand breaks and oxidative modifications from ROS, potentially altering mRNA stability, translation efficiency, and protein synthesis, though these effects are generally less studied than DNA damage. The sensitivity to radiation varies across cellular compartments, with the nucleus being the most vulnerable due to its housing of genomic DNA, where even low doses can induce dozens of DSBs per gray of exposure. The cytoplasm follows in sensitivity, as damage here affects metabolic processes and cytoskeletal integrity through ROS-mediated oxidation. Organelles like mitochondria are particularly impacted, where radiation-generated ROS disrupt mitochondrial DNA and respiratory chain components, leading to energy production failure, persistent oxidative stress, and amplification of apoptotic signals. Cells mitigate damage to these targets primarily through DNA repair pathways focused on DSBs, with (NHEJ) and (HR) being the dominant mechanisms. NHEJ rapidly ligates broken ends using proteins like Ku70/80, , and ligase IV, operating error-pronely throughout the but with higher efficiency in and non-dividing cells. In contrast, HR employs a sister chromatid template for accurate repair via resection factors (e.g., CtIP, Exo1) and Rad51, predominating in S and G2 phases where template availability enhances fidelity, though its efficiency diminishes in quiescent or differentiated cell types. The balance between these pathways influences radiosensitivity, as defects in either can exacerbate lethal outcomes from unrepaired DSBs.

Types of Cellular Damage

Ionizing radiation primarily induces lethal cellular damage through irreparable DNA double-strand breaks (DSBs), which overwhelm repair mechanisms and trigger cell death pathways such as mitotic catastrophe or apoptosis. These DSBs, the most lethal lesions produced by radiation, often result from direct ionization of DNA or indirect effects via reactive oxygen species, leading to chromosomal fragmentation during mitosis if unrepaired. In radiosensitive cells, such as those in rapidly dividing tissues, this damage manifests as reproductive death, where cells lose clonogenic potential, contributing to overall tissue radiosensitivity. Sublethal damage refers to radiation-induced lesions, such as single-strand breaks (SSBs) or oxidatively damaged bases, that cells can repair individually without immediate lethality but may become lethal if they interact with additional damage from subsequent exposures. Potentially lethal damage encompasses more complex clustered lesions, including closely spaced SSBs or base damage that could evolve into DSBs during attempted repair, particularly under conditions that delay or inhibit repair processes like prolonged arrest. The concept of exploits this repairability, as sublethal and potentially lethal damage from fractionated low doses can be repaired between exposures, reducing overall killing and highlighting variations in radiosensitivity based on repair . Chromosomal aberrations arise from misrepair of radiation-induced DSBs, serving as key indicators of cellular damage and radiosensitivity. Common types include dicentric chromosomes, formed by asymmetrical exchanges that create unstable structures with two centromeres, and acentric fragments, which lack centromeres and are lost during , often leading to mitotic death. These aberrations can also result in micronuclei formation, where acentric fragments or whole lagging chromosomes are enclosed in separate cytoplasmic nuclei, providing a quantifiable marker for exposure dose and repair proficiency in biodosimetry assays. Beyond DNA-targeted damage, causes non-DNA cellular effects through , where excess (ROS) generated in mitochondria damage proteins, lipids, and membranes, potentially inducing mutations via indirect genomic instability. This oxidative burden promotes , an irreversible growth arrest state characterized by enlarged morphology and secretion of pro-inflammatory factors (, SASP), which correlates with increased radiosensitivity in normal cells through pathways like ATM-p53-p21 signaling. Additionally, bystander effects enable irradiated cells to transmit damage signals—via exosomes containing miRNAs and proteins or soluble factors like cytokines—to unirradiated neighboring cells, inducing , ROS elevation, and DNA damage in a non-targeted manner, thereby amplifying overall radiosensitivity.

