Next Article in Journal
Development of a Polyclonal Antibody for the Immunoanalysis of Ochratoxin A (OTA) by Employing a Specially Designed Synthetic OTA Derivative as the Immunizing Hapten
Previous Article in Journal
The Emerging Mycotoxin 2-Amino-14, 16-Dimethyloctadecan-3-ol (AOD) Alters Transcriptional Regulation and Sphingolipid Metabolism and Undergoes N-Acylation by HepG2 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Aflatoxin Exposure in Immunocompromised Patients: Current State and Future Perspectives

by
Temitope R. Fagbohun
1,*,
Queenta Ngum Nji
1,
Viola O. Okechukwu
2,
Oluwasola A. Adelusi
1,
Lungani A. Nyathi
3,
Patience Awong
4 and
Patrick B. Njobeh
1,*
1
Department of Biotechnology and Food Technology, University of Johannesburg, Doornfontein Campus, Johannesburg 2094, South Africa
2
Department of Environmental, Water and Earth Sciences, Tshwane University of Technology, Arcadia Campus, Private Bag X680, Pretoria 0001, South Africa
3
Centre for Innovative Food Research (CIFR), Department of Biotechnology and Food Technology, University of Johannesburg, Doornfontein Campus, Johannesburg 2094, South Africa
4
Department of Health Sciences, University of Johannesburg, Doornfontein Campus, Johannesburg 2094, South Africa
*
Authors to whom correspondence should be addressed.
Toxins 2025, 17(8), 414; https://doi.org/10.3390/toxins17080414
Submission received: 30 April 2025 / Revised: 11 July 2025 / Accepted: 25 July 2025 / Published: 16 August 2025
(This article belongs to the Section Mycotoxins)

Abstract

Aflatoxins (AFs), harmful secondary metabolites produced by the genus Aspergillus, particularly Aspergillus flavus and Aspergillus parasiticus, are one of the best-known potent mycotoxins, posing a significant risk to public health. The primary type, especially aflatoxin B1 (AFB1), is a potent carcinogen associated with liver cancer, immunosuppression, and other health problems. Environmental factors such as high temperatures, humidity, and inadequate storage conditions promote the formation of aflatoxin in staple foods such as maize, peanuts, and rice. Immunocompromised individuals, including those with HIV/AIDS, hepatitis, cancer, or diabetes, are at increased risk due to their reduced detoxification capacity and weakened immune defenses. Chronic exposure to AF in these populations exacerbates liver damage, infection rates, and disease progression, particularly in developing countries and moderate-income populations where food safety regulations are inadequate and reliance on contaminated staple foods is widespread. Biomarkers such as aflatoxin-albumin complexes, urinary aflatoxin M1, and aflatoxin (AF) DNA adducts provide valuable insights but remain underutilized in resource-limited settings. Despite the globally recognized health risk posed by AF, research focused on monitoring human exposure remains limited, particularly among immunocompromised individuals. This dynamic emphasizes the need for targeted studies and interventions to address the particular risks faced by immunocompromised individuals. This review provides an up-to-date overview of AF exposure in immunocompromised populations, including individuals with cancer, hepatitis, diabetes, malnutrition, pregnant women, and the elderly. It also highlights exposure pathways, biomarkers, and biomonitoring strategies, while emphasizing the need for targeted interventions, advanced diagnostics, and policy frameworks to mitigate health risks in these vulnerable groups. Addressing these gaps is crucial to reducing the health burden and developing public health strategies in high-risk regions.
Key Contribution: Biomonitoring of aflatoxins is critical for assessing exposure in immunocompromised patients and other vulnerable populations and assists in risk assessment and mitigation strategies. Advances in the use of analytical techniques have improved the identification and measurement of aflatoxin biomarkers, enhancing early diagnosis and intervention efforts. Emerging mitigation strategies leading to nanotechnological solutions and detoxification approaches offer promising opportunities to reduce aflatoxin toxicity in high-risk groups.

1. Introduction

Aflatoxins, toxic secondary metabolites produced predominantly by the genera Aspergillus, particularly Aspergillus flavus (A. flavus) and Aspergillus parasiticus (A. parasiticus), are among the most potent mycotoxins and have serious public health implications [1,2]. The most important types of aflatoxins (AFs) include aflatoxin B1 (AFB1), the most potent and well-documented carcinogen among them, as well as aflatoxin B2 (AFB2), aflatoxin G1 (AFG1), and aflatoxin G2 (AFG2). Among these, AFB1 and AFG1 are considered highly toxic and mutagenic due to their strong genotoxic and hepatocarcinogenic properties [3,4]. These toxins are formed under environmental conditions such as high temperatures (25–35 °C), high humidity (≥70%), and during pre-harvest or improper storage of crops [2]. Other factors that favor their formation in agricultural products are insect infestation [5], drought stress [6], and improper food and feed handling. The biosynthesis of AF by toxic fungal species, particularly A. flavus and A. parasiticus, is a complex process regulated by a specific gene cluster in the fungal genome. Their biosynthesis occurs via a polyketide pathway in which the precursor, acetyl-CoA, undergoes enzymatic transformations to produce intermediates such as norsolorinic acid, leading to AFB1 and other variants [7,8].
Staple foods such as maize [9,10], peanuts [9,11] rice [12,13], sorghum [11,12], wheat [13,14], pearl millet [15], and milk in the form of aflatoxin M1 [10,16] are frequently contaminated with AF, leading to serious food safety and health concerns worldwide. Exposure to AF occurs primarily through ingestion, although inhalation, dermal contact, and transplacental ingestion are also possible routes [17]. Cao et al. [18] reported that the toxicity of AF is primarily mediated by its metabolic activation in the liver, where cytochrome P450 enzymes convert AFB1 into a highly reactive epoxide metabolite (AFB1-8,9-epoxide). This metabolite attaches to deoxyribonucleic acid (DNA), and forms adducts that induce mutations and genomic instability, thus contributing to carcinogenesis [18]. The negative effects of AF range from immediate toxicity to long-term health problems, such as liver cancer, gallbladder cancer, hepatotoxicity, genotoxicity, immunosuppression, growth retardation in children, and in severe cases, death [3,8,19,20].
As confirmed by Costa et al. [20], AFB1, the most dangerous and widespread AF variant, has been classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC). While AF exposure poses a health risk to the general population, immunocompromised individuals such as those living with HIV/AIDS, cancer, diabetes, chronic hepatitis, autoimmune disorders, or organ transplant recipients are particularly at risk due to their impaired immune function [21,22,23]. AF-induced immunosuppression can impair cytokine production and reduce the activity of macrophages and natural killer (NK) cells, key components of the innate immune system [3,24]. Additionally, Chu et al. [25] reported that AFB1 exposure significantly increases the risk of hepatocellular carcinoma and liver cirrhosis, especially in individuals chronically infected with the hepatitis B virus.
In regions with high HIV prevalence and poor food safety practices, the combination of HIV-related immunosuppression and AF exposure represents a dual health crisis. This includes increased susceptibility to infection and disease due to weakened immunity [26], and an increased risk of AF-related health problems such as liver damage, hepatocellular carcinoma, and other immunodeficiencies [21,27].The global burden of AF is disproportionately prevalent in low- and middle-income countries (LMICs), where regulatory frameworks for food safety are weak, and contamination of staple foods is widespread [28]. These regions also have a greater incidence of immunocompromising illnesses, such as HIV/AIDS, making the intersection of AF exposure and immunosuppression an urgent public health challenge [29].
The lack of robust surveillance systems, combined with limited access to safe food, exacerbates the risks for vulnerable populations in these areas. Efforts to detect and monitor AF exposure have primarily focused on the use of biomarkers such as AF-albumin adducts [28,30], AFM1 in urine [13] and AF DNA adducts [31,32]. While these methods provide valuable insights into the extent of exposure, their application in immunocompromised populations has not yet been sufficiently researched. More importantly, prevention strategies for AF contamination have focused on improving agricultural practices [33], biological control of AF-producing fungi [34,35], chemical detoxification of AF [35], and proper post-harvest storage to reduce the growth of AF-producing fungal species [34].
However, these measures have not been able to combat the increased risks to immunocompromised individuals. Despite the globally recognized health risk posed by AF, there has been limited research focused on monitoring AF exposure in humans, let alone in immunocompromised individuals [36]. This gap emphasizes the need for targeted studies and interventions to address the particular risks to immunocompromised individuals. Therefore, this review provides a detailed overview of AF exposure in immunocompromised populations, including people with cancer, hepatitis, diabetes, malnutrition, pregnant women, and the elderly. It also provides a thorough and detailed examination of the various pathways of AF exposure in these vulnerable populations, including ingestion, carriage from animal products, in utero and maternal transmission, and inhalation. The review also looks in detail at the commonly used matrices and biomarkers for AF detection and explores the impacts of AF exposure on diseases such as diabetes, hepatitis, HIV, and cancer. Particular emphasis is placed on the increased susceptibility of vulnerable populations, including the elderly, pregnant women, malnourished individuals, and patients with HIV, diabetes, hepatitis, or cancer, who are at increased risk of AF-induced toxicity due to their weakened immune systems. More importantly, the review highlights future perspectives and emphasizes the need for targeted interventions, advanced diagnostic tools, and policy frameworks to reduce the health impact of AFs in this high-risk group. By bridging the existing knowledge gaps, this study aims to inform research priorities and public health initiatives and ensure the development of tailored approaches to safeguard the health of immunocompromised individuals in the face of this persistent threat.

2. Biosynthetic Pathway of AFs in Aspergillus Species

The biosynthesis of AFs occurs via a complex polyketide pathway, involving a series of oxidative, reductive, cyclization, and methylation reactions. According to Liao et al. [37], Ma et al. [38], Okechukwu et al. [8], and Yang et al. [39], the pathway begins with the condensation of acetyl-CoA and several units of malonyl-CoA through the activity of a polyketide synthase (PKS) enzyme complex encoded by the aflC (also known as pksA) gene. This reaction leads to the formation of the first stable intermediate, norsolorinic acid (NOR), a red pigment and an important early marker in aflatoxin biosynthesis. Norsolorinic acid (NOR) is reduced by aflD (nor-1) to form averantin (AVN). Subsequent hydroxylation and oxidation reactions mediated by enzymes such as aflG (avnA) and aflH lead to the formation of averufin (AVF). AVF is then further modified via several intermediate steps involving key enzymes such as aflI (ver-1), aflV (cypX), and aflW (moxY) to form versiconal hemiacetal acetate (VHA), which cyclizes into versicolorin A (VERA). VERA is a crucial branching intermediate that ultimately leads to the formation of demethylsterigmatocystin (DMST) and subsequently to sterigmatocystin (ST) through oxidative reactions. ST, an intrinsically toxic compound, is then o-methylated by O-methyltransferase A (aflP, also known as omtA) to O-methylsterigmatocystin (OMST).
The final steps of the pathway involve a series of oxidation and rearrangement reactions catalyzed by enzymes such as aflQ (ordA) and aflU (cypA), which convert OMST into the four major AFs: AFB1 and AFG1, followed by the reduced forms AFB2 and AFG2. The structural differences between B- and G-type AFs are due to variations in the oxidation state and the lactone ring structure [4]. AFB1 is the most potent and widely studied aflatoxin, known for its strong hepatocarcinogenic properties in humans and animals. This biosynthetic pathway operates within a large gene cluster spanning approximately 70 kilobases and includes around 25–30 genes, many of which encode enzymes for each step of the pathway [40]. Although this section focuses on the biosynthetic sequence, it is important to recognize that the expression of these genes is highly dependent on environmental factors such as temperature, moisture content, substrate composition, and oxidative stress. Understanding the enzymatic steps and key intermediates in this pathway provides a basis for developing strategies to monitor, detect, and mitigate AF contamination in agricultural products.

3. AF Exposure Pathways in Humans

Despite extensive research and ongoing efforts to tighten food safety regulations, exposure to mycotoxins remains high worldwide. An estimated five billion people remain at risk, with populations in sub-Saharan Africa disproportionately affected [41]. This increased vulnerability is largely attributed to favorable climatic conditions for fungal growth, recurrent food insecurity, and poor implementation of legislation to control mycotoxin contamination in food, with some regions of Asia having been regularly exposed [42]. Although people can come into contact with mycotoxins through various pathways (see Figure 1), the global extent of this exposure is still not well understood. This data gap limits our ability to fully understand health implications. To date, more than 400 mycotoxins have been discovered; only a small number are considered a significant threat to public health and agriculture, particularly in sub-Saharan Africa [43,44]. The tropical environment of the region, with its harsh and erratic climate, combined with traditional farming methods, makes the crop highly susceptible to mold infestation. This, in turn, leads to the formation of toxic compounds such as AF [45]. Cereal grains are one of the main food categories that are particularly susceptible to AF, but dried fruit, nuts, coffee, and spices are also affected. Intake of AF-contaminated foods and/or animal-derived foods (such as milk and eggs), tissues, and dairy products is the main pathway of AF exposure, with a significant proportion of AF being absorbed dermally and through inhalation. Otherwise, direct exposure to AFs occurs via consumption of contaminated food/feed and indirect exposure via animal foods such as tissues, eggs, milk, and other dairy products from animals fed with AF-contaminated feed as well as through dermal contact and inhalation of mold-contaminated dust.

3.1. Exposure to AF Through Ingestion

Aflatoxin exposure represents a substantial public health challenge, particularly in regions with inadequate food safety controls and suboptimal storage conditions. Aflatoxin risk assessment is a structured and methodical approach aimed at evaluating the potential health hazards associated with exposure to this group of food contaminants. The assessment process generally comprises four key components: hazard identification, exposure assessment, dose-response assessment, and risk characterization [46]. According to Kibugu et al. [47], hazard identification involves determining the adverse health effects associated with AFs, particularly AFB1. Exposure assessment quantifies the levels and frequency of aflatoxin ingestion through diet across various populations. The dose-response assessment establishes the relationship between the magnitude of exposure and the likelihood of specific toxicological outcomes, such as hepatotoxicity and liver cancer [48]. Risk characterization integrates data from the previous steps to estimate the overall health risk, with particular attention to vulnerable populations, including young children, the elderly, and immunocompromised individuals.
Exposure to AF through ingestion remains a significant global food safety concern, particularly in areas where staple foods are susceptible to fungal contamination. In 2015, the World Health Organization (WHO) stated that approximately 600 million people become ill each year due to contaminated food, resulting in 420,000 deaths and the loss of 33 million healthy life years [49]. Among the greatest threats to food safety are mycotoxins, secondary metabolites produced by toxic fungi such as Aspergillus, Penicillium, and Fusarium species [50]. In response, various nations have established stringent regulations to limit mycotoxin exposure. The Joint FAO/WHO Expert Committee on Food Additives estimates the average dietary exposure to AFs at 0.93 to 2.45 ng/kg/day in Europe, 3.5 to 180 ng/kg/day in Africa, and 0.3 to 53 ng/kg/day in Asia [51]. Africa in particular has the highest incidence of liver cancer, which is largely due to chronic ingestion of AFs. Previous estimates suggest that around 250,000 deaths in China and Africa are due to hepatocellular carcinoma [52]. Dietary exposure to AFs is influenced by consumption habits, processing methods, and geographical factors. Staple foods such as maize, peanuts, and other grains are highly susceptible to AF exposure, especially in low-income regions. For instance, researchers have indicated extremely high rates of AF contamination in food (see Table 1). Alberts et al. [53] indicate that the daily maize intake in some rural communities reaches 1–2 kg per person, which significantly increases the AF load. AFs have also been detected in processed foods, including fermented beverages [54], and cheese [55]. Despite the widespread assumption that food processing eliminates mycotoxins, studies confirm that AFs exhibit high thermal and chemical stability, rendering conventional food processing methods insufficient for complete detoxification [56,57]. Consequently, contamination by AF in various foods remains an ongoing concern emphasizing the need for continuous monitoring and effective remediation strategies.