Biological Effects

Deterministic Effects

Deterministic effects, also known as reactions, are non- biological responses to that occur only when the exceeds a specific , with the severity of the effect increasing in a predictable manner as the dose rises above that . These effects result from the killing or malfunction of a large number of cells in a or , leading to observable clinical manifestations rather than random cellular changes. Unlike effects, deterministic effects have a clear dose-response relationship where no response is expected below the , but higher doses amplify the damage through mechanisms such as and dysfunction. For example, the for transient skin , an early sign of radiation injury, is typically around 2 for a single acute exposure. Acute radiation syndromes represent severe deterministic effects following whole-body or significant partial-body , categorized by the dose range and affected systems. The hematopoietic syndrome emerges at doses of 2-6 , primarily due to failure, resulting in , increased infection risk, and hemorrhage; this underlies the depletion of rapidly dividing stem cells, leading to lethal cellular damage if untreated. At higher doses of 6-10 , the gastrointestinal syndrome predominates, characterized by denudation of the intestinal mucosa, severe , , and from the destruction of rapidly proliferating crypt cells. The cerebrovascular syndrome, occurring above 20 , involves acute neurological damage, including , , and rapid death within days, stemming from vascular and neuronal cell . These syndromes progress through distinct s: a prodromal (hours to days post-) with , , and ; a latent of apparent recovery (days to weeks); a manifest illness with peak symptoms; and a recovery or death lasting weeks to months, depending on dose and supportive care. Specific tissue reactions exemplify deterministic effects in localized exposures, such as radiation dermatitis and . Radiation dermatitis manifests as skin , dry or moist , and ulceration, with thresholds starting at 2-6 for early and escalating to severe reactions above 10-15 , following a similar phased time course from prodromal redness to potential in recovery. Oral , common in head and neck radiotherapy, involves mucosal inflammation, ulceration, and pain, with deterministic thresholds around 20-30 cumulative dose, leading to impaired swallowing and nutrition during the manifest phase. For whole-body exposure, the (LD50/30)—the dose causing 50% mortality within 30 days without treatment—is approximately 3-4 , primarily due to hematopoietic , though this can be mitigated to higher levels (up to 7-10 ) with medical countermeasures like (G-CSF) to support recovery.

Stochastic Effects

Stochastic effects of are probabilistic health outcomes, such as cancer and hereditary disorders, where the likelihood of occurrence increases with dose but the severity of the effect remains independent of dose. These effects are modeled under the linear no-threshold (LNT) framework, which assumes a linear dose-response relationship with no safe threshold, implying that even low doses carry some risk proportional to exposure. The LNT model underpins standards by extrapolating risks from higher-dose data to lower levels. Cancer induction represents a primary stochastic effect, with radiation damaging DNA and leading to oncogenic transformations over time. Leukemia typically exhibits a shorter latency period, peaking 5-7 years post-exposure, followed by a decline, unlike solid tumors which manifest after a minimum of 10 years. For instance, the Chernobyl accident in 1986 resulted in elevated thyroid cancer rates among exposed children due to radioiodine release, demonstrating dose-dependent increases in incidence. The relative biological effectiveness (RBE) varies by radiation type; for stochastic effects at low doses, photons like X-rays have an RBE of 1, while neutrons can reach 20 or higher, amplifying risk per unit absorbed dose. Hereditary effects arise from mutations in germ cells transmitted to offspring, potentially causing genetic disorders. In humans, these effects have low incidence, with radiation-induced contributions estimated at less than 1% of all genetic diseases, reflecting the rarity observed in epidemiological data despite animal model evidence of mutagenicity. Epidemiological evidence from the Life Span Study (LSS) of atomic bomb survivors in and provides key insights, showing a dose-dependent excess (ERR) per of approximately 0.47 for solid cancers among those exposed at age 30 and reaching age 70. This cohort demonstrates linear risk elevation without a threshold, supporting the LNT model for cancer risks across a wide dose range.

Clinical and Practical Applications

In Radiotherapy

In radiotherapy, radiosensitivity principles are leveraged to maximize tumor destruction while minimizing damage to surrounding normal tissues, creating a therapeutic window that exploits inherent differences in cellular responses to radiation. Radiosensitive tumors, such as lymphomas, exhibit high susceptibility to due to their rapid proliferation and limited repair capacity, allowing effective control with relatively low doses. In contrast, normal tissues like the are more radioresistant, characterized by slower cell turnover and robust mechanisms, which permits selective targeting of malignant cells. This differential sensitivity forms the basis for treatment planning, where rapidly dividing tumors are prioritized for radiosensitive exploitation. Fractionation regimens in radiotherapy are designed based on the linear-quadratic (LQ) model, which quantifies cell survival as a function of dose, incorporating linear (α) and quadratic (β) components of damage. Early-responding tissues and tumors typically have an α/β ratio of approximately 10 Gy, reflecting sensitivity to fraction size, while late-responding normal tissues have a lower ratio of 2-4 Gy, emphasizing the need for smaller, multiple fractions to spare them. This model guides dose escalation strategies, balancing tumor control against late toxicities. Post-2010s trends have shifted toward hypofractionation—delivering higher doses per fraction in fewer sessions—for certain cancers like breast and prostate, supported by clinical trials demonstrating equivalent efficacy and reduced treatment burden without increased complications. To enhance radiosensitivity, radiotherapy is often combined with sensitization strategies that amplify DNA damage or impair repair in tumor cells. Chemotherapy agents like act as radiosensitizers by inhibiting DNA-protein kinase activity, thereby potentiating radiation-induced double-strand breaks, particularly in head and neck and cancers. modifiers, such as nitroimidazoles (e.g., nimorazole), target oxygen-deficient tumor regions that resist radiation by mimicking oxygen's role in fixing free radical damage, improving outcomes in hypoxic solid tumors like head and neck squamous cell carcinomas. For cancers with DNA repair defects, such as BRCA-mutated breast and ovarian tumors, (e.g., ) selectively radiosensitize defective cells by preventing alternative repair pathways, widening the therapeutic window without excessively harming proficient normal tissues. Treatment success is evaluated through tumor control probability (TCP), which estimates the likelihood of eradicating all clonogenic tumor cells, versus normal tissue complication probability (NTCP), which predicts risks like or in healthy organs. Balancing high TCP with low NTCP remains challenging due to inter-patient variability in radiosensitivity, but advances in intensity-modulated radiation therapy (IMRT) and have improved precision by 2025, conforming doses more tightly to tumors and reducing integral doses to adjacent structures, as evidenced by phase III trials showing comparable quality-of-life outcomes to traditional methods. These technologies exploit intrinsic factors like tumor proliferation rates to optimize radiosensitivity in .

Radiation Protection and Testing

Radiation protection in contexts involving potential exposure to ionizing radiation emphasizes minimizing risks through established principles and targeted assessments. The ALARA principle, which stands for "as low as reasonably achievable," guides efforts to reduce radiation doses by optimizing shielding, , and time of while balancing practical constraints. The (ICRP) recommends occupational effective dose limits of 20 mSv per year, averaged over five consecutive years, with no single year exceeding 50 mSv, to prevent deterministic effects and limit risks. These limits apply to workers in facilities, settings, and environments, ensuring that exposures remain below thresholds associated with significant impacts. Assessing individual radiosensitivity is crucial for tailoring protection strategies, particularly for those at elevated risk. Common methods include lymphocyte sensitivity assays, which evaluate DNA damage in peripheral blood lymphocytes exposed to low-dose radiation in vitro to predict personal response. Dicentric chromosome analysis, a gold-standard cytogenetic technique, quantifies unstable chromosomal aberrations formed by radiation-induced double-strand breaks, providing retrospective dose estimates in biodosimetry. Genomic profiling, such as the G2 chromosomal radiosensitivity assay, measures chromatid aberrations in lymphocytes arrested in the G2 phase after irradiation, offering insights into inherent cellular repair deficiencies. Certain populations exhibit heightened radiosensitivity, necessitating enhanced protective measures. Children are particularly vulnerable due to their rapid cellular and longer lifespan for potential effects to manifest, with risks amplified during organ phases. Pregnant women require special consideration, as fetal exposure below 100 mGy poses no justification for termination based on risk alone, though doses are minimized to protect embryonic . Individuals with hypersensitive syndromes, such as ataxia-telangiectasia caused by ATM mutations, display extreme sensitivity to , leading to severe cellular damage even at low doses and informing strict avoidance protocols. Recent advancements up to 2025 have integrated into radiosensitivity prediction and biodosimetry for more precise risk management. AI-driven models, leveraging multi-omics data like and transcriptomics, enable personalized risk stratification by forecasting individual responses to with improved accuracy over traditional assays. In post-accident scenarios, such as those informed by the Fukushima Daiichi incident, Japan's enhanced biodosimetry networks now incorporate AI-assisted cytogenetic analysis for rapid , accelerating dose estimation and medical response in mass casualty events. These tools support proactive and occupational screening, reducing uncertainties in effect quantification like cancer risk.