3.1.1. Exposure Through Carryover from Animal Sources

Farmers often feed mold-contaminated and physically damaged crops to livestock, assuming they are unsuitable for human consumption but still usable as animal feed. However, these crops may contain high levels of mycotoxins, particularly AFs, which pose serious health risks to animals [74,75]. AFs. Various types of mycotoxins have been detected in animal feed, including AFs, which are usually associated with cereals and nuts that make up the majority of feed ingredients or formulations [76]. Depending on the animal class and/or species, there is a possibility that microorganisms in the rumen affect the concentration of AFs transferred to animal tissues, leading to consumer exposure through the consumption of tissues contaminated with AFs [77,78]. Monogastric species, for example, are more susceptible to the effects of AFs compared to ruminants.
This is because the microbiota in the rumen of ruminants degrades most AFs [79]. Nevertheless, the ability of the rumen to detoxify AFs may also be limited within species, age, as well as sex, resulting in some AFs accumulating in animal tissues and representing a potential source of exposure for humans if these contaminated tissues are consumed [80]. Therefore, the proportion of AFs in the consumable tissues of ruminants is comparatively low, as some of the AFs are degraded in the rumen, while others are processed in the liver, and therefore have no significant impact on human exposure compared to non-ruminants [80]. AFs ingested by animals can either be excreted in urine, excreted in milk, or assimilated into tissues. As mentioned above, AFs are low in edible tissues, and milk contaminated with mycotoxins has been identified as one of the main sources of human exposure [81,82].
However, the degree of carryover is also influenced by dietary factors such as feeding, intake rate, digestion rate, health status of the livestock, age, sex, ability to biotransform in the liver, husbandry systems, seasons, geographical location, and environmental conditions that affect the extent of AF carryover in animals. Consumption of eggs and meat from animals fed with AF-contaminated feed is another possibility of indirect exposure. Numerous research papers have documented the effects of AFs on fish [83,84,85], and AF-contaminated feed for fish in aquaculture has been reported [86]. When such AF-contaminated feed is consumed, humans and other animals are exposed to AFs.

3.1.2. In Utero and Maternal Exposure

Research has shown that exposure to AFs in utero can occur via the transplacental route [38,87]. Although breast milk is rich in nutrients and immune factors, it can also be a potential source of exposure to AFs for infants. The hydroxylated derivative of AF (AFM1) is found in breast milk 12–24 h after consumption of AF-contaminated foods. Magoha et al. [88] analyzed the breast milk of 143 Tanzanian lactating mothers for AFM1 and reported 100% contamination. Similarly, Mahdavi et al. [89] reported AFM1 levels in the breast milk of Iranian breastfeeding mothers who had exclusively breastfed their children. Waseem et al. [51] found that 99.5% of the breast milk of 445 mothers from Abu Dhabi was contaminated with AFM1, and 36% of Egyptian mothers were contaminated with AFM1. High concentrations of ochratoxins and AFs were also identified in Italian mothers. Infant formula was a source of AF contamination.
Exposure in utero and through the mother is therefore a feasible source of human exposure to AFs. The first 1000 days spent alive, starting from conception to 24 months of age, are an important period for the growth and development of the child. Therefore, food intake during pregnancy plays a fundamental role in the future health of the child when it comes to the mother’s consumption of food contaminated with AFs [90,91]. In addition, children of weaning age are considered a high-risk group for AF exposure. Foods suitable for weaning include corn and peanuts which have been reported to be particularly susceptible to mycotoxin contamination (Table 1).

3.2. Exposure to AF Through Inhalation

Many studies have shown that exposure to AFs is not always related to food consumption and that the work and residential environments (see Table 2) are also potential sources of AF exposure exposure [92,93,94,95,96,97]. The nasal passage is the principal target for many inhaled toxins, with the epithelial mucosa being the first to be damaged by these inhaled spores or AFs [98]. Systemic effects of AF through inhalation of some AFs have been reported such as lung cancer, whose mechanism of carcinogenicity is due to oxidative DNA damage [95,98,99]. Although this is the least studied route of exposure, some reports have shown that breathing in a portion of AFs may pose greater risks than ingestion. Exposure to AFs in the environment or workplace is mainly through inhalation. Airborne dust often contains fungal spores and hyphae fragments and can transport AFs to the lungs via inhalation, which is common in the workplace [98,100]. Work environments with poor ventilation subject employees to an increased danger of aflatoxin contamination [93]. Environmental exposure to AF through molds in water-damaged buildings is well known. Various mycotoxins have been detected in human biopsies of lung, brain, and liver tissue and body fluids of people who have been exposed to toxin-producing molds in their environments [76].

3.3. Exposure to AF Through Dermal Contact

As diet is the primary factorial method regarding exposure to AF, exposure from the work environment or occupation should not be disregarded as in some cases it may also contribute directly to the overall exposure. When AF-containing dust particles come into contact with the skin, either through contaminated work surfaces or in situations where workers must frequently handle AF-contaminated materials such as food or feed without protection, exposure to AFs is likely [111].
Workplaces, especially in agriculture and the food industry, including tasks like loading, handling, or milling of AF-tainted substances (grain, waste, and feed), as well as feed processing plants, are good examples of aflatoxin exposure sites, as most aflatoxin concentrations are 10 times higher than in human food. Work environments with inappropriate protective clothing and equipment put workers at higher risk of AF contamination [81]. Taevernier et al. [112] have shown that mycotoxins can penetrate the skin and cause skin damage. Therefore, sporadic exposure to AF also increases the risk of cell death in the epidermis, skin cancer, and immune-related diseases [111].

4. AF Biomarkers

AFs are among the most commonly detected fungal toxins in food and animals, resulting in their status as the most biomonitored mycotoxins [113,114,115,116]. The metabolism of AFs, particularly AFB1, along with biochemical reactions such as hydroxylation and epoxidation, determines the resultant forms of these compounds. AFM1 is a thoroughly studied metabolite and a confirmed indicator of AFB1 exposure, detectable in various biological liquids such as urine and breast milk [117]. Wild and Gong [118] highlighted several biomarkers used to monitor AF exposure, including urinary metabolites such as AFM1, AFB1-N7-Gua, AFP1, AFQ1, and AFB1-mercapturic acid, as well as AF-albumin adducts [119].
Understanding the sources of AF exposure and their metabolic pathways is essential for the advancement of appropriate biomarkers for exposure estimation. Furthermore, the effective use of biomarkers necessitates the availability of detailed information regarding individual AF toxicokinetics, bioavailability, and metabolism to accurately interpret the results [120]. Human data on toxicokinetic studies for various toxins are limited, as most studies have predominantly been conducted in animal models; consequently, exposure assessment using biomarkers presents a quantitative approach fraught with significant uncertainties [121]. Biomonitoring includes the detection of AFs in readily accessible biological liquids like blood, milk, feces, and urine (see Figure 2). The main benefit of these biomarkers rests in their capacity to assess AF exposure levels within biological matrices, which integrate all previously identified sources of [122,123]. The detection of biomarkers can be conducted using one of the following approaches: direct biomarkers, which involve the application of particular standardized methods that are refined and confirmed methods utilizing parent compounds as reference substances [124], indirect biomarker quantification, which entails assessing non-specific structural or functional alterations in the body induced by exposure to certain drugs or toxins; or non-targeted biomarkers, which involve the measurement of unknown aflatoxin derivatives.
Biomarkers for AF exposure have been identified in various biological fluids (see Table 3) and tissues, including milk, plasma, saliva, feces, hair, nails, liver, kidneys, and lungs [125]. Among these biomarkers, urine is the most utilized matrix, not only because it contains a wide array of AFs but also due to the ease of obtaining urine samples through non-invasive methods [126]. Different biomarkers are suitable for varying purposes and contexts; for instance, breast milk is particularly effective for evaluating aflatoxin exposure in breastfed infants, whereas other biological fluids, such as serum and plasma, require more invasive collection methods and trained medical personnel, making them more applicable for long-term exposure studies [126]. Thus, the biomonitoring of AFs in mycotoxin exposure can be conducted across diverse vulnerable groups, thereby reducing assumptions about dietary exposure [127]. Determining the precise level of AFs within bodily fluids highlights the ambiguity associated with the presence of emerging or modified AF types that might go unnoticed by traditional analytical methods, particularly those mycotoxins lacking legal limits and/or defined biomarkers, which present additional challenges. The following section will outline commonly used biomarkers, along with their respective advantages and disadvantages.

4.1. Urine

AFs are excreted in urine as metabolites following the ingestion of contaminated food. After absorption in the gastrointestinal tract, AFs, particularly AFB1, are transported to the liver, where they undergo biotransformation primarily by cytochrome P450 enzymes [8,128]. This hepatic metabolism converts AFB1 into several hydroxylated metabolites, including aflatoxin M1 (AFM1), aflatoxin Q1 (AFQ1), and aflatoxin P1 (AFP1). These metabolites are subsequently released into the bloodstream, filtered by the kidneys, and excreted in urine as part of the body’s detoxification and elimination processes [129].
Urine provides a convenient, non-invasive, and efficient matrix for the biomonitoring of aflatoxin exposure in humans [130,131]. Following the ingestion of aflatoxin-contaminated food, the compounds undergo hepatic metabolism, resulting in the formation of metabolites such as AFM1, AFQ1, AFP1, and AFB1-N7-Gua, which are subsequently excreted in urine. Quantification of these metabolites allows for an accurate assessment of recent aflatoxin exposure. Due to the ease of urine collection, this approach is particularly suitable for large-scale epidemiological studies, including those involving high-risk groups such as children, the elderly, HIV-infected individuals, and malnourished populations [132,133]. To enhance the reliability of exposure estimates, urinary aflatoxin concentrations are commonly normalized to creatinine levels, thereby accounting for interindividual variability in physiological factors such as age, sex, body mass, and hydration status [134,135]. Collectively, urine-based biomonitoring offers a robust, accessible, and scientifically sound method for evaluating aflatoxin exposure across diverse demographic cohorts.
Hydroxylation at the C-7 position results in the production of AFM1, while epoxidation at the C-8 and C-9 positions results in either internal or external configurations. The exo-epoxidized type of AFB1 can react with DNA and glutathione or undergo hydrolysis to form AFB-diol [96]. Specifically, AFB1-N7-guanine and AFB1-glutathione conjugates are excreted in urine as AFB1-mercapturic acid conjugates, serving as indicators of exposure [136,137]. Panel et al. [138] also reported measurable levels of AFB1-N7-guanine in urine samples. Additionally, other metabolites such as AFM1, AFQ1, and AFBN7-Gua are excreted in both feces and urine as a result of AFB1 biotransformation mediated by specific cytochrome P450 isoforms. To assess exposure, urinary AF concentrations are converted to levels of intake that account for their kinetics, body weight, and the daily output of urine, primarily characterized by creatinine levels [134,136,137].
Variability in urine volume among individuals can influence the levels of AFs and their metabolites that are excreted, with daily fluctuations in aflatoxin intake posing a significant challenge [134]. This issue can be mitigated by normalizing aflatoxin levels to creatinine concentrations, as the AF/creatinine ratio allows for comparisons between individuals, accounting for factors such as muscle mass, sex, age, and season [139]. Thus, urinary creatinine concentrations serve to adjust the urinary concentrations of AFs or their metabolites. Furthermore, the analysis of AFs and their metabolites in urine samples requires low LOQs, which can be achieved through targeted analysis and/or enzymatic treatment of urine samples with β-glucuronidase/sulfatase to convert conjugates back to the parent compound [115].

4.2. Blood

Blood-based biomonitoring is a critical tool for evaluating long-term aflatoxin exposure. Repeated ingestion of AFs leads to the formation of aflatoxin–albumin adducts, which circulate in the bloodstream for extended periods due to the stability and longevity of albumin [3,140]. Quantifying these adducts in serum or plasma provides a reliable indicator of chronic exposure. This technique is particularly beneficial for assessing exposure in high-risk populations, such as individuals with cancer, diabetes, or HIV, who may exhibit increased susceptibility to the adverse effects of AFs [138]. While conventional blood sampling is invasive, the use of dried blood spot (DBS) methods offers a minimally invasive and more practical alternative, especially in remote or resource-limited settings where access to clinical infrastructure is restricted. DBS facilitates effective population-level monitoring without requiring extensive medical equipment or personnel.
AFs are produced by fungi like Aspergillus flavus in contaminated food. After ingestion, AFs are absorbed in the gut and transported to the liver. In the liver, they are metabolized by cytochrome P450 enzymes into various toxic and non-toxic forms. Some metabolites, such as AFM1, AFQ1, AFP1, and AFB1-8,9-epoxide, enter the bloodstream. In blood, AFs can bind to albumin, forming aflatoxin–albumin adducts used as biomarkers. These circulating metabolites are later excreted in urine or bile.
AF-albumin adducts have long been used as a substitute efficacy biomarker of AFB1 exposure for assessment due to the long half-life of albumin in humans [141,142]. Reports indicate that over 95% of blood samples in various regions of West Africa contain AF-albumin adducts as biomarkers, encompassing individuals of all ages. The advantages of these biomarkers include their higher levels of toxins, making them particularly useful in long-term exposure studies [126,143]. While serum and plasma are not commonly employed as routine biomarkers due to the invasive techniques required for collection, a non-invasive method for detecting AFB1-albumin adducts from dried blood spots has been reported [144].

4.3. Breast Milk

Breast milk plays a vital role in assessing recent exposure to AFs, especially AFM1, which appears following maternal ingestion of AFB1-contaminated food. This is particularly concerning for breastfeeding infants, whose immature detoxification systems make them highly vulnerable to toxin exposure [145]. Measuring aflatoxin levels in breast milk enables accurate evaluation of exposure in both lactating mothers and their infants, groups often overlooked in traditional dietary assessments [146]. Consequently, breast milk monitoring provides critical insights into aflatoxin-related health risks for neonates and young infants, supporting targeted interventions to protect these sensitive populations.
Lactating mothers and their infants constitute a particularly vulnerable population due to the potential transfer of AFs from the maternal system to breast milk. When a lactating woman consumes aflatoxin-contaminated food, mainly AFB1, it is absorbed and metabolized in the liver. The liver converts AFB1 into AFM1, a hydroxylated metabolite. AFM1 is then transported through the bloodstream to the mammary glands. It is excreted into breast milk, especially within 12–24 h of exposure [90,147]. AFM1 in breast milk is heat-stable and toxic, though less potent than AFB1. The presence of AFM1 in breast milk provides a direct route of exposure for breastfeeding infants, who are especially susceptible due to their developing organs and immature detoxification systems. Monitoring AFs in breast milk is critical for assessing exposure levels in lactating mothers and their infants, guiding public health interventions, and mitigating associated risks [148,149]. Detecting and quantifying AFs in breast milk necessitates sensitive and reliable analytical techniques.
Breast milk serves as a valuable matrix for estimating the exposure of breastfed infants to AFs; however, its limitation lies in its applicability solely to lactating women. AFM1 is the predominant biomarker of exposure, having been detected in both milk and urine during exposure assessments [150]. However, while breast milk analysis provides critical data on infant exposure, its applicability is limited to lactating women, and the concentration of AFM1 in milk is influenced by maternal dietary habits, metabolic efficiency, and interindividual differences in detoxification pathways [80,140]. Detecting and quantifying AFM1 in breast milk requires highly sensitive and reliable analytical techniques such as HPLC, ELISA, and LC-MS/MS. These methods ensure accurate assessment of exposure levels, contributing to risk mitigation strategies aimed at protecting maternal and infant health.