References

  1. [1]
    Cell Radiosensitivity - Engineered Radiation Safety - NDE-Ed.org
    Radiosensitivity is the relative susceptibility of cells, tissues, organs, organisms, or other substances to the injurious action of radiation.
  2. [2]
    Human Radiosensitivity and Radiosusceptibility: What Are the ... - NIH
    Jul 2, 2021 · The radiosensitivity is the proneness to the adverse tissue events that are considered as non-cancer radiation-induced effects and attributable ...
  3. [3]
    The law of Bergonié and Tribondeau: A nice formula for a first ...
    ... Bergonié and Tribondeau established a link between radiosensitivity and proliferation as a 'law' in 1906. Although it is still popular and taken as one of ...<|control11|><|separator|>
  4. [4]
    Radiosensitivity | Radiology - RSNA Journals
    THE general laws governing radiosensitivity were recognized many years ago and have always been an important guide in radiotherapy.
  5. [5]
    Biological Effects of Radiation and Units of Dose
    Age, species, and fractionation. Other factors affect radiosensitivity. As expected, radiosensitivity is greatest during the fetal stage and becomes ...
  6. [6]
    Cellular and Molecular Mechanisms Underlying Oxygen-Dependent ...
    May 4, 2015 · Molecular oxygen has long been recognized as a powerful radiosensitizer that enhances the cell-killing efficiency of ionizing radiation.
  7. [7]
    BfS - Individual radiosensitivity - Bundesamt für Strahlenschutz
    The term "radiation sensitivity" describes the sensitivity of organisms to the effect of ionising radiation. This can relate to the level of individual cells, ...Individual Radiation... · Dna Repair Defects As... · Biomarkers Or Bioassays To...
  8. [8]
    Normal tissue radiosensitivity — How important is it?
    The success of radiotherapy in eradicating tumours depends on the total radiation dose, but what limits this dose is the tolerance of the normal tissues ...
  9. [9]
    Radiosensitivity index emerges as a potential biomarker for ... - Nature
    Jun 2, 2021 · Radiotherapy is one of the most effective treatments for uncomplicated locoregional tumours, and around half of all the cancer patients receive ...
  10. [10]
    [PDF] RADIOSENSITIVITY OF CELLS - International Atomic Energy Agency
    Mammalian cells are very sensitive to radiation, while some micro-organisms are highly radioresistant, with varying doses needed to kill them.Missing: definition | Show results with:definition
  11. [11]
    Radiation Quantities and Units, Definitions, Acronyms - NCBI - NIH
    gray (Gy) The SI unit of absorbed dose. 1 Gy equals an absorbed dose of 1 J/kg (100 rad). internal dose The dose received from radioactive material taken into ...
  12. [12]
    Linear-Non-Threshold Model - Canadian Nuclear Safety Commission
    The LNT model assumes a direct and proportional relationship between radiation exposure and cancer risk with all radiation doses.Linear-Non-Threshold Model · April 2013 · Radiation Risk Models
  13. [13]
    German scientist discovers X-rays | November 8, 1895 - History.com
    Initially, it was believed X-rays passed through flesh as harmlessly as light. However, within several years, researchers began to report cases of burns and ...
  14. [14]
    The Evolution of Radiation Safety: A History of X-Ray Badges
    Jun 4, 2024 · X-ray safety awareness began after Wilhelm Conrad Röntgen discovered X-rays in 1895. Reports of radiation "burns" and other injuries emerged as ...
  15. [15]
    The law of Bergonié and Tribondeau: a nice formula for a ... - PubMed
    Bergonié and Tribondeau established a link between radiosensitivity and proliferation as a 'law' in 1906. Although it is still popular and taken as one of the ...Missing: 1912 | Show results with:1912
  16. [16]
    Theoretical survival curves for radiation damage in bacteria
    Theoretical survival curves for radiation damage in bacteria. Author ... Alper and Gillies, 1958. T. Alper, N.E. Gillies. J. gen. Microbiol, 18 (1958) ...
  17. [17]
    The linear quadratic model: usage, interpretation and challenges
    Dec 19, 2018 · The linear-quadratic model is one of the key tools in radiation biology and physics. It provides a simple relationship between cell survival and delivered dose.
  18. [18]
    Linking the History of Radiation Biology to the Hallmarks of Cancer
    May 8, 2014 · Because the formation of chromosomal aberrations correlated with cell killing, it was established early that DNA was the lethal target of ...
  19. [19]
    ICRP Publication 26
    Recommendations of the ICRP. Recommended citation. ICRP, 1977. Recommendations of the ICRP. ICRP Publication 26. Ann. ICRP 1 (3). Download PDF. Translations.Missing: guidelines | Show results with:guidelines
  20. [20]
    [PDF] Evolution of ICRP Recommendations 1977, 1990 and 2007
    Firstly, it is necessary to adopt a set of risk estimates for the effects on health per unit of exposure to ionising radiation that are applicable for.
  21. [21]
    Genomic predictors of radiation response: recent progress towards ...
    Dec 18, 2024 · Current personalized radiogenomics approaches for all indications include a 10-gene signature for radiosensitivity, which includes expression ...
  22. [22]
    Individual Radiosensitivity Testing in 2025: Current Advances and ...
    Sep 13, 2025 · Advances in omics technologies - genomics, transcriptomics, proteomics, and metabolomics - have opened new avenues for understanding RS.
  23. [23]
    Radiation‐induced tissue damage and response - PMC
    Acute responding tissues, like intestine, bone marrow, skin and testes, turn over rapidly and have well‐defined stem cell populations, at least a portion of ...
  24. [24]
    Role of cell cycle in mediating sensitivity to radiotherapy - PubMed
    Jul 15, 2004 · The cell cycle phase also determines a cell's relative radiosensitivity, with cells being most radiosensitive in the G(2)-M phase, less ...
  25. [25]
    Cell Cycle Dependence of Ionizing Radiation-Induced DNA ... - NIH
    HeLa cells in M phase were the most sensitive to X-ray cell killing, G1 and G2 were the most radioresistant, and S-phase cells were intermediately sensitive. ...
  26. [26]
    SUMMARY OF HEALTH EFFECTS OF IONIZING RADIATION - NCBI
    Cells that are rapidly growing and dividing (such as those found in the gastrointestinal tract, bone marrow, reproductive and lymphoid tissues, and fetal nerve ...
  27. [27]
    Increased Radiosensitivity of Solid Tumors Harboring ATM ... - NIH
    Tumors with ATM and BRCA1/2 mutations exhibited superior tumor response and local control after RT compared to tumors without these mutations.
  28. [28]
    BRCA1 Mutation: A Predictive Marker for Radiation Therapy? - PMC
    The impaired ability for DSBs repair contributes to radiation sensitivity when BRCA1 is deficient. However, which DSBs repair pathways controlled by BRCA1 play ...
  29. [29]
    Radiosensitivities of Selected Amphibians in Relation to Their ... - jstor
    There exists among species of amphibians a wide range of nuclear (1) and. (1, 2) size, with many species having relatively large chromosomes (3-8). C.Missing: interspecies | Show results with:interspecies
  30. [30]
    A review on lymphocyte radiosensitivity and its impact on radiotherapy
    B cells were the most sensitive, followed by T cells (CD4+, CD8+) and NK cells. CD34+ progenitor cells appeared to be highly radioresistant. Note that the ...
  31. [31]
    Quercetin regulates sensitivity to X-ray radiation of hepatocellular ...