4.4. Feces

AFs are excreted in feces as unmetabolized toxins and metabolites. After ingestion, AFs are absorbed and metabolized mainly in the liver. Some unmetabolized AFs and their metabolites are secreted into the bile which transports these compounds into the intestines [151]. From the intestines, AFs and metabolites are eliminated via feces.
Fecal biomarkers enable the assessment of recent aflatoxin exposure by detecting unmetabolized toxins and their metabolites excreted via the gastrointestinal tract. This non-invasive approach is practical for large-scale population studies; however, its validity and applicability in vulnerable populations, such as the elderly, pregnant women, and children, remain to be fully established. The analyses and effective method for detecting AFs and their metabolites in humans and animals provide valuable insights into short-term dietary exposure and metabolism [152]. AFB1, the most toxic and prevalent aflatoxin, is often detected in feces in its unmetabolized form, serving as a direct indicator of recent dietary intake [147,151]. In addition to AFB1, feces contain a range of metabolites, including AFQ1 and AFM1, which result from hepatic biotransformation processes. Notably, higher levels of AFQ1 are often detected in feces compared to AFM1, indicating a significant excretory route for metabolism [147,151]. Since feces reflect unabsorbed AFs and metabolic byproducts excreted through bile, fecal biomarkers provide a complementary assessment of systemic exposure and detoxification efficiency [152].
The presence of AFM1 is particularly relevant for populations consuming AF-contaminated food, as it highlights both dietary intake and metabolic processing capacity [153,154]. The detection of these adducts is invaluable for assessing genotoxicity, as persistent exposure to AFs has been linked to hepatocellular carcinoma (HCC) and other adverse health effects [138,155]. Using fecal biomarkers is especially advantageous for large-scale biomonitoring studies, as sample collection is non-invasive, and fecal matter provides a stable medium for mycotoxin analysis. Recent studies have demonstrated the potential of fecal biomarkers to reflect both direct dietary intake and metabolic processing of AFs.
For instance, a 2024 study investigated the effects of AFB1 on human intestinal Caco-2 cells, identifying critical genes affected by AFB1 exposure [156]. The study highlighted changes in gene expression related to cell proliferation, migration, apoptosis, and metabolism, suggesting that these genetic alterations could serve as biomarkers for AF contamination detection. Integrating fecal biomarker assessments with other biological matrices, such as urine and blood, enhances the accuracy of exposure assessments and improves the understanding of interindividual variability in AF metabolism and excretion. As analytical techniques such as LC-MS and HPLC continue to advance, fecal biomarker analysis will remain a critical tool in mycotoxin research and public health monitoring.
Table 3. Matrices and AFs biomarkers commonly used in exposure assessment.
Table 3. Matrices and AFs biomarkers commonly used in exposure assessment.
MatrixBiomarkerTechniqueSample Size% ContaminationConcentration Range (ppb)References
UrineAFM1HPLC7229.170.00067–0.00787[157]
UrineAFM1HPLC6978.260.0006–0.0399[158]
UrineAFM1LC-MS/MS220140.06–4.7[159]
FecesAFB1PLE and HPLC-MS/MS310.02[160]
UrineAFM1HPLC30027.670.01–0.33[161]
UrineAFM1LC-ESI-MS/MS14510.340.17–1.38[162]
UrineAFB1-N7-GuaHPLC-MS/MS20800.9–7.2[163]
UrineAFB1-N7-GuaHPLC2740.746.6–494.9[164]
UrineAFM1ELISA20557.560.00020–0.0193 creatinine[165]
UrineAFs2-D TLC601000.07–0.2[166]
UrineAFM1HPLC50640.008–0.801[167]
UrineAFM1ELISA9347.31500–59,900 creatinine[168]
UrineAFM1ELISA16061.25LOD-0.0747[169]
UrineAFM1HPLC-ESI-MS/MS175-0.005-0.5[170]
UrineAFM1HPLC-MS/MS2810.74LOD-0.33[171]
UrineAFM1LC-MS/MS12014.20.3–1.5[172]
PlasmaAFB1-lysLC-MS/MS26019.610.5–74.5[115]
PlasmaAFB1
AFB2
LC-MS/MS6039.4
74.91
1.23–4.56
1.16–3.75
[173]
PlasmaAFB1-lysLC-Orbitrap5823.450.2–59.2[174]
SerumAFB1-lysLC-FLDLC-MS/MS34831.08–102.6[31]
PlasmaAFB1-lysLC-MS3246.88-[175]
SerumAFB1-lysLC-MS160610.80–20.24[176]
SerumAFB1-lysLC-FLD2201000.71–95.6[177]
SerumAFB1-lysLC-FLD34799.4-[178]
SerumAFB1-lysLC-FLD8841006.04–8.90[179]
SerumAFB1-lysLC-MS4611000.2–814.8[180]
PlasmaAFB1-lysLC-MS/MS60723.5–6.6[181]
PlasmaAFB1-lysLC-MS/MS85--[181]
PlasmaAFB1-lysLC-MS/MS167620.04–123.5[182]
PlasmaAFB1-lysELISA1151003.9–458.4[183]
SerumAFB1-lysELISA23067-[184]
SerumAFB1-lysELISA30588.2-[185]
PlasmaAFB1-lysELISA37495-[186]
SerumAFB1
AFM1
LC-MS/MS21323
50
0.0–0.73
0.0–1.91
[187]
SerumAFB1-lysLC-FLD713900.4–168[188]
PlasmaAFB1-lysELISA16667–984.7–23.50[189]
SerumAFB1-albELISA34983.0–35.1[168]
Blood/serumAFB1-albELISA24100LOD-32.8[190]
Blood/serumAFs2-D TLC601000.15–0.38[166]
Blood/serumAFB1-albHPLC5071000.44–268.73[191]
Blood/serumAFB1-albELISA25098LOD-66[192]
Blood/serumAFB1-albLC-MS/MS597780.02–211[193]
Blood/serumAFB1-albHPLC170970.2–23.16[194]
Blood/serumAFM1HPLC131390.3–56[195]
Blood/serumAFB1-albELISA1191004.8–260.8[196]
Blood/serumAFB1-albHPLC17020.61.01–16.57[197]
BreastmilkAFM1HPLC388365.5–5131[198]
BreastmilkAFM1HPLC7510060.9–299.9[199]
BreastmilkAFB1HPLC7510094.5–4123.8[199]
BreastmilkAFM1HPLC140925–3400[200]

5. AF Biomonitoring in Immunocompromised Individuals

Biomonitoring of AFs in at-risk populations involves the detection of AF metabolites in biological samples such as urine, serum, and breast milk [201]. Techniques such as LC-MS, HPLC, and ELISA have been widely used for this purpose [202]. The diagram (Figure 3) illustrates key factors contributing to the susceptibility of immunocompromised individuals to AF toxicity. It categorizes vulnerable groups, including pregnant women, HIV patients, diabetes patients, cancer patients, and elderly individuals, highlighting their specific risk factors that compromise detoxification and immune responses to AF exposure. Therefore, regular biomonitoring can provide insights into the exposure levels of immunocompromised individuals helping in formulating dietary interventions to reduce risks [203,204].

5.1. Elderly Individuals

The natural decline in immune function with age is one of the main contributors to the vulnerability of elderly individuals to the adverse effects of mycotoxins. The role of an intact immune system and barrier function in the successful elimination of pathogenic fungi and their toxic metabolites cannot be overemphasized [205]. Mycotoxins are implicated in facilitating a decline in the body’s natural defenses. For example, the A. flavus toxins AFB1 and AFM1 compromise the intestinal epithelial cell barrier by inhibiting intestinal cell growth and viability. Since these epithelial cells act as the body’s primary defense against fungal toxins, their disruption facilitates inappropriate immune responses such as chronic inflammation of vital organs [206]. Additionally, disturbances in the enzyme and antimicrobial agent secretory functions of epithelial cells and poor nutrient retention have been reported by Gonkowski et al. [207].
This double burden, namely the natural decline in immune function and the additional compromise caused by mycotoxin exposure, places the elderly at a heightened risk of developing severe health complications. Biomonitoring in this population becomes critical as an opportunity to identify early signs of exposure and implement timely interventions. Strategies such as routine screening for mycotoxin biomarkers and dietary adjustments to reduce exposure can help mitigate health risks [141]. However, a critical limitation of existing studies is the exclusion of elderly individuals over 60 years of age. Available studies tend to group all adults into a single category, that is, those aged 18 years or older. This limits insights into mycotoxin exposure in the elderly immunocompromised population, which is the focus of this review.
In a mycotoxin exposure biomonitoring study by Patriarca et al. [201] using the urinary multi-biomarker approach, different analytes were detected using LC–ESI–MS/MS in 110 out of 175 samples collected from Cameroonian adults (up to 58 years of age) in Bamenda and Yaoundé. Among the mycotoxins identified, AFM1 was a key compound detected, contributing significantly to the overall percentage of analytes found in urine samples. Notably, an HIV-positive adult in the study was found to be exposed to a mixture of multiple mycotoxins, including AFM1, highlighting the relevance of this particular mycotoxin in the exposure assessment. An assessment of the food consumption practices of the same study population revealed the consumption of maize and cassava-based products such as corn fufu, garri, rice, plantain, cocoyam, pumpkin, and potatoes. These dishes are generally accompanied by groundnut, egusi, or vegetable soup. Typically, combined maize and groundnut-based foods were consumed once to five times a week in both Bamenda and Yaoundé. Maize and groundnuts are mycotoxin-vulnerable foods, which explains the high mycotoxin exposure rates in the elderly in these two regions of Cameroon [201]. Based on the roughly calculated amounts of mycotoxins ingested per day from these foods, the urinary levels of AFM1 were estimated to exceed the recommended tolerable daily intake (TDI) limits proposed by the 2003 and 2013 Scientific Committee on Food.
In another study by Huang et al. [198], urine specimens from 227 adults in China aged 20–88 years were analyzed for mycotoxin exposure. The biomarker for AFB1 and AFM1 was noted at a low prevalence in only 2.20% of the specimens (mean ± SD: 0.35 ± 0.17 ppb). Despite the low occurrence, given that AFM1 is a class 1 carcinogen and highly toxic even at lower concentrations, it is still necessary to monitor and reduce these levels to non-detectable concentrations. While young adults (20–45 years), middle-aged individuals (46–64 years), and senior adults (65–88 years) are all prone to mycotoxin contamination, this same study showed that mycotoxins were more frequently detected in young adults, possibly due to a greater food intake compared to older adults. The relatively low prevalence and concentration of toxins in samples from young adults could be due to a high detoxification rate of toxins, which limits their bioavailability [208]. This may suggest a decline in detoxification capacity and efficiency with age. Interestingly, females were found to be more prone to AFM1 exposure than males. Correlation studies between urinary AFM1 concentrations and food consumption indicated that exposure was linked to the consumption of aflatoxin-prone foods, such as seeds and nuts.

5.2. Pregnant Women

Breast milk serves as a valuable biological matrix for assessing the vulnerability of infants to mycotoxin contamination. However, urine and plasma samples are also effective for monitoring mycotoxin exposure during pregnancy. Exposure to multiple mycotoxins in early pregnancy can pose significant health and developmental risks to the fetus, as these toxins can cross the placental barrier [209]. For instance, in utero exposure has been linked to low birth weight and, in some cases, stunted growth. Specifically, mycotoxin exposure is associated with differential gene methylation, affecting growth and immune-related genes, therefore potentially impairing the child’s development and immune function [210].
A 2018–2019 study of 447 pregnant women in rural Bangladesh revealed exposure to AFs, among other mycotoxins of interest in the study. While AFs were not among the most prevalent in the study population, only 17 of the samples were clear of mycotoxin exposure. These figures indicate widespread exposure of rural populations in Bangladesh to mycotoxins, which, if unmonitored, can have adverse effects on pregnancy outcomes. Market studies in Bangladesh have revealed high consumption of mycotoxin-prone foods such as rice, wheat flour, groundnuts, spices, dates, and milk, with mycotoxin levels in some exceeding U.S. maximum regulatory thresholds [211]. Comprehensive mycotoxin control strategies must be prioritized across the entire value chain to mitigate exposure risks effectively. This includes promoting effective agricultural practices, proper post-harvest management, storage, and transportation to inhibit fungal growth and mycotoxin production contamination. Additionally, enforcing country-specific regulatory limits, conducting regular surveillance, and raising public awareness of mycotoxins and their impacts are crucial to ensuring food safety.
Simultaneous detection of mycotoxins using LC-MS/MS in biological fluids, serum (n = 71), urine (n = 18), and amniotic fluid (n = 21) samples collected from pregnant women in Alessandria, Italy, demonstrated that mycotoxins could be present in fetal-maternal biological fluids. However, the detected levels were too low to pose significant risks to the mother or fetus. Specifically, AFG1 was detected in one serum and four urine samples and AFB1 and AFB2 were detected in two urine samples [212]. In contrast, an assessment of biological samples from 98 pregnant women in Egypt revealed a mean serum AF-albumin (AF-Alb) contamination of 4.9 pg/mg albumin in 34 of the samples and a urinary AFM1 mean concentration of 19.7 pg/mg creatinine in 44 of 93 samples. Essentially, AFs were detected in 41% of the subjects studied. In another study, an ELISA analysis of 80 human breast milk samples obtained from clinics in Jordan revealed AFM1 contamination in most samples, with only three falling below the maximum tolerance standards established by the European Union and U.S. regulations [213]. These findings highlight the variation in AF exposure across different regions, emphasizing the need for country-specific regulations and tailored control strategies to effectively address localized risks. Also, the prevalence of AFs in breast milk poses a major risk for the conveyance of toxins to infants, potentially leading to serious health issues.
Southern African countries are among the regions most at risk for mycotoxin contamination due to the frequent consumption of cereal-based foods and groundnuts. However, research on exposure assessment and biomonitoring of AFs among pregnant women in this area appears to be limited. Smith et al. [180] conducted a study to determine aflatoxin exposure among pregnant women in Zimbabwe by measuring urinary AFM1 levels. Urine specimens were gathered from 1580 participants in rural regions, specifically the Chirumanzu and Shurugwi regions in the Midlands Province, and analyzed for AFM1 using direct ELISA. The study found that 484 women, or 30%, were exposed to aflatoxin, with AFM1 mean values ranging from 31 to 6046 pg/mg. The greatest levels of exposure were identified in areas with lower elevations and limited rainfall. The consumption of high-risk items like maize, peanuts, and milk was linked to the observed mycotoxin exposure rates in this study [180].

5.3. Malnourished Individuals

Malnutrition, particularly in children, remains a critical health burden in developing economies. The widespread prevalence of AF exposure in these regions further exacerbates the problem, as these toxins impair nutrient absorption, compromise immune function, and contribute to stunted growth and developmental delays [206,214]. A study conducted in Nigeria involving 58 children (aged 6 to 48 months) from both rural and urban communities revealed alarming findings [174]. Among these children, 47 were diagnosed with severe acute malnutrition or undernutrition (SAM), with 21 suffering from marasmus and 26 suffering from kwashiorkor: additionally, 43 exhibited stunting. Notably, 81% of these children had measurable levels of AFB1-Lys adducts in their serum, with concentrations ranging from 0.2 to 59.2 pg/mg albumin. Children with SAM and stunting had significantly higher concentrations compared to the control group without malnutrition. Furthermore, AFB1-Lys adduct levels varied notably between children with kwashiorkor and those with marasmus, with kwashiorkor patients exhibiting the highest concentrations. Although AF exposure may not directly cause kwashiorkor or marasmus, evidence suggests that malnutrition significantly impairs the body’s ability to detoxify mycotoxins [181]. This is supported by the lower mycotoxin concentrations observed in non-malnourished children, as reported by [174].
On the contrary, Gong et al. [215] reported a connection between chronic AF exposure and growth impairment and underweight in children from Benin and Togo. Interestingly, AF levels were low in breastfed infants, suggesting that children are most vulnerable to mycotoxin contamination post-weaning, primarily through contaminated solid foods. This highlights an urgent need for integrated strategies to address both malnutrition and mycotoxin exposure in children to improve health outcomes, especially in vulnerable populations.

5.4. AF Biomonitoring in HIV Individuals

Biomonitoring of AF exposure in HIV-infected individuals is essential for assessing potential health risks and understanding its implications for disease progression. Biomarkers such as AF-albumin adducts provide valuable insights into long-term exposure, particularly in resource-limited settings where dietary contamination with AFs is common [216,217]. The connection between AF exposure and HIV progression has been increasingly studied, with evidence suggesting that chronic exposure may exacerbate immune suppression, increase viral replication, and contribute to associated comorbidities.
Recent studies highlight the high prevalence of AF exposure among HIV-infected populations, particularly in sub-Saharan Africa. For instance, a cross-sectional study conducted in Nigeria reported that 95.9% of 196 HIV-positive patients had detectable levels of urinary AFM1, indicating widespread exposure [218]. Similarly, research by Jolly et al. [219] demonstrated that high baseline AFB1 albumin adduct levels were significantly linked to lower CD4 T-cell counts in ART-naïve HIV patients, with an average count of 66.5 cells/µL and a statistically significant p-value of 0.043. These findings suggest that AF exposure may accelerate HIV-related immunosuppression, contributing to increased susceptibility to opportunistic infections, liver dysfunction, and complications associated with disease progression.
The interaction between AFs and HIV infection is mediated through multiple mechanisms. AFs have well-documented immunosuppressive properties that impair immune cell function, further weakening the already compromised immune system of HIV-positive individuals [220]. The compromised immune response may lead to faster progression of HIV-related complications. This suppression can reduce the body’s ability to fight opportunistic infections, leading to more severe disease outcomes. Additionally, some studies suggest that AF exposure is associated with increased HIV viral replication, potentially due to the disruption of cellular pathways involved in viral regulation [221]. Furthermore, AFs induce DNA damage through the formation of reactive metabolites that bind to DNA, leading to mutations. In the context of HIV, this mutagenic potential may influence viral evolution and contribute to the development of drug resistance, complicating antiretroviral therapy (ART) efficacy. AF exposure is also known to contribute to malnutrition, which can further impair immune function and reduce ART effectiveness, making disease management more challenging. The health implications of aflatoxin exposure in HIV patients are significant. Immunosuppression caused by AFs increases the risk of opportunistic infections, leading to higher hospitalization rates and greater healthcare burdens. Additionally, the interaction between AFs and HIV may complicate treatment strategies, as patients with higher aflatoxin exposure could experience reduced ART effectiveness, necessitating individualized treatment modifications. Chronic exposure to AFs also raises concerns regarding long-term health outcomes, particularly the heightened risk of hepatocellular carcinoma, a concern for HIV-positive individuals who are already at an elevated risk for liver disease.
Given these risks, routine biomonitoring of AF exposure in HIV patients, especially in endemic regions, is critical. Implementing public health strategies such as improved food storage practices, dietary diversification, and targeted interventions to reduce aflatoxin contamination could help mitigate these risks [222]. Further research is needed to establish definitive causal relationships and explore intervention strategies aimed at minimizing aflatoxin exposure in HIV-vulnerable populations.

5.5. AF Biomonitoring in Diabetes Patients

Diabetes mellitus, characterized by chronic hyperglycemia, is associated with immune dysfunction and an increased susceptibility to infections. The immunocompromised state of diabetic individuals makes them particularly vulnerable to environmental and dietary toxins such as AF, that have been implicated in a variety of negative health impacts, such as immunosuppression, hepatotoxicity, and carcinogenesis [223,224]. AFB1 is recognized as a potent hepatocarcinogen and metabolic disruptor, and its effects in diabetic populations warrant careful investigation. Given that diabetes is already linked to a higher risk of metabolic disorders and infection-related complications, AF exposure may further amplify these health risks, creating a cycle of worsening disease outcomes [223,225]. The interaction between AF and diabetes involves multiple biochemical pathways that can exacerbate disease progression. One of the primary concerns is the impact of AFs on glycemic control. AF exposure has been connected to disruptions in glucose metabolism and insulin signaling, leading to insulin resistance and increased difficulty in maintaining blood sugar levels [226,227]. This disruption can exacerbate hyperglycemia in diabetic patients, complicating their condition [223].
Additionally, AFs induce oxidative stress, which damages pancreatic β-cells responsible for insulin production, thereby impairing insulin secretion and contributing to hyperglycemia [23,228]. Long-term exposure to AFs has also been shown to elicit inflammatory responses, further exacerbating insulin resistance and metabolic dysfunction [229]. The inflammatory cytokines produced in reaction to aflatoxin exposure can aggravate systemic inflammation, which is already a key factor in diabetes pathogenesis, leading to further metabolic imbalances.
In addition to metabolic disturbances, AF exposure may contribute to an increased risk of infections in diabetic patients. Given that diabetes itself impairs immune function, the immunosuppressive effects of AFs may further compromise the body’s capacity to combat infections, raising the likelihood of complications such as diabetic ketoacidosis and poor wound healing [225].
Moreover, long-term exposure to AFs has been linked to an elevated risk of cardiovascular disease, neuropathy, and retinopathy—complications that are already prevalent in diabetic populations [230,231]. Another critical concern is the potential interaction between AFs and antidiabetic medications. AF exposure may interfere with the metabolism and bioavailability of these drugs, reducing their efficacy and necessitating adjustments in treatment regimens [223]. Additionally, diabetic individuals are already at a higher likelihood of developing certain cancers, and long-term AF exposure further amplifies this risk, particularly for hepatocellular carcinoma [232].
The intersection of diabetes and AF exposure poses a considerable public health issue, especially in areas with high dietary AF contamination. Tackling this problem necessitates a multifaceted approach that includes routine biomonitoring of AF exposure in diabetic individuals, particularly those in high-risk regions. Public health initiatives should focus on food safety measures, improved agricultural practices, and dietary diversification to reduce AF exposure. Increased awareness among healthcare providers and patients is also essential for promoting preventive strategies and early intervention. Educational campaigns targeting both the medical community and the public can help improve understanding of the risks associated with AF exposure and encourage proactive measures to mitigate its impact [232]. While existing evidence highlights the adverse effects of AFs in diabetic individuals, further research is needed to fully elucidate the molecular mechanisms underlying these interactions and to develop targeted strategies for reducing exposure and associated health risks.

5.6. AF Biomonitoring in Cancer Patients

Cancer is defined as a complex medical condition characterized by the uncontrolled growth of cells, leading to tumor formation and, in many cases, metastasis to other organs [233]. The progression of cancer is influenced by various genetic, environmental, and lifestyle factors, including exposure to dietary carcinogens such as AFs. AFs, particularly AFB1, are potent hepatocarcinogens that undergo metabolic activation in the liver, leading to the formation of reactive epoxides that bind to DNA and induce mutagenic changes [2,8,234]. These interactions contribute to cellular damage, inflammation, and oxidative stress, all of which play a role in tumor initiation and progression. Cancer patients, particularly those undergoing chemotherapy or radiotherapy, are immunocompromised and may be more susceptible to the harmful effects of AF exposure due to their reduced ability to detoxify and repair cellular damage. The carcinogenic effects of AFs are mediated through their metabolic activation by cytochrome P450 enzymes in the liver, which convert this aflatoxin (AFB1) to highly reactive intermediate forms [2,8].
These metabolites interact with DNA, RNA, and proteins, leading to the creation of DNA adducts that trigger mutations and genomic instability, raising the chances of oncogenesis [235]. AF-induced DNA damage can further contribute to the disruption of critical cellular processes, including apoptosis and cell cycle regulation, thereby promoting the survival of malignant cells. Moreover, AFs induce oxidative stress through the excessive production of reactive oxygen species (ROS) that harm cellular macromolecules, disrupt metabolic pathways, and promote chronic inflammation [236,237]. This inflammatory environment facilitates tumor progression by enhancing cellular proliferation and angiogenesis, further supporting cancer development.
The immunosuppressive effects of AFs compound the challenges faced by cancer patients, as these toxins impair immune surveillance and reduce the body’s ability to eliminate pre-cancerous and cancerous cells [3,238]. Cancer patients undergoing chemotherapy or radiotherapy are particularly vulnerable, as these treatments already diminish the immune system’s effectiveness, making them more vulnerable to infections and other environmental toxins. Additionally, AFs can interfere with the metabolism of chemotherapeutic drugs, potentially reducing their efficacy and complicating treatment outcomes [239]. The interaction between AFs and other carcinogenic substances, such as alcohol and certain dietary components, may also contribute to an increased cancer risk by enhancing mutagenic potential and promoting tumorigenesis [240,241,242].
Furthermore, nutritional deficiencies common in cancer patients, including deficiencies in vitamins A, C, and E, may impair the detoxification of AFs, exacerbating oxidative stress and DNA damage [240,241,242].
The consequences for public health of AF exposure in cancer patients are significant, particularly in regions where food contamination is prevalent. Awareness and education about the risks associated with AF-contaminated food are crucial for minimizing exposure, particularly for immunocompromised individuals [3,239]. Strengthening food safety regulations and monitoring AF levels in food supplies are essential measures to protect at-risk populations, including cancer patients [243]. Further studies are necessary to better understand the effect of AF exposure on cancer progression and treatment efficacy, as well as to develop targeted strategies for reducing exposure and mitigating associated health risks. Implementing biomonitoring programs that assess AF levels in cancer patients could provide valuable insights into the relationship between dietary aflatoxin intake and disease outcomes, ultimately contributing to improved clinical management and public health interventions.

5.7. AF Biomonitoring in Hepatitis Patients

AF biomonitoring involves the detection and quantification of AF metabolites in biological samples like blood, urine, and tissues [244]. Hepatitis B or C patients are at heightened risk due to their weakened immune systems [245]. AFB1, the most potent hepatotoxic variant, has been linked to liver disease progression, leading to increased morbidity and mortality among these populations [3]. The hepatotoxic effects of AFs are primarily mediated through their metabolic activation in the liver. Upon ingestion, AFB1 undergoes bioactivation through the cytochrome P450 enzyme, leading to the development of highly reactive epoxide metabolites that form adducts with DNA and proteins [3]. This DNA damage triggers mutagenic processes, increasing the risk of liver cancer [2,8]. Additionally, AFs contribute to immune suppression, further impairing the body’s capacity to fight infections and exacerbating disease progression in hepatitis patients.
Various analytical techniques are employed for biomonitoring, including enzyme-linked immunosorbent assays (ELISA), high-performance liquid chromatography (HPLC), and mass spectrometry (MS) techniques [246]. These methods facilitate accurate exposure assessment, allowing for early detection and intervention in high-risk individuals (Table 4). Recent studies indicate a significant correlation between AF exposure and liver disease progression among hepatitis patients [247]). Research conducted in West Africa demonstrated that chronic hepatitis patients had significantly elevated levels of AF metabolites compared to healthy controls, emphasizing the importance of focused monitoring strategies in these vulnerable populations [248]. The integration of biomonitoring into routine clinical assessments could improve disease management and mitigate AF-associated health risks in immunocompromised individuals. Effective monitoring programs can identify high-risk populations and inform targeted interventions to minimize exposure to AFs [249,250]. Furthermore, strategies such as improved food storage practices, stricter regulatory frameworks, and enhanced surveillance systems can mitigate AF contamination.
Finally, raising awareness among healthcare professionals and at-risk patients, while strengthening collaborations between public health authorities, researchers, and clinicians, is essential for developing effective policies to mitigate AF exposure and enhance health outcomes in hepatitis patients.

6. Conclusions

AF biomonitoring in immunocompromised and high-risk populations is critical for assessing exposure risks and associated health outcomes. Elderly individuals, pregnant women, malnourished individuals, and patients with HIV, diabetes, cancer, or hepatitis are highly susceptible to aflatoxin-induced toxicity due to compromised immune function and metabolic vulnerabilities. AFs, particularly AFB1, are potent hepatotoxins and carcinogens, exacerbating liver damage in hepatitis and cancer patients while increasing the risk of hepatocellular carcinoma. In malnourished individuals, aflatoxin exposure impairs nutrient absorption and immune response, further worsening health outcomes. Advanced detection techniques, including LC-MS, ELISA, and HPLC, have improved AF identification in biological matrices, yet challenges remain in defining exposure thresholds and understanding chronic low-dose effects.

7. Future Perspectives and Recommendations

There is a wide range of immunocompromised individuals, but this review focuses on the most commonly affected groups: pregnant women, the elderly, cancer patients, individuals with diabetes, and those living with HIV. To enhance future research, additional vulnerable populations such as organ transplant recipients, patients with chronic kidney or liver disease, malnourished individuals, and young children should be considered to better address the broader challenges of immunosuppression. Addressing aflatoxin exposure in immunocompromised patients will require integrated strategies aimed at preventing further exposure, enhancing detoxification, and supporting immune function. Future research should prioritize real-time, non-invasive detection methods using sensors and wearable technologies for continuous biomonitoring. Nanotechnology is revolutionizing the detection of AFs through the development of highly sensitive methods. Nanosensors, utilizing materials such as gold nanoparticles and carbon nanotubes, enhance the detection of AFs in food and biological samples. These sensors offer rapid and efficient results, making them ideal for on-site testing, which is particularly beneficial for farmers and food safety inspectors. CRISPR technology is being repurposed for aflatoxin detection. This cutting-edge method exploits CRISPR’s precision to identify specific DNA sequences associated with aflatoxin-producing fungi. As a result, contamination can be detected early and accurately, thereby facilitating more effective food safety management.
Significant advancements are being made in the development of portable devices (Portable Analytical Devices) designed for on-site AF testing. These devices typically employ microfluidics and lab-on-a-chip technology, enabling efficient analysis without the complexity of traditional laboratory setups. This innovation is particularly advantageous for farmers and food safety inspectors in rural settings, allowing for immediate food safety assessments. However, artificial intelligence and machine learning are increasingly being applied to predict aflatoxin contamination in crops. By analyzing environmental variables and historical data, these technologies contribute to risk assessment and management. This proactive strategy enables farmers to implement preventive measures before contamination occurs, thus protecting both their crops and public health. Innovative biosensors are being developed that integrate biological recognition elements with electronic signal transducers to detect AFs, even at low concentrations. These sophisticated sensors can be tailored to target specific AFs and facilitate real-time monitoring of food safety, providing reassurance to both consumers and producers. Research is ongoing into novel detoxification methods, such as the use of biochar and other adsorbents, to address AF contamination in agricultural products. These techniques have the potential to significantly reduce aflatoxin levels in contaminated foods, enhancing their safety for consumption. Additionally, researchers are investigating genetic engineering as a strategy to develop crop varieties that are resistant to AFs. By modifying the genes that make crops vulnerable to aflatoxin-producing fungi, scientists aim to minimize contamination at its source, thereby promoting safer food production.
Omics-based approaches like metabolomics and proteomics can help elucidate biochemical changes caused by AFs, while longitudinal studies are needed to establish dose-response relationships and refine regulatory limits. Understanding the mechanisms of aflatoxin-induced immunosuppression is essential, especially with emerging AF-producing fungi such as A. nomius and A. pseudotamarii, which call for epidemiological studies and control measures. Climate change is expected to worsen aflatoxin contamination, emphasizing the need for climate-resilient agricultural practices and predictive models. Innovative solutions using nanotechnology for detection and prevention, along with advances in personalized medicine, can further enhance mitigation strategies. The development of rapid point-of-care diagnostic tests for aflatoxin biomarkers will facilitate timely interventions, while detoxification methods and nutritional support may reduce toxicity in vulnerable groups. Additionally, future efforts should focus on developing effective chemoprotective agents and antioxidants to reduce liver damage and oxidative stress. Early detection and monitoring of liver health and cancer risk will be crucial, as will tailored nutritional support to strengthen immunity and optimized management of underlying conditions like HIV. Improving patient education on food safety and implementing routine aflatoxin screening in high-risk populations will also be key to reducing exposure. Advances in these areas will help protect vulnerable groups from the harmful effects of AFs.