    HCC is only moderately sensitive to IR, similar to the radiosensitivity of poorly differentiated squamous cell carcinoma [7], which is a major cause of ...
  32. [32]
    Effects of Ionizing Radiation on Biological Molecules—Mechanisms ...
    DNA damaging events inflicted by IR alone include the deleterious alteration of bases and sugars, cross-link formation, single- and double-strand breaks (SSBs/ ...
  33. [33]
    Ionizing radiation-induced DNA injury and damage detection in ...
    Ionizing radiation causes DSBs directly, but in addition base damages due to indirect effects are also induced. This radiation causes formation of ROS (reactive ...
  34. [34]
    Initiation and Prevention of Biological Damage by Radiation ... - NIH
    Dec 30, 2021 · Ionizing radiations cause chemical damage to proteins. In aerobic aqueous solutions, the damage is commonly mediated by the hydroxyl free ...
  35. [35]
    Multiple radiations and its effect on biological system
    Apr 12, 2025 · RNA strand breaks are one of the most frequent types of damage caused by ionizing radiation. Radiation can induce reactive oxygen species (ROS) ...
  36. [36]
    Introduction to Radiobiology of Targeted Radionuclide Therapy - PMC
    Mar 17, 2015 · DNA, mitochondria, and the cell membrane are the main sensitive targets of radiation. ... cytoplasm, organelles, or nucleus) according to ...
  37. [37]
    Mitochondria-targeted antioxidant enzyme activity regulates ...
    We investigated the hypothesis that mitochondrial antioxidant enzyme activity regulates radioresistance in human pancreatic cancer cells.Mnsod Activity Regulates... · Mnsod Overexpression Alters... · Mitochondrial Ros Regulate...
  38. [38]
    Detection and repair of ionizing radiation induced DNA double ...
    Non-homologous end joining (NHEJ) is the major pathway for the repair of ionizing radiation (IR)-induced DNA double strand breaks (DSBs) in human cells.
  39. [39]
    The Determinant of DNA Repair Pathway Choices in Ionising ...
    Aug 25, 2020 · IR can attack DNA and produce a variety of DNA lesions, mainly including DNA double-strand breaks (DSBs), DNA single-strand breaks (SSBs), ...
  40. [40]
    Targeting the Double-Strand DNA Break Repair Pathway as a ...
    Aug 9, 2006 · DNA double-strand breaks (DSB) can be particularly problematic; modest numbers of DSBs caused by ionizing radiation can be lethal.Mechanisms Of Dsb Repair · Chk1 And Chk2 Kinases As... · Synthetic Lethal Approaches...
  41. [41]
    Molecular Mechanisms of Radiation-Induced Cancer Cell Death
    We provide an updated primer on the major types of radiation-induced cell death, focussing on their molecular mechanisms, factors affecting their initiation,
  42. [42]
    Sublethal damage, potentially lethal damage, and chromosomal ...
    Special feature. Sublethal damage, potentially lethal damage, and chromosomal aberrations in mammalian cells exposed to ionizing radiations.
  43. [43]
    Radiation-Induced Chromosomal Aberrations and Immunotherapy
    May 29, 2018 · Radiation-induced chromosomal aberrations represent an early marker of late effects, including cell killing and transformation.
  44. [44]
    Ionizing Radiation-Induced Cellular Senescence in Normal, Non ...
    May 21, 2018 · Research has revealed that senescent phenotype is observable in irradiated organs. This process starts with DNA damage mediated by radiation.
  45. [45]
    Radiation-Induced Senescence Bystander Effect: The Role of ... - NIH
    Jul 27, 2020 · Ionizing Radiation (IR), especially at high doses, induces cellular senescence in exposed cultures. IR also induces “bystander effects” through signals ...
  46. [46]
    Radiation-Induced Bystander Response: Mechanism and Clinical ...
    Significance: Absorption of energy from ionizing radiation (IR) to the genetic material in the cell gives rise to damage to DNA in a dose-dependent manner.
  47. [47]
    Radiation Dictionary - CDC
    Apr 22, 2024 · Deterministic effects: effects that can be related directly to the radiation dose received. The severity increases as the dose increases. A ...
  48. [48]
    Medical management of the acute radiation syndrome - PMC
    The classic syndromes are: hematopoietic (doses >2–3 Gy), gastrointestinal (doses 5–12 Gy) and cerebrovascular syndrome (doses 10–20 Gy). There is no ...
  49. [49]
    [PDF] Safety Reports Series No. 122
    A deterministic effect can occur only if the individual radiation dose exceeds a certain threshold. At increasing doses above this threshold level, the ...
  50. [50]
    Radiation-Induced Skin Injuries to Patients: What the Interventional ...
    May 11, 2017 · These studies identify increased TEWL as an early surrogate marker for radiation dermatitis and raise the possibility that preservation of ...
  51. [51]
    Acute Radiation Syndrome: Information for Clinicians - CDC
    ARS (sometimes known as acute radiation sickness) is an acute illness caused by radiation exposure (or irradiation) of the entire body (or most of the body)
  52. [52]
    Radiobiology of the acute radiation syndrome - PMC - PubMed Central
    Jul 6, 2011 · The gastrointestinal syndrome occurs at doses of between 5 and 20 Gy and the cerebrovascular syndrome at doses higher than 20 Gy.
  53. [53]
    Radiation-Related Injuries and Their Management: An Update - PMC
    A comprehensive symptom management guideline regarding radiation dermatitis was created by the BC Cancer Agency and may be consulted for an extensive review.
  54. [54]
    Radiation-Induced Oral Mucositis - PMC - PubMed Central
    Radiation-induced oral mucositis occurs in up to 80% of head and neck cancer irradiated patients and reaches up to 100% in patients with altered fractionation ...
  55. [55]
    Lethal dose (LD) - Nuclear Regulatory Commission
    The dose of radiation expected to cause death to 50 percent of an exposed population within 30 days (LD 50/30). Typically, the LD 50/30 is in the range from ...
  56. [56]
    Linear No-Threshold Model and Standards for Protection Against ...
    Aug 17, 2021 · 99, “Low-dose extrapolation of radiation-related cancer risk,” the threshold theory posits that “there is some threshold dose below which there ...Supplementary Information · Iii. Petitioners' Assertions · Iv. Public Comments On The...
  57. [57]
    [PDF] SAT Chapter 2: Biological Effects
    Radiation effects may be stochastic, such as causing cancer or genetic effects. The probability of stochastic effects increases with dose. The NRC has adopted ...
  58. [58]
    Latency period of radiation-induced cancer - PMC - NIH
    The latency period for induction of leukemia is 5–7 years, and for solid tumours is at least 10 years, so cancers occurring earlier than this should be ...Missing: peak | Show results with:peak
  59. [59]
    Chernobyl: Chapter V. Health impact - Nuclear Energy Agency (NEA)
    These late effects, cancer induction and hereditary defects, are known as "stochastic effects" and are those effects whose frequency, not severity, is dose ...
  60. [60]
    Relative biological effectiveness - ICRPaedia
    RBE values vary with the dose, dose rate, and biological endpoint considered. In radiological protection, the RBE for stochastic effects at low doses (RBEM) is ...
  61. [61]
    Genetic Effects of Radiation - NCBI - NIH
    Ionizing radiation damages the genetic material in reproductive cells and results in mutations that are transmitted from generation to generation.
  62. [62]
    A Quick Look at Life Span Study Report 12, Part I
    Figure 1. Excess relative risk per sievert for solid cancer as a function of age for atomic-bomb survivors exposed at 10, 30, or 50 years of age.
  63. [63]
    Atomic Bomb Survivors Life-Span Study: Insufficient Statistical ... - NIH
    After sex-averaging, the excess risk should be communicated as about 2.5% per Sv on the top of ~20% mortality of the unexposed population. We should stress that ...
  64. [64]
    Clinical Radiobiology for Radiation Oncology - SpringerLink
    Sep 24, 2023 · The therapeutic window is large in radiosensitive tumor types like lymphoma, but small for other tumor types such as brain and pancreatic cancer ...