Author Contributions

Conceptualization, T.R.F. and O.A.A.; methodology, T.R.F., Q.N.N., O.A.A., V.O.O., L.A.N. and P.A.; validation, T.R.F., Q.N.N., O.A.A., V.O.O., L.A.N. and P.A.; investigation, T.R.F., Q.N.N., O.A.A., V.O.O., L.A.N. and P.A.; resources, T.R.F., Q.N.N., O.A.A., V.O.O., L.A.N. and P.A.; writing—original draft preparation, T.R.F., Q.N.N., O.A.A., V.O.O., L.A.N. and P.A.; visualization, V.O.O.; writing–review & editing, T.R.F., O.A.A., V.O.O. and L.A.N.; supervision P.B.N.; project administration, P.B.N.; funding acquisition, P.B.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Maize Trust, grant number MTM23-01 and the APC was funded by the University of Johannesburg, South Africa.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We would like to extend our heartfelt thanks to the Mycotoxin Laboratory for the technical support. unwavering support in bringing this review manuscript to completion. Your assistance has truly been invaluable to our research journey.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Adelusi, O.A.; Gbashi, S.; Adebo, J.A.; Aasa, A.O.; Oladeji, O.M.; Kah, G.; Adebo, O.A.; Changwa, R.; Njobeh, P.B. Seasonal and geographical impact on the mycotoxigenicity of Aspergillus and Fusarium species isolated from smallholder dairy cattle feeds and feedstuffs in Free State and Limpopo provinces of South Africa. Toxins 2023, 15, 128. [Google Scholar] [CrossRef]
  2. Okechukwu, V.O.; Kappo, A.P.; Njobeh, P.B.; Mamo, M.A. Morphed aflaxotin concentration produced by Aspergillus flavus strain VKMN22 on maize grains inoculated on agar culture. Food Chem. Mol. Sci. 2024, 8, 100197. [Google Scholar] [CrossRef] [PubMed]
  3. Benkerroum, N. Chronic and acute toxicities of Aflatoxins: Mechanisms of action. Int. J. Environ. Res. Public Health 2020, 17, 423. [Google Scholar] [CrossRef] [PubMed]
  4. Ramadan, N.A.; Al-Ameri, H.A. Aflatoxins, in Aflatoxins-Occurrence, Detoxification, Determination and Health Risks; IntechOpen: London, UK, 2022. [Google Scholar]
  5. Danso, J.K.; Mbata, G.N.; Holton, R.L. Preharvest insect pests of peanuts and associated aflatoxin contaminants in Georgia, USA. J. Econ. Entomol. 2024, 117, 993–1000. [Google Scholar] [CrossRef] [PubMed]
  6. Aminou, M.M.; Falalou, H.; Abdou, H.; Mendu, V. Aflatoxin B1 Contamination Association with the Seed Coat Biochemical Marker Polyphenol in Peanuts Under Intermittent Drought. J. Fungi 2024, 10, 850. [Google Scholar] [CrossRef]
  7. Muaz, K.; Manzoor, S.; Akhtar, S.; Riaz, M.; Amir, M.; Akram, K.; Ismail, A. Aflatoxin Biosynthesis. In Aflatoxins in Food: A Recent Perspective; Springer: Berlin/Heidelberg, Germany, 2022; pp. 19–40. [Google Scholar]
  8. Okechukwu, V.O.; Adelusi, O.A.; Kappo, A.P.; Njobeh, P.B.; Mamo, M.A. Aflatoxins: Occurrence, biosynthesis, mechanism of action and effects, conventional/emerging detection techniques. Food Chem. 2024, 436, 137775. [Google Scholar] [CrossRef]
  9. Boadu, R.O.; Dankyi, E.; Apalangya, V.A.; Osei-Safo, D. Aflatoxins in maize and groundnuts on markets in Accra and consumers risk. Food Addit. Contam. Part B 2024, 17, 213–222. [Google Scholar] [CrossRef]
  10. Kos, J.; Radić, B.; Radović, R.; Šarić, B.; Jovanov, P.; Šarić, L. Aflatoxins in maize, milk and dairy products from Serbia. Food Addit. Contam. Part B 2024, 17, 296–307. [Google Scholar] [CrossRef]
  11. Falade, T.D.; Neya, A.; Bonkoungou, S.; Dagno, K.; Basso, A.; Senghor, A.L.; Atehnkeng, J.; Ortega-Beltran, A.; Bandyopadhyay, R. Aflatoxin contamination of maize, groundnut, and sorghum grown in Burkina Faso, Mali, and Niger and aflatoxin exposure assessment. Toxins 2022, 14, 700. [Google Scholar] [CrossRef]
  12. Wenndt, A.; Sudini, H.K.; Pingali, P.; Nelson, R. Exploring aflatoxin contamination and household-level exposure risk in diverse Indian food systems. PLoS ONE 2020, 15, e0240565. [Google Scholar] [CrossRef]
  13. Xia, L.; Routledge, M.N.; Rasheed, H.; Ismail, A.; Dong, Y.; Jiang, T.; Gong, Y.Y. Biomonitoring of aflatoxin B1 and deoxynivalenol in a rural pakistan population using ultra-sensitive LC-MS/MS method. Toxins 2020, 12, 591. [Google Scholar] [CrossRef]
  14. Heshmati, A.; Mozaffari Nejad, A.S.; Mehri, F. Occurrence, dietary exposure, and risk assessment of Aflatoxins in wheat flour from Iran. Int. J. Environ. Anal. Chem. 2023, 103, 9395–9408. [Google Scholar] [CrossRef]
  15. Houissa, H.; Lasram, S.; Sulyok, M.; Šarkanj, B.; Fontana, A.; Strub, C.; Krska, R.; Schorr-Galindo, S.; Ghorbel, A. Multimycotoxin LC-MS/MS analysis in pearl millet (Pennisetum glaucum) from Tunisia. Food Control 2019, 106, 106738. [Google Scholar] [CrossRef]
  16. Tadesse, S.; Berhanu, T.; Woldegiorgis, A.Z. Aflatoxin M1 in milk and milk products marketed by local and industrial producers in Bishoftu town of Ethiopia. Food Control 2020, 118, 107386. [Google Scholar] [CrossRef]
  17. Mathew, G.; Pingle, S. Assessing Aflatoxin Exposure: Exploring Types of Exposure and Diverse Biomarkers—A Minireview. Curr. Fungal Infect. Rep. 2024, 18, 163–171. [Google Scholar] [CrossRef]
  18. Cao, W.; Yu, P.; Yang, K.; Cao, D. Aflatoxin B1: Metabolism, toxicology, and its involvement in oxidative stress and cancer development. Toxicol. Mech. Methods 2022, 32, 395–419. [Google Scholar] [CrossRef]
  19. Nabwire Wangia-Dixon, R.; Xue, K.S.; Alcala, J.; Quach, T.H.T.; Song, X.; Tang, L.; Ombaka, J.; Githanga, D.P.; Anzala, O.A.; Wang, J.-S. Nutrition and growth outcomes are affected by aflatoxin exposures in Kenyan children. Food Addit. Contam. Part A 2020, 37, 2123–2134. [Google Scholar] [CrossRef]
  20. Costa, J.; Lima, N.; Santos, C. An overview on possible links between aflatoxin B1 exposure and gallbladder cancer. Mycotoxin Res. 2021, 37, 205–214. [Google Scholar] [CrossRef]
  21. Otedo, A.; Simbiri, K.O.; Were, V.; Ongati, O.; Estambale, B.A. Risk factors for liver Cancer in HIV endemic areas of Western Kenya. Infect. Agents Cancer 2018, 13, 41. [Google Scholar] [CrossRef]
  22. Kabak, B. Aflatoxins in foodstuffs: Occurrence and risk assessment in Turkey. J. Food Compos. Anal. 2021, 96, 103734. [Google Scholar] [CrossRef]
  23. Akash, M.S.H.; Haq, M.E.U.; Qader, A.; Rehman, K. Biochemical investigation of human exposure to aflatoxin M1 and its association with risk factors of diabetes mellitus. Environ. Sci. Pollut. Res. 2021, 28, 62907–62918. [Google Scholar] [CrossRef]
  24. Saha Turna, N.; Comstock, S.S.; Gangur, V.; Wu, F. Effects of aflatoxin on the immune system: Evidence from human and mammalian animal research. Crit. Rev. Food Sci. Nutr. 2024, 64, 9955–9973. [Google Scholar] [CrossRef] [PubMed]
  25. Chu, Y.J.; Yang, H.I.; Wu, H.C.; Liu, J.; Wang, L.Y.; Lu, S.N.; Lee, M.H.; Jen, C.L.; You, S.L.; Santella, R.M.; et al. Aflatoxin B1 exposure increases the risk of cirrhosis and hepatocellular carcinoma in chronic hepatitis B virus carriers. Int. J. Cancer 2017, 141, 711–720. [Google Scholar] [CrossRef] [PubMed]
  26. Cribbs, S.K.; Crothers, K.; Morris, A. Pathogenesis of HIV-related lung disease: Immunity, infection, and inflammation. Physiol. Rev. 2020, 100, 603–632. [Google Scholar] [CrossRef] [PubMed]
  27. Boni, S.; Beed, F.; Kimanya, M.; Koyano, E.; Mponda, O.; Mamiro, D.; Kaoneka, B.; Bandyopadhyay, R.; Korie, S.; Mahuku, G. Aflatoxin contamination in Tanzania: Quantifying the problem in maize and groundnuts from rural households. World Mycotoxin J. 2021, 14, 553–564. [Google Scholar] [CrossRef]
  28. Rasheed, H.; Xu, Y.; Kimanya, M.E.; Pan, X.; Li, Z.; Zou, X.; Shirima, C.P.; Holmes, M.; Routledge, M.N.; Gong, Y.Y. Estimating the health burden of aflatoxin attributable stunting among children in low income countries of Africa. Sci. Rep. 2021, 11, 1619. [Google Scholar] [CrossRef]
  29. Phan, J.M.; Kim, S.; Linh, Đ.T.T.; Cosimi, L.A.; Pollack, T.M. Telehealth interventions for HIV in low-and middle-income countries. Curr. HIV/AIDS Rep. 2022, 19, 600–609. [Google Scholar] [CrossRef]
  30. Saltzmann, J.; Xu, Y.; Gong, Y.Y.; Lindahl, J.; Kersten, S.; Dänicke, S.; Routledge, M.N. Preliminary study on the relationship between aflatoxin-bovine serum albumin adducts in blood and aflatoxin M1 levels in milk of dairy cows. Mycotoxin Res. 2020, 36, 207–211. [Google Scholar] [CrossRef]
  31. Diaz de Leon-Martinez, L.; Rodríguez-Aguilar, M.; Wong-Arce, A.; Díaz-Barriga, F.; Bañuelos-Hernández, B.; Rosales-Mendoza, S.; Flores-Ramírez, R. Evaluation of acute and chronic exposure to aflatoxin B1 in indigenous women of the Huasteca Potosina, Mexico. Environ. Sci. Pollut. Res. 2020, 27, 30583–30591. [Google Scholar] [CrossRef]
  32. Autrup, H.; Autrup, J.L. Human exposure to aflatoxins-biological monitoring. In Handbook of Applied Mycology; CRC Press: Boca Raton, FL, USA, 2024; pp. 213–230. [Google Scholar]
  33. Gichohi-Wainaina, W.N.; Kumwenda, N.; Zulu, R.; Munthali, J.; Okori, P. Aflatoxin contamination: Knowledge disparities among agriculture extension officers, frontline health workers and small holder farming households in Malawi. Food Control 2021, 121, 107672. [Google Scholar] [CrossRef]
  34. Marshall, H.; Meneely, J.P.; Quinn, B.; Zhao, Y.; Bourke, P.; Gilmore, B.F.; Zhang, G.; Elliott, C.T. Novel decontamination approaches and their potential application for post-harvest aflatoxin control. Trends Food Sci. Technol. 2020, 106, 489–496. [Google Scholar] [CrossRef]
  35. Shabeer, S.; Asad, S.; Jamal, A.; Ali, A. Aflatoxin contamination, its impact and management strategies: An updated review. Toxins 2022, 14, 307. [Google Scholar] [CrossRef]
  36. Yusuf Aliyu, A.; Adeleke, O.A. Latest Progress on Tuberculosis and HIV Co-Infection: A Closer Look at People of Different Ages. Adv. Ther. 2025, 8, 2400033. [Google Scholar] [CrossRef]
  37. Liao, J.; He, Z.; Xia, Y.; Lei, Y.; Liao, B. A review on biosynthesis and genetic regulation of aflatoxin production by major Aspergillus fungi. Oil Crop Sci. 2020, 5, 166–173. [Google Scholar] [CrossRef]
  38. Ma, L.; Li, X.; Xing, F.; Ma, J.; Ma, X.; Jiang, Y. Fus3, as a critical kinase in MAPK cascade, regulates aflatoxin biosynthesis by controlling the substrate supply in Aspergillus flavus, rather than the cluster genes modulation. Microbiol. Spectr. 2022, 10, e01269-21. [Google Scholar] [CrossRef] [PubMed]
  39. Yang, K.; Tian, J.; Keller, N.P. Post-translational modifications drive secondary metabolite biosynthesis in Aspergillus: A review. Environ. Microbiol. 2022, 24, 2857–2881. [Google Scholar] [CrossRef] [PubMed]
  40. Smit, S.J.; Lichman, B.R. Plant biosynthetic gene clusters in the context of metabolic evolution. Nat. Prod. Rep. 2022, 39, 1465–1482. [Google Scholar] [CrossRef] [PubMed]
  41. Dickerson, S.; Cannon, M.; O’Neill, B. Climate change risks to human development in sub-Saharan Africa: A review of the literature. Clim. Dev. 2022, 14, 571–589. [Google Scholar] [CrossRef]
  42. Awuchi, C.G.; Ondari, E.N.; Ogbonna, C.U.; Upadhyay, A.K.; Baran, K.; Okpala, C.O.R.; Korzeniowska, M.; Guiné, R.P. Mycotoxins affecting animals, foods, humans, and plants: Types, occurrence, toxicities, action mechanisms, prevention, and detoxification strategies—A revisit. Foods 2021, 10, 1279. [Google Scholar] [CrossRef]
  43. Ashby, E.; Nicholson, A.; Haag, T. The Role of Plant Agricultural Practices on Development of Antimicrobial Resistant Fungi Affecting Human Health; Proceedings of a Workshop Series; National Academies Press: Washington, DC, USA, 2023; pp. 43–56. [Google Scholar]
  44. Marchante, H.; Palhas, J.; Núñez, F.A.L.; Marchante, E. Invasive species impacts and management. In Life on Land; Springer: Cham, Switzerland, 2021; pp. 560–571. [Google Scholar]
  45. Nji, Q.N.; Babalola, O.O.; Ekwomadu, T.I.; Nleya, N.; Mwanza, M. Six Main Contributing Factors to High Levels of Mycotoxin Contamination in African Foods. Toxins 2022, 14, 318. [Google Scholar] [CrossRef]
  46. Williams, P.R.; Paustenbach, D.J. Risk characterization. In Human and Ecological Risk Assessment: Theory and Practice; John Wiley & Sons: Hoboken, NJ, USA, 2024; Volume 1, pp. 263–331. [Google Scholar]
  47. Kibugu, J.; Munga, L.; Mburu, D.; Maloba, F.; Auma, J.E.; Grace, D.; Lindahl, J.F. Dietary mycotoxins: An overview on toxicokinetics, toxicodynamics, toxicity, epidemiology, detection, and their mitigation with special emphasis on aflatoxicosis in humans and animals. Toxins 2024, 16, 483. [Google Scholar] [CrossRef]
  48. Moffett, D.B.; Mumtaz, M.M.; Sullivan, D.W., Jr.; Whittaker, M.H. General considerations of dose-effect and dose-response relationships. In Handbook on the Toxicology of Metals; Academic Press: Cambridge, MA, USA, 2022; pp. 299–317. [Google Scholar]
  49. FAO/WHO. Joint FAO/WHO Food Standards Programme Codex Committee on Contaminants in Foods; WHO, Geneva 9th Session; WHO: Geneva, Switzerland, 2015. [Google Scholar]
  50. Marrez, D.A.; Ayesh, A.M. Mycotoxins: The threat to food safety. Egypt. J. Chem. 2022, 65, 353–372. [Google Scholar] [CrossRef]
  51. Waseem, A.; Ahmad Shah, S.; Sajjad, A.; Rauf Siddiqi, A.; Nafees, M. Human Exposure to Mycotoxins: A Retrospective Review of Leading Toxins and Metabolites in Human Biological Matrices. J. Chem. Soc. Pak. 2014, 36, 1196. [Google Scholar]
  52. Partnership for Aflatoxin Control in Africa (PACA) Secretariat, PACA Strategy: Overview of Intervention and Results. African Union Commission. 2020. Available online: https://archives.au.int/handle/123456789/5549 (accessed on 25 March 2025).
  53. Alberts, J.; Lilly, M.; Rheeder, J.; Burger, H.-M.; Shephard, G.S.; Gelderblom, W. Technological and community-based methods to reduce mycotoxin exposure. Food Control 2017, 73, 101–109. [Google Scholar] [CrossRef]
  54. Misihairabgwi, J.; Cheikhyoussef, A. Traditional fermented foods and beverages of Namibia. J. Ethn. Foods 2017, 4, 145–153. [Google Scholar] [CrossRef]
  55. Zebib, H.; Abate, D.; Woldegiorgis, A.Z. Aflatoxin M1 in raw milk, pasteurized milk and cottage cheese collected along value chain actors from three regions of Ethiopia. Toxins 2022, 14, 276. [Google Scholar] [CrossRef] [PubMed]
  56. Medina, A.; Akbar, A.; Baazeem, A.; Rodriguez, A.; Magan, N. Climate change, food security and mycotoxins: Do we know enough? Fungal Biol. Rev. 2017, 31, 143–154. [Google Scholar] [CrossRef]
  57. Authority, E.F.S.; Maggiore, A.; Afonso, A.; Barrucci, F.; Sanctis, G.D. Climate change as a driver of emerging risks for food and feed safety, plant, animal health and nutritional quality. EFSA Support. Publ. 2020, 17, 1881E. [Google Scholar]
  58. Martins, C.; Assunção, R.; Cunha, S.C.; Fernandes, J.O.; Jager, A.; Petta, T.; Oliveira, C.A.; Alvito, P. Assessment of multiple mycotoxins in breakfast cereals available in the Portuguese market. Food Chem. 2018, 239, 132–140. [Google Scholar] [CrossRef]
  59. Akello, J.; Ortega-Beltran, A.; Katati, B.; Atehnkeng, J.; Augusto, J.; Mwila, C.M.; Mahuku, G.; Chikoye, D.; Bandyopadhyay, R. Prevalence of aflatoxin-and fumonisin-producing fungi associated with cereal crops grown in Zimbabwe and their associated risks in a climate change scenario. Foods 2021, 10, 287. [Google Scholar] [CrossRef]
  60. Agriopoulou, S.; Stamatelopoulou, E.; Varzakas, T. Advances in analysis and detection of major mycotoxins in foods. Foods 2020, 9, 518. [Google Scholar] [CrossRef] [PubMed]
  61. Mngqawa, P.; Shephard, G.; Green, I.; Ngobeni, S.; De Rijk, T.; Katerere, D. Mycotoxin contamination of home-grown maize in rural northern South Africa (Limpopo and Mpumalanga Provinces). Food Addit. Contam. Part B 2016, 9, 38–45. [Google Scholar] [CrossRef] [PubMed]
  62. Omotayo, O.P.; Omotayo, A.O.; Babalola, O.O.; Mwanza, M. Comparative study of aflatoxin contamination of winter and summer ginger from the North West Province of South Africa. Toxicol. Rep. 2019, 6, 489–495. [Google Scholar] [CrossRef] [PubMed]
  63. Adekoya, I.; Obadina, A.; Adaku, C.C.; De Boevre, M.; Okoth, S.; De Saeger, S.; Njobeh, P. Mycobiota and co-occurrence of mycotoxins in South African maize-based opaque beer. Int. J. Food Microbiol. 2018, 270, 22–30. [Google Scholar] [CrossRef]
  64. Odhav, B.; Naicker, V. Mycotoxins in South African traditionally brewed beers. Food Addit. Contam. 2002, 19, 55–61. [Google Scholar] [CrossRef]
  65. Ndisio, B.; Wachira, P.; Kagot, V.; Okoth, S. Susceptibility of locally cultivated groundnut (Arachis hypogaea) varieties to aflatoxin accumulation in Homa Bay County, Kenya. Afr. J. Microbiol. Res. 2017, 11, 1329–1337. [Google Scholar] [CrossRef]
  66. Dada, T.A.; Ekwomadu, T.I.; Mwanza, M. Multi-mycotoxin determination in dried beef using liquid chromatography coupled with triple quadrupole mass spectrometry (LC-MS/MS). Toxins 2020, 12, 357. [Google Scholar] [CrossRef]
  67. Daou, R.; Afif, C.; Joubrane, K.; Khabbaz, L.R.; Maroun, R.; Ismail, A.; El Khoury, A. Occurrence of aflatoxin M1 in raw, pasteurized, UHT cows’ milk, and dairy products in Lebanon. Food Control 2020, 111, 107055. [Google Scholar] [CrossRef]
  68. Ekpakpale, D.O.; Kraak, B.; Meijer, M.; Ayeni, K.I.; Houbraken, J.; Ezekiel, C.N. Fungal diversity and Aflatoxins in maize and rice grains and cassava-based flour (Pupuru) from Ondo State, Nigeria. J. Fungi 2021, 7, 635. [Google Scholar] [CrossRef]
  69. Afolabi, C.G.; Ezekiel, C.N.; Ogunbiyi, A.E.; Oluwadairo, O.J.; Sulyok, M.; Krska, R. Fungi and mycotoxins in cowpea (Vigna unguiculata L.) on Nigerian markets. Food Addit. Contam. Part B 2020, 13, 52–58. [Google Scholar] [CrossRef]
  70. Onyedum, S.C.; Adefolalu, F.S.; Muhammad, H.L.; Apeh, D.O.; Agada, M.; Imienwanrin, M.; Makun, H.A. Occurrence of major mycotoxins and their dietary exposure in North-Central Nigeria staples. Sci. Afr. 2020, 7, e00188. [Google Scholar] [CrossRef]
  71. Iqbal, S.Z.; Asi, M.R.; Malik, N. The seasonal variation of aflatoxin M1 in milk and dairy products and assessment of dietary intake in Punjab, Pakistan. Food Control 2017, 79, 292–296. [Google Scholar] [CrossRef]
  72. Hajnal, J.; Kos, J.; Krulj, J.; Krstović, S.; Jajić, I.; Pezo, L.; Šarić, B.; Nedeljković, N. Aflatoxins contamination of maize in Serbia: The impact of weather conditions in 2015. Food Addit. Contam. Part A 2017, 34, 1999–2010. [Google Scholar] [CrossRef] [PubMed]
  73. Kos, J.; Janić Hajnal, E.; Šarić, B.; Jovanov, P.; Mandić, A.; Đuragić, O.; Kokić, B. Aflatoxins in maize harvested in the Republic of Serbia over the period 2012–2016. Food Addit. Contam. Part B 2018, 11, 246–255. [Google Scholar] [CrossRef]
  74. Matumba, L.; Monjerezi, M.; Kankwamba, H.; Njoroge, S.M.; Ndilowe, P.; Kabuli, H.; Kambewa, D.; Njapau, H. Knowledge, attitude, and practices concerning presence of molds in foods among members of the general public in Malawi. Mycotoxin Res. 2016, 32, 27–36. [Google Scholar] [CrossRef]
  75. Phokane, S.; Flett, B.C.; Ncube, E.; Rheeder, J.P.; Rose, L.J. Agricultural practices and their potential role in mycotoxin contamination of maize and groundnut subsistence farming. S. Afr. J. Sci. 2019, 115, 1–6. [Google Scholar] [CrossRef]
  76. Nji, Q.N.; Babalola, O.O.; Nleya, N.; Mwanza, M. Underreported human exposure to mycotoxins: The case of South Africa. Foods 2022, 11, 2714. [Google Scholar] [CrossRef]
  77. Jiang, Y.; Ogunade, I.M.; Vyas, D.; Adesogan, A.T. Aflatoxin in dairy cows: Toxicity, occurrence in feedstuffs and milk and dietary mitigation strategies. Toxins 2021, 13, 283. [Google Scholar] [CrossRef]
  78. Smaoui, S.; D’Amore, T.; Tarapoulouzi, M.; Agriopoulou, S.; Varzakas, T. Aflatoxins contamination in feed commodities: From occurrence and toxicity to recent advances in analytical methods and detoxification. Microorganisms 2023, 11, 2614. [Google Scholar] [CrossRef]
  79. Zhao, W.; Abdelsattar, M.M.; Wang, X.; Zhang, N.; Chai, J. In vitro modulation of rumen fermentation by microbiota from the recombination of rumen fluid and solid phases. Microbiol. Spectr. 2023, 11, e03387-22. [Google Scholar] [CrossRef]
  80. Fink-Gremmels, J.; van der Merwe, D. Mycotoxins in the food chain: Contamination of foods of animal origin. In Chemical Hazards in Foods of Animal Origin; Wageningen Academic: Wageningen, The Netherlands, 2019; pp. 241–261. [Google Scholar]
  81. Anelli, P.; Haidukowski, M.; Epifani, F.; Cimmarusti, M.T.; Moretti, A.; Logrieco, A.; Susca, A. Fungal mycobiota and mycotoxin risk for traditional artisan Italian cave cheese. Food Microbiol. 2019, 78, 62–72. [Google Scholar] [CrossRef]
  82. Younis, G.; Ibrahim, D.; Awad, A.; El Bardisy, M. Determination of aflatoxin M1 and ochratoxin A in milk and dairy products in supermarkets located in Mansoura City, Egypt. Adv. Anim. Vet. Sci. 2016, 4, 114–121. [Google Scholar] [CrossRef]
  83. Mwihia, E.W.; Mbuthia, P.G.; Eriksen, G.S.; Gathumbi, J.K.; Maina, J.G.; Mutoloki, S.; Waruiru, R.M.; Mulei, I.R.; Lyche, J.L. Occurrence and Levels of Aflatoxins in Fish Feeds and Their Potential Effects on Fish in Nyeri, Kenya. Toxins 2018, 10, 543. [Google Scholar] [CrossRef] [PubMed]
  84. Namulawa, V.T.; Mutiga, S.; Musimbi, F.; Akello, S.; Ngángá, F.; Kago, L.; Kyallo, M.; Harvey, J.; Ghimire, S. Assessment of fungal contamination in fish feed from the Lake Victoria Basin, Uganda. Toxins 2020, 12, 233. [Google Scholar] [CrossRef] [PubMed]
  85. Chelimo, S.J.; Isaboke, J.; Osano, O.; Menya, D. Risk assessment of aflatoxin and fumonisin in fish feeds, Kenya a review. Risk 2021, 11, 31–42. [Google Scholar]
  86. Matejova, I.; Svobodova, Z.; Vakula, J.; Mares, J.; Modra, H. Impact of mycotoxins on aquaculture fish species: A review. J. World Aquac. Soc. 2017, 48, 186–200. [Google Scholar] [CrossRef]
  87. Triarico, S.; Rivetti, S.; Capozza, M.A.; Romano, A.; Maurizi, P.; Mastrangelo, S.; Attinà, G.; Ruggiero, A. Transplacental passage and fetal effects of antineoplastic treatment during pregnancy. Cancers 2022, 14, 3103. [Google Scholar] [CrossRef]
  88. Magoha, H.; Kimanya, M.; De Meulenaer, B.; Roberfroid, D.; Lachat, C.; Kolsteren, P. Risk of dietary exposure to Aflatoxins and fumonisins in infants less than 6 months of age in Rombo, Northern Tanzania. Matern. Child Nutr. 2016, 12, 516–527. [Google Scholar] [CrossRef]
  89. Mahdavi, R.; Nikniaz, L.; Arefhosseini, S.; Vahed Jabbari, M. Determination of Aflatoxin M1 in breast milk samples in Tabriz–Iran. Matern. Child Health J. 2010, 14, 141–145. [Google Scholar] [CrossRef]
  90. Benkerroum, N.; Ismail, A. Human breast milk contamination with Aflatoxins, impact on children’s health, and possible control means: A review. Int. J. Environ. Res. Public Health 2022, 19, 16792. [Google Scholar] [CrossRef]
  91. Kayanda, R.A.; Ngure, F.M.; Kassim, N. Dietary Exposure of Infants and Young Children to Aflatoxins and Fumonisins in the East African Region: A Review. Curr. Res. Nutr. Food Sci. J. 2024, 12, 471–489. [Google Scholar] [CrossRef]
  92. Viegas, S.; Assunção, R.; Martins, C.; Nunes, C.; Osteresch, B.; Twarużek, M.; Kosicki, R.; Grajewski, J.; Ribeiro, E.; Viegas, C. Occupational exposure to mycotoxins in swine production: Environmental and biological monitoring approaches. Toxins 2019, 11, 78. [Google Scholar] [CrossRef]
  93. Viegas, S.; Assunção, R.; Twarużek, M.; Kosicki, R.; Grajewski, J.; Viegas, C. Mycotoxins feed contamination in a dairy farm–potential implications for milk contamination and workers’ exposure in a One Health approach. J. Sci. Food Agric. 2020, 100, 1118–1123. [Google Scholar] [CrossRef]
  94. Viegas, S.; Assunção, R.; Nunes, C.; Osteresch, B.; Twarużek, M.; Kosicki, R.; Grajewski, J.; Martins, C.; Alvito, P.; Almeida, A.; et al. Exposure assessment to mycotoxins in a Portuguese fresh bread dough company by using a multi-biomarker approach. Toxins 2018, 10, 342. [Google Scholar] [CrossRef]
  95. Viegas, S.; Viegas, C.; Oppliger, A. Occupational exposure to mycotoxins: Current knowledge and prospects. Ann. Work. Expo. Health 2018, 62, 923–941. [Google Scholar] [CrossRef]
  96. Cramer, B.; Humpf, H.-U. Human biomonitoring of mycotoxins for the detection of nutritional, environmental and occupational exposure. In Exposure to Microbiological Agents in Indoor and Occupational Environments; Springer: Cham, Switzerland, 2017; pp. 191–212. [Google Scholar]
  97. Franco, L.T.; Oliveira, C.A. Assessment of occupational and dietary exposures of feed handling workers to mycotoxins in rural areas from São Paulo, Brazil. Sci. Total Environ. 2022, 837, 155763. [Google Scholar] [CrossRef]
  98. Huttunen, K.; Korkalainen, M. Microbial secondary metabolites and knowledge on inhalation effects. In Exposure to Microbiological Agents in Indoor and Occupational Environments; Springer: Cham, Switzerland, 2017; pp. 213–234. [Google Scholar]
  99. Guindon-Kezis, K.A.; Mulder, J.E.; Massey, T.E. In vivo treatment with aflatoxin B1 increases DNA oxidation, base excision repair activity and 8-oxoguanine DNA glycosylase 1 levels in mouse lung. Toxicology 2014, 321, 21–26. [Google Scholar] [CrossRef] [PubMed]
  100. Viegas, S.; Veiga, L.; Almeida, A.; dos Santos, M.; Carolino, E.; Viegas, C. Occupational exposure to Aflatoxin B1 in a Portuguese poultry slaughterhouse. Ann. Occup. Hyg. 2016, 60, 176–183. [Google Scholar] [CrossRef] [PubMed]
  101. Mo, X.; Lai, H.; Yang, Y.; Xiao, J.; He, K.; Liu, C.; Chen, J.; Lin, Y. How does airway exposure of aflatoxin B1 affect serum albumin adduct concentrations? Evidence based on epidemiological study and animal experimentation. J. Toxicol. Sci. 2014, 39, 645–653. [Google Scholar] [CrossRef] [PubMed]