  65. [65]
    The linear quadratic model: usage, interpretation and challenges
    Dec 19, 2018 · ... late-responding tissues having α/β values in the range from 1 to 4 Gy, while early-responding tissues have α/β ratios in the range of 8–15 ...
  66. [66]
    Trends in Radiation Oncology Treatment Fractionation at a Single ...
    The mean number of fractions per treatment course for all cancers declined from 17.5 in 2010 to 13.6 in 2020. There was increased adoption of hypofractionation ...Missing: post- | Show results with:post-
  67. [67]
    Cisplatin sensitizes cancer cells to ionizing radiation via inhibition of ...
    These results demonstrate that the molecular mechanism of cisplatin sensitization of cells to IR involves inhibition DNA-PK catalyzed phosphorylation of target ...
  68. [68]
    Randomized Phase 3 Trial of the Hypoxia Modifier Nimorazole ...
    Dec 8, 2023 · Nimorazole is a hypoxic radiosensitizer with high electron affinity, which enables the drug to mimic the effect of oxygen in rendering hypoxic ...
  69. [69]
    Interfering with Tumor Hypoxia for Radiotherapy Optimization
    Jun 21, 2021 · The prototype of hypoxic radiosensitizers are the electron-affinic nitroimidazoles, such as misonidazole, etanidazole, pimonidazole and the ...
  70. [70]
    PARP inhibition radiosensitizes BRCA1 wildtype and mutated breast ...
    Dec 28, 2024 · We found that PARP inhibition increases radiosensitization independent of BRCA1 status for both protons and photons.
  71. [71]
    PARP inhibitors combined with radiotherapy: are we ready? - Frontiers
    Oct 25, 2023 · The combination of PARP inhibitors and radiotherapy can enhance the sensitivity of cancer cells to radiation, intensify DNA damage, and trigger cell death.Mechanism of anti-tumor... · Sensitization effect of PARP... · Future perspective of...
  72. [72]
    Modeling clinical outcomes in radiotherapy: NTCP, TCP ... - PubMed
    Achieving treatments with high tumor control probability (TCP) yet low normal tissue complication probability (NTCP) is often a struggle.
  73. [73]
    Intensity-modulated radiation therapy (IMRT) matches proton ...
    Sep 29, 2025 · IMRT is an advanced photon-based technique that shapes and modulates radiation beams to precisely target tumors. Photon beams effectively ...
  74. [74]
  75. [75]
    Dose limits - ICRPaedia
    Dose limits help ensure that no person is exposed to an excessive amount of radiation in normal, planned situations.Missing: 2020 ALARA
  76. [76]
    Assessment of Radiosensitivity and Biomonitoring of Exposure to ...
    Nov 28, 2019 · Two of the most used cytogenetics assays are the Dicentric Chromosome Assay ... method for simultaneous interphase-metaphase chromosome analysis ...
  77. [77]
    About Dicentric Chromosome Assays
    Dicentric Chromosome Assay is based on the principle that radiation exposure causes DNA strand breaks, in particular double strand breaks.Missing: G2 | Show results with:G2
  78. [78]
    G2 Chromosomal Radiosensitivity Assay for Testing Individual ...
    The G2 chromosomal radiosensitivity assay or, simply G2 assay, measures the number of chromatid type aberrations induced by radiation in G2 phase.Missing: assessment lymphocyte dicentric
  79. [79]
    Children Are Very Sensitive to Radiation - PMC - NIH
    The reasons for the extreme radiation sensitivity in children include: Growth means a high rate of cell division in all organ systems. Cell division is the ...
  80. [80]
  81. [81]
    Ataxia-telangiectasia - Genetics - MedlinePlus
    Sep 19, 2022 · Affected individuals are very sensitive to the effects of radiation exposure, including medical x-rays. Ataxia-telangiectasia has no cure, ...Missing: radiosensitivity | Show results with:radiosensitivity
  82. [82]
    Harmonization and strengthening of Japan's biodosimetry network ...
    Oct 21, 2025 · Purpose: The development of AI-assisted biodosimetry systems brings significant advances in cytogenetic dosimetry. The introduction of deep ...