  102. Lai, H.; Mo, X.; Yang, Y.; He, K.; Xiao, J.; Liu, C.; Chen, J.; Lin, Y. Association between aflatoxin B1 occupational airway exposure and risk of hepatocellular carcinoma: A case-control study. Tumor Biol. 2014, 35, 9577–9584. [Google Scholar] [CrossRef]
  103. Brera, C.; Caputi, R.; Miraglia, M.; Iavicoli, I.; Salerno, A.; Carelli, G. Exposure assessment to mycotoxins in workplaces: Aflatoxins and Ochratoxin A occurrence in airborne dusts and human sera. Microchem. J. 2002, 73, 167–173. [Google Scholar] [CrossRef]
  104. Selim, M.I.; Juchems, A.M.; Popendorf, W. Assessing airborne aflatoxin B1 during on-farm grain handling activities. Am. Ind. Hyg. Assoc. J. 1998, 59, 252–256. [Google Scholar] [CrossRef] [PubMed]
  105. Autrup, J.L.; Schmidt, J.; Seremet, T.; Autrup, H. Determination of exposure to Aflatoxins among Danish workers in animal-feed production through the analysis of aflatoxin B1 adducts to serum albumin. Scand. J. Work Environ. Health 1991, 17, 436–440. [Google Scholar] [CrossRef] [PubMed]
  106. Viegas, S.; Veiga, L.; Figueiredo, P.; Almeida, A.; Carolino, E.; Viegas, C. Assessment of workers’ exposure to aflatoxin B1 in a Portuguese waste industry. Ann. Occup. Hyg. 2015, 59, 173–181. [Google Scholar]
  107. Viegas, S.; Veiga, L.; Malta-Vacas, J.; Sabino, R.; Figueredo, P.; Almeida, A.; Viegas, C.; Carolino, E. Occupational exposure to aflatoxin (AFB1) in poultry production. J. Toxicol. Environ. Health Part A 2012, 75, 1330–1340. [Google Scholar] [CrossRef]
  108. Saad-Hussein, A.; Beshir, S.; Moubarz, G.; Elserougy, S.; Ibrahim, M.I. Effect of occupational exposure to Aflatoxins on some liver tumor markers in textile workers. Am. J. Ind. Med. 2013, 56, 818–824. [Google Scholar] [CrossRef]
  109. Viegas, S.; Osteresch, B.; Almeida, A.; Cramer, B.; Humpf, H.-U.; Viegas, C. Enniatin B and Ochratoxin A in the blood serum of workers from the waste management setting. Mycotoxin Res. 2018, 34, 85–90. [Google Scholar] [CrossRef]
  110. Oluwafemi, F.; Odebiyi, T.; Kolapo, A. Occupational aflatoxin exposure among feed mill workers in Nigeria. World Mycotoxin J. 2012, 5, 385–389. [Google Scholar] [CrossRef]
  111. Wangia, R.N.; Tang, L.; Wang, J.-S. Occupational exposure to Aflatoxins and health outcomes: A review. J. Environ. Sci. Health Part C 2019, 37, 215–234. [Google Scholar] [CrossRef]
  112. Taevernier, L.; Veryser, L.; Roche, N.; Peremans, K.; Burvenich, C.; Delesalle, C.; De Spiegeleer, B. Human skin permeation of emerging mycotoxins (Beauvericin and Enniatins). J. Expo. Sci. Environ. Epidemiol. 2016, 26, 277–287. [Google Scholar] [CrossRef]
  113. Pallares, N.; Tolosa, J.; Ferrer, E.; Berrada, H. Mycotoxins in raw materials, beverages and supplements of botanicals: A review of occurrence, risk assessment and analytical methodologies. Food Chem. Toxicol. 2022, 165, 113013. [Google Scholar] [CrossRef] [PubMed]
  114. Carballo, D.; Fernández-Franzón, M.; Ferrer, E.; Pallarés, N.; Berrada, H. Dietary exposure to mycotoxins through alcoholic and non-alcoholic beverages in Valencia, Spain. Toxins 2021, 13, 438. [Google Scholar] [CrossRef] [PubMed]
  115. Fan, K.; Xu, J.; Jiang, K.; Liu, X.; Meng, J.; Di Mavungu, J.D.; Guo, W.; Zhang, Z.; Jing, J.; Li, H.; et al. Determination of multiple mycotoxins in paired plasma and urine samples to assess human exposure in Nanjing, China. Environ. Pollut. 2019, 248, 865–873. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, M.; Guo, X. Emerging strategies in fluorescent aptasensor toward food hazard Aflatoxins detection. Trends Food Sci. Technol. 2022, 129, 621–633. [Google Scholar] [CrossRef]
  117. Zarba, A.; Wild, C.P.; Hall, A.J.; Montesano, R.; Hudson, G.J.; Groopman, J.D. Aflatoxrn M1 in human breast milk from The Gambia, West Africa, quantified by combined monoclonal antibody immunoaffinity chromatography and HPLC. Carcinogenesis 1992, 13, 891–894. [Google Scholar] [CrossRef]
  118. Wild, C.P.; Gong, Y.Y. Mycotoxins and human disease: A largely ignored global health issue. Carcinogenesis 2010, 31, 71–82. [Google Scholar] [CrossRef]
  119. Mamo, F.T.; Abate, B.A.; Zheng, Y.; Nie, C.; He, M.; Liu, Y. Distribution of Aspergillus fungi and recent aflatoxin reports, health risks, and advances in developments of biological mitigation strategies in China. Toxins 2021, 13, 678. [Google Scholar] [CrossRef]
  120. Escrivá, L.; Font, G.; Manyes, L.; Berrada, H. Studies on the presence of mycotoxins in biological samples: An overview. Toxins 2017, 9, 251. [Google Scholar] [CrossRef]
  121. Vallero, D.A. Translating Diverse Environmental Data into Reliable Information: How to Coordinate Evidence from Different Sources; Academic Press: Cambridge, MA, USA, 2017; pp. 251–296. [Google Scholar]
  122. Belbasis, L.; Mavrogiannis, M.C.; Emfietzoglou, M.; Evangelou, E. Environmental factors, serum biomarkers and risk of atrial fibrillation: An exposure-wide umbrella review of meta-analyses. Eur. J. Epidemiol. 2020, 35, 223–239. [Google Scholar] [CrossRef]
  123. Filipoiu, D.C.; Bungau, S.G.; Endres, L.; Negru, P.A.; Bungau, A.F.; Pasca, B.; Radu, A.-F.; Tarce, A.G.; Bogdan, M.A.; Behl, T.; et al. Characterization of the toxicological impact of heavy metals on human health in conjunction with modern analytical methods. Toxics 2022, 10, 716. [Google Scholar] [CrossRef]
  124. Frusciante, L.; Visibelli, A.; Geminiani, M.; Santucci, A.; Spiga, O. Artificial Intelligence Approaches in Drug Discovery: Towards the Laboratory of the Future. Curr. Top. Med. Chem. 2022, 22, 2176–2189. [Google Scholar] [CrossRef] [PubMed]
  125. Quig, D. Metal toxicity: Assessment of exposure and retention. In Textbook of Natural Medicine; Elsevier: Amsterdam, The Netherlands, 2020; 187p. [Google Scholar]
  126. Habschied, K.; Kanižai Šarić, G.; Krstanović, V.; Mastanjević, K. Mycotoxins—Biomonitoring and human exposure. Toxins 2021, 13, 113. [Google Scholar] [CrossRef] [PubMed]
  127. Bocca, B.; Battistini, B.; Petrucci, F. Silver and gold nanoparticles characterization by SP-ICP-MS and AF4-FFF-MALS-UV-ICP-MS in human samples used for biomonitoring. Talanta 2020, 220, 121404. [Google Scholar] [CrossRef] [PubMed]
  128. Alvarado, A.M.; Zamora-Sanabria, R.; Granados-Chinchilla, F. A focus on Aflatoxins in feedstuffs: Levels of contamination, prevalence, control strategies, and impacts on animal health. Aflatoxin-Control. Anal. Detect. Health Risks 2017, 2017, 116–152. [Google Scholar]
  129. Thompson, L.E.; Joy, M.S. Endogenous markers of kidney function and renal drug clearance processes of filtration, secretion, and reabsorption. Curr. Opin. Toxicol. 2022, 31, 100344. [Google Scholar] [CrossRef]
  130. Owolabi, I.O.; Siwarak, K.; Greer, B.; Rajkovic, A.; Dall’asta, C.; Karoonuthaisiri, N.; Uawisetwathana, U.; Elliott, C.T.; Petchkongkaew, A. Applications of mycotoxin biomarkers in human biomonitoring for exposome-health studies: Past, present, and future. Expo. Health 2024, 16, 837–859. [Google Scholar] [CrossRef]
  131. Arce-López, B.; Lizarraga, E.; Vettorazzi, A.; González-Peñas, E. Human biomonitoring of mycotoxins in blood, plasma and serum in recent years: A review. Toxins 2020, 12, 147. [Google Scholar] [CrossRef]
  132. Bore, M.G. Nutritional Status Among Adolescents Living with Human Immuno-Deficiency Virus on Anti-Retroviral Therapy Follow-Up Living in Selected Regions of Ethiopia: A Mixed Methods Study. 2024. Available online: http://hdl.handle.net/10453/187481 (accessed on 29 April 2025).
  133. Wobudeya, E.; Bonnet, M.; Walters, E.G.; Nabeta, P.; Song, R.; Murithi, W.; Mchembere, W.; Dim, B.; Taguebue, J.-V.; Orne-Gliemann, J.; et al. Diagnostic advances in childhood tuberculosis—Improving specimen collection and yield of microbiological diagnosis for intrathoracic tuberculosis. Pathogens 2022, 11, 389. [Google Scholar] [CrossRef]
  134. Elabed, S.; Khaled, R.; Farhat, N.; Madkour, M.; Zadeh, S.A.M.; Shousha, T.; Taneera, J.; Semerjian, L.; Abass, K. Assessing Aflatoxin Exposure in the United Arab Emirates (UAE): Biomonitoring AFM1 levels in Urine Samples and their Association with Dietary Habits. Environ. Toxicol. Pharmacol. 2025, 114, 104644. [Google Scholar] [CrossRef]
  135. Nasir, U.; Naeem, I.; Asif, M.; Ismail, A.; Gong, Y.Y.; Routledge, M.N.; Amjad, A.; Fazal, A.; Ismail, Z. Assessment of Aflatoxins exposure through urinary biomarker approach and the evaluation of the impacts of Aflatoxins exposure on the selected health parameters of the children of Multan city of Pakistan. Food Control 2021, 123, 107863. [Google Scholar] [CrossRef]
  136. Kuempel, E.D.; Roberts, J.R.; Roth, G.; Dunn Kevin, L.; Zumwalde, R.D.; Hubbs, A.F.; Trout, D.; Holdsworth, G. Health Effects of Occupational Exposure to Silver Nanomaterials. 2021. Available online: https://www.cdc.gov/niosh/docs/2021-112/default.html (accessed on 29 April 2025).
  137. Liu, B.; Wang, Y.; Zhang, Y.; Yan, B. Mechanisms of protective effects of SGLT2 inhibitors in cardiovascular disease and renal dysfunction. Curr. Top. Med. Chem. 2019, 19, 1818–1849. [Google Scholar] [CrossRef]
  138. Panel, E.C.; Schrenk, D.; Bignami, M.; Bodin, L.; Chipman, J.K.; Del Mazo, J.; Grasl-Kraupp, B.; Hogstrand, C.; Hoogenboom, L.R.; Leblanc, J.-C. Risk assessment of aflatoxins in food. EFSA J. 2020, 18, e06040. [Google Scholar] [CrossRef]
  139. Ouhibi, S.; Vidal, A.; Martins, C.; Gali, R.; Hedhili, A.; De Saeger, S.; De Boevre, M. LC-MS/MS methodology for simultaneous determination of patulin and citrinin in urine and plasma applied to a pilot study in colorectal cancer patients. Food Chem. Toxicol. 2020, 136, 110994. [Google Scholar] [CrossRef]
  140. Dabuo, B.; Avogo, E.W.; Koomson, G.O.; Akantibila, M.; Gbati, D.A. Aflatoxins: Toxicity, Occurrences and Chronic Exposure, in Aflatoxins-Occurrence, Detection and Novel Detoxification Strategies; IntechOpen: London, UK, 2022. [Google Scholar] [CrossRef]
  141. Turner, P.C.; Snyder, J.A. Development and limitations of exposure biomarkers to dietary contaminants mycotoxins. Toxins 2021, 13, 314. [Google Scholar] [CrossRef]
  142. Xue, K.S. Toxic Effects and Intervention Strategies for Co-Exposure to Aflatoxins and Fumonisins in Animals and High-Risk Human Populations. University of Georgia. 2017. Available online: https://getd.libs.uga.edu/pdfs/xue_kathy_s_201705_phd.pdf (accessed on 29 April 2025).
  143. Slobodchikova, I.; Vuckovic, D. Liquid chromatography–high resolution mass spectrometry method for monitoring of 17 mycotoxins in human plasma for exposure studies. J. Chromatogr. A 2018, 1548, 51–63. [Google Scholar] [CrossRef]
  144. Xue, K.S.; Cai, W.; Tang, L.; Wang, J.-S. Aflatoxin B1-lysine adduct in dried blood spot samples of animals and humans. Food Chem. Toxicol. 2016, 98, 210–219. [Google Scholar] [CrossRef]
  145. Adaku Chilaka, C.; Mally, A. Mycotoxin occurrence, exposure and health implications in infants and young children in Sub-Saharan Africa: A review. Foods 2020, 9, 1585. [Google Scholar] [CrossRef] [PubMed]
  146. Fakhri, Y.; Ghorbani, R.; Taghavi, M.; Keramati, H.; Amanidaz, N.; Moradi, B.; Nazari, S.H.; Shariatifar, N.; Khaneghah, A.M. Concentration and prevalence of aflatoxin M1 in human breast milk in Iran: Systematic review, meta-analysis, and carcinogenic risk assessment: A review. J. Food Prot. 2019, 82, 785–795. [Google Scholar] [CrossRef]
  147. Min, L.; Fink-Gremmels, J.; Li, D.; Tong, X.; Tang, J.; Nan, X.; Yu, Z.; Chen, W.; Wang, G. An overview of aflatoxin B1 biotransformation and Aflatoxin M1 secretion in lactating dairy cows. Anim. Nutr. 2021, 7, 42–48. [Google Scholar] [CrossRef]
  148. Li, K.; Cai, H.; Luo, B.; Duan, S.; Yang, J.; Zhang, N.; He, Y.; Wu, A.; Liu, H. Recent Progress of Mycotoxin in Various Food Products—Human Exposure and Health Risk Assessment. Foods 2025, 14, 865. [Google Scholar] [CrossRef]
  149. Ezekiel, C.N.; Abia, W.A.; Braun, D.; Šarkanj, B.; Ayeni, K.I.; Oyedele, O.A.; Michael-Chikezie, E.C.; Ezekiel, V.C.; Mark, B.; Ahuchaogu, C.P.; et al. Comprehensive mycotoxin exposure biomonitoring in breastfed and non-exclusively breastfed Nigerian children. medRxiv 2020. [Google Scholar] [CrossRef]
  150. Lehmann, G.M.; LaKind, J.S.; Davis, M.H.; Hines, E.P.; Marchitti, S.A.; Alcala, C.; Lorber, M. Environmental chemicals in breast milk and formula: Exposure and risk assessment implications. Environ. Health Perspect. 2018, 126, 096001. [Google Scholar] [CrossRef] [PubMed]
  151. Rushing, B.R.; Selim, M.I. Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food Chem. Toxicol. 2019, 124, 81–100. [Google Scholar] [CrossRef] [PubMed]
  152. Mupunga, I.; van Rensburg, I.J.; Luthuli, N.; Abafe, O.A.; Shai, L.J.; Katerere, D.R. Analysis of aflatoxin biomarkers in the hair of experimental animals. Toxins 2021, 13, 570. [Google Scholar] [CrossRef]
  153. Wang, L.; Huang, Q.; Wu, J.; Wu, W.; Jiang, J.; Yan, H.; Huang, J.; Sun, Y.; Deng, Y. The metabolism and biotransformation of AFB1: Key enzymes and pathways. Biochem. Pharmacol. 2022, 199, 115005. [Google Scholar] [CrossRef]
  154. Eaton, D.L.; Williams, D.E.; Coulombe, R.A. Species differences in the biotransformation of aflatoxin B1: Primary determinants of relative carcinogenic potency in different animal species. Toxins 2025, 17, 30. [Google Scholar] [CrossRef]
  155. Kensler, T.W.; Groopman, J.D. Aflatoxin Exposure, Human Liver Cancer Risk, and Chemoprevention. In Carcinogens, DNA Damage and Cancer Risk: Mechanisms of Chemical Carcinogenesis; World Scientific: Singapore, 2019; pp. 143–169. [Google Scholar]
  156. Hamzehloo-Moghadam, M.; Asri, N.; Sherafat, S.J.; Mansouri, V. Detection of aflatoxin contamination through biomarker discovery in human intestinal Caco-2 cells. Appl. Food Biotechnol. 2024, 11, e4. [Google Scholar]
  157. Giolo, M.P.; Oliveira, C.M.D.; Bertolini, D.A.; Lonardoni, M.V.C.; Gouveia, M.S.; Netto, D.P.; Nixdorf, S.L.; Machinski Junior, M. Aflatoxin M1 in the urine of non-carriers and chronic carriers of hepatitis B virus in Maringa, Brazil. Braz. J. Pharm. Sci. 2012, 48, 447–452. [Google Scholar] [CrossRef]
  158. de Cássia Romero, A.; Ferreira, T.R.B.; dos Santos Dias, C.T.; Calori-Domingues, M.A.; da Gloria, E.M. Occurrence of AFM1 in urine samples of a Brazilian population and association with food consumption. Food Control 2010, 21, 554–558. [Google Scholar] [CrossRef]
  159. Ediage, E.N.; Di Mavungu, J.D.; Song, S.; Sioen, I.; De Saeger, S. Multimycotoxin analysis in urines to assess infant exposure: A case study in Cameroon. Environ. Int. 2013, 57, 50–59. [Google Scholar] [CrossRef]
  160. Cao, X.; Wu, S.; Yue, Y.; Wang, S.; Wang, Y.; Tao, L.; Tian, H.; Xie, J.; Ding, H. A high-throughput method for the simultaneous determination of multiple mycotoxins in human and laboratory animal biological fluids and tissues by PLE and HPLC–MS/MS. J. Chromatogr. B 2013, 942, 113–125. [Google Scholar] [CrossRef] [PubMed]
  161. Mykkänen, H.; Zhu, H.; Salminen, E.; Juvonen, R.O.; Ling, W.; Ma, J.; Polychronaki, N.; Kemiläinen, H.; Mykkänen, O.; Salminen, S. Fecal and urinary excretion of aflatoxin B1 metabolites (AFQ1, AFM1 and AFB-N7-guanine) in young Chinese males. Int. J. Cancer 2005, 115, 879–884. [Google Scholar] [CrossRef] [PubMed]
  162. Abia, W.A.; Warth, B.; Sulyok, M.; Krska, R.; Tchana, A.; Njobeh, P.B.; Turner, P.C.; Kouanfack, C.; Eyongetah, M.; Dutton, M.; et al. Bio-monitoring of mycotoxin exposure in Cameroon using a urinary multi-biomarker approach. Food Chem. Toxicol. 2013, 62, 927–934. [Google Scholar] [CrossRef] [PubMed]
  163. Egner, P.A.; Groopman, J.D.; Wang, J.-S.; Kensler, T.W.; Friesen, M.D. Quantification of aflatoxin-b1-n7-guanine in human urine by high-performance liquid chromatography and isotope dilution tandem mass spectrometry1. Chem. Res. Toxicol. 2006, 19, 1191–1195. [Google Scholar] [CrossRef]
  164. Wang, J.-S.; Huang, T.; Su, J.; Liang, F.; Wei, Z.; Liang, Y.; Luo, H.; Kuang, S.-Y.; Qian, G.-S.; Sun, G. Hepatocellular carcinoma and aflatoxin exposure in Zhuqing village, Fusui county, People’s Republic of China. Cancer Epidemiol. Biomark. Prev. 2001, 10, 143–146. [Google Scholar]
  165. Ostry, V.; Malir, F.; Roubal, T.; Skarkova, J.; Ruprich, J.; Cerna, M.; Creppy, E.E. Monitoring of mycotoxin biomarkers in the Czech Republic. Mycotoxin Res. 2005, 21, 49–52. [Google Scholar] [CrossRef]
  166. Hatem, N.L.; Hassab, H.M.; Al-Rahman, E.M.A.; El-Deeb, S.A.; Ahmed, R.L.E.-S. Prevalence of Aflatoxins in blood and urine of Egyptian infants with protein–energy malnutrition. Food Nutr. Bull. 2005, 26, 49–56. [Google Scholar] [CrossRef]
  167. Polychronaki, N.; Wild, C.P.; Mykkänen, H.; Amra, H.; Abdel-Wahhab, M.; Sylla, A.; Diallo, M.; El-Nezami, H.; Turner, P.C. Urinary biomarkers of aflatoxin exposure in young children from Egypt and Guinea. Food Chem. Toxicol. 2008, 46, 519–526. [Google Scholar] [CrossRef]
  168. Piekkola, S.; Turner, P.; Abdel-Hamid, M.; Ezzat, S.; El-Daly, M.; El-Kafrawy, S.; Savchenko, E.; Poussa, T.; Woo, J.; Mykkänen, H. Characterisation of aflatoxin and deoxynivalenol exposure among pregnant Egyptian women. Food Addit. Contam. Part A 2012, 29, 962–971. [Google Scholar] [CrossRef]
  169. Mohd Redzwan, S.; Rosita, J.; Mohd Sokhini, A.M.; Nurul Aqilah, A.R. Association between aflatoxin M 1 excreted in human urine samples with the consumption of milk and dairy products. Bull. Environ. Contam. Toxicol. 2012, 89, 1115–1119. [Google Scholar] [CrossRef]
  170. Warth, B.; Sulyok, M.; Fruhmann, P.; Mikula, H.; Berthiller, F.; Schuhmacher, R.; Hametner, C.; Abia, W.A.; Adam, G.; Fröhlich, J.; et al. Development and validation of a rapid multi-biomarker liquid chromatography/tandem mass spectrometry method to assess human exposure to mycotoxins. Rapid Commun. Mass Spectrom. 2012, 26, 1533–1540. [Google Scholar] [CrossRef] [PubMed]
  171. Song, S.; Ediage, E.N.; Wu, A.; De Saeger, S. Development and application of salting-out assisted liquid/liquid extraction for multi-mycotoxin biomarkers analysis in pig urine with high performance liquid chromatography/tandem mass spectrometry. J. Chromatogr. A 2013, 1292, 111–120. [Google Scholar] [CrossRef]
  172. Ezekiel, C.N.; Warth, B.; Ogara, I.M.; Abia, W.A.; Ezekiel, V.C.; Atehnkeng, J.; Sulyok, M.; Turner, P.C.; Tayo, G.O.; Krska, R.; et al. Mycotoxin exposure in rural residents in northern Nigeria: A pilot study using multi-urinary biomarkers. Environ. Int. 2014, 66, 138–145. [Google Scholar] [CrossRef]
  173. Cao, X.; Li, X.; Li, J.; Niu, Y.; Shi, L.; Fang, Z.; Zhang, T.; Ding, H. Quantitative determination of carcinogenic mycotoxins in human and animal biological matrices and animal-derived foods using multi-mycotoxin and analyte-specific high performance liquid chromatography-tandem mass spectrometric methods. J. Chromatogr. B 2018, 1073, 191–200. [Google Scholar] [CrossRef]
  174. McMillan, A.; Renaud, J.B.; Burgess, K.M.; Orimadegun, A.E.; Akinyinka, O.O.; Allen, S.J.; Miller, J.D.; Reid, G.; Sumarah, M.W. Aflatoxin exposure in Nigerian children with severe acute malnutrition. Food Chem. Toxicol. 2018, 111, 356–362. [Google Scholar] [CrossRef]
  175. Koshiol, J.; Gao, Y.-T.; Dean, M.; Egner, P.; Nepal, C.; Jones, K.; Wang, B.; Rashid, A.; Luo, W.; Van Dyke, A.L.; et al. Association of aflatoxin and gallbladder cancer. Gastroenterology 2017, 153, 488–494.e1. [Google Scholar] [CrossRef]
  176. Mohammed, A.; Chala, A.; Dejene, M.; Fininsa, C.; Hoisington, D.A.; Sobolev, V.S.; Arias, R.S. Aspergillus and aflatoxin in groundnut (Arachis hypogaea L.) and groundnut cake in Eastern Ethiopia. Food Addit. Contam. Part B 2016, 9, 290–298. [Google Scholar] [CrossRef]
  177. Lauer, J.M.; Duggan, C.P.; Ausman, L.M.; Griffiths, J.K.; Webb, P.; Wang, J.S.; Xue, K.S.; Agaba, E.; Nshakira, N.; Ghosh, S. Maternal aflatoxin exposure during pregnancy and adverse birth outcomes in Uganda. Matern. Child Nutr. 2019, 15, e12701. [Google Scholar] [CrossRef]
  178. Leroy, J.L.; Sununtnasuk, C.; García-Guerra, A.; Wang, J.S. Low level aflatoxin exposure associated with greater linear growth in southern Mexico: A longitudinal study. Matern. Child Nutr. 2018, 14, e12619. [Google Scholar] [CrossRef]
  179. Leroy, J.L.; Wang, J.-S.; Jones, K. Serum aflatoxin B1-lysine adduct level in adult women from Eastern Province in Kenya depends on household socio-economic status: A cross sectional study. Soc. Sci. Med. 2015, 146, 104–110. [Google Scholar] [CrossRef]
  180. Smith, L.E.; Mbuya, M.N.; Prendergast, A.J.; Turner, P.C.; Ruboko, S.; Humphrey, J.H.; Nelson, R.J.; Chigumira, A.; Kembo, G.; Stoltzfus, R.J. Determinants of recent aflatoxin exposure among pregnant women in rural Zimbabwe. Mol. Nutr. Food Res. 2017, 61, 1601049. [Google Scholar] [CrossRef] [PubMed]
  181. Chen, C.; Mitchell, N.J.; Gratz, J.; Houpt, E.R.; Gong, Y.; Egner, P.A.; Groopman, J.D.; Riley, R.T.; Showker, J.L.; Svensen, E.; et al. Exposure to aflatoxin and fumonisin in children at risk for growth impairment in rural Tanzania. Environ. Int. 2018, 115, 29–37. [Google Scholar] [CrossRef] [PubMed]
  182. Mahfuz, M.; Alam, M.A.; Fahim, S.M.; Gazi, M.A.; Raihan, M.J.; Hossain, M.; Egner, P.A.; Bessong, P.O.; Petri, W.A.; Groopman, J.D.; et al. Aflatoxin exposure in children living in Mirpur, Dhaka: Data from MAL-ED companion study. J. Expo. Sci. Environ. Epidemiol. 2019, 29, 655–662. [Google Scholar] [CrossRef]
  183. Hernandez-Vargas, H.; Castelino, J.; Silver, M.J.; Dominguez-Salas, P.; Cros, M.-P.; Durand, G.; Calvez-Kelm, F.L.; Prentice, A.M.; Wild, C.P.; Moore, S.E.; et al. Exposure to aflatoxin B1 in utero is associated with DNA methylation in white blood cells of infants in The Gambia. Int. J. Epidemiol. 2015, 44, 1238–1248. [Google Scholar] [CrossRef] [PubMed]
  184. Seetha, A.; Monyo, E.S.; Tsusaka, T.W.; Msere, H.W.; Madinda, F.; Chilunjika, T.; Sichone, E.; Mbughi, D.; Chilima, B.; Matumba, L. Aflatoxin-lysine adducts in blood serum of the Malawian rural population and aflatoxin contamination in foods (groundnuts, maize) in the corresponding areas. Mycotoxin Res. 2018, 34, 195–204. [Google Scholar] [CrossRef]
  185. Watson, S.; Chen, G.; Sylla, A.; Routledge, M.N.; Gong, Y.Y. Dietary exposure to aflatoxin and micronutrient status among young children from Guinea. Mol. Nutr. Food Res. 2016, 60, 511–518. [Google Scholar] [CrossRef]
  186. Watson, S.; Moore, S.E.; Darboe, M.K.; Chen, G.; Tu, Y.-K.; Huang, Y.-T.; Eriksen, K.G.; Bernstein, R.M.; Prentice, A.M.; Wild, C.P.; et al. Impaired growth in rural Gambian infants exposed to aflatoxin: A prospective cohort study. BMC Public Health 2018, 18, 1247. [Google Scholar] [CrossRef]
  187. Xie, J.; Jiang, H.; Shen, J.; Peng, T.; Wang, J.; Yao, K.; Sun, S.; Shao, B.; Tan, J. Design of multifunctional nanostructure for ultrafast extraction and purification of Aflatoxins in foodstuffs. Anal. Chem. 2017, 89, 10556–10564. [Google Scholar] [CrossRef]
  188. Kang, M.-S.; Nkurunziza, P.; Muwanika, R.; Qian, G.; Tang, L.; Song, X.; Xue, K.; Nkwata, A.; Ssempebwa, J.; Lutalo, T. Longitudinal evaluation of aflatoxin exposure in two cohorts in south-western Uganda. Food Addit. Contam. Part A 2015, 32, 1322–1330. [Google Scholar] [CrossRef]
  189. Shirima, C.P.; Kimanya, M.E.; Routledge, M.N.; Srey, C.; Kinabo, J.L.; Humpf, H.-U.; Wild, C.P.; Tu, Y.-K.; Gong, Y.Y. A prospective study of growth and biomarkers of exposure to aflatoxin and fumonisin during early childhood in Tanzania. Environ. Health Perspect. 2015, 123, 173–178. [Google Scholar] [CrossRef]
  190. Turner, P.C.; Loffredo, C.; Kafrawy, S.E.; Ezzat, S.; Eissa, S.A.L.; Daly, M.E.; Nada, O.; Abdel-Hamid, M. Pilot survey of aflatoxin–albumin adducts in sera from Egypt. Food Addit. Contam. 2008, 25, 583–587. [Google Scholar] [CrossRef]
  191. Shuaib, F.M.; Jolly, P.E.; Ehiri, J.E.; Yatich, N.; Jiang, Y.; Funkhouser, E.; Person, S.D.; Wilson, C.; Ellis, W.O.; Wang, J.S.; et al. Association between birth outcomes and aflatoxin B1 biomarker blood levels in pregnant women in Kumasi, Ghana. Trop. Med. Int. Health 2010, 15, 160–167. [Google Scholar] [CrossRef] [PubMed]
  192. Turner, P.C.; Sylla, A.; Gong, Y.Y.; Diallo, M.S.; Sutcliffe, A.E.; Hall, A.J.; Wild, C.P. Reduction in exposure to carcinogenic Aflatoxins by postharvest intervention measures in West Africa: A community-based intervention study. Lancet 2005, 365, 1950–1956. [Google Scholar] [CrossRef] [PubMed]
  193. Yard, E.E.; Daniel, J.H.; Lewis, L.S.; Rybak, M.E.; Paliakov, E.M.; Kim, A.A.; Montgomery, J.M.; Bunnell, R.; Abudo, M.U.; Akhwale, W.; et al. Human aflatoxin exposure in Kenya, 2007: A cross-sectional study. Food Addit. Contam. Part A 2013, 30, 1322–1331. [Google Scholar] [CrossRef]
  194. Leong, Y.-H.; Rosma, A.; Latiff, A.A.; Izzah, A.N. Associations of serum aflatoxin B1–lysine adduct level with socio-demographic factors and Aflatoxins intake from nuts and related nut products in Malaysia. Int. J. Hyg. Environ. Health 2012, 215, 368–372. [Google Scholar] [CrossRef] [PubMed]
  195. Jonsyn-Ellis, F.E. Aflatoxins and ochratoxins in serum samples of school children. J. Nutr. Environ. Med. 2007, 16, 52–58. [Google Scholar] [CrossRef]
  196. Turner, P.C.; Collinson, A.C.; Cheung, Y.B.; Gong, Y.; Hall, A.J.; Prentice, A.M.; Wild, C.P. Aflatoxin exposure in utero causes growth faltering in Gambian infants. Int. J. Epidemiol. 2007, 36, 1119–1125. [Google Scholar] [CrossRef]
  197. Johnson, N.M.; Qian, G.; Xu, L.; Tietze, D.; Marroquin-Cardona, A.; Robinson, A.; Rodriguez, M.; Kaufman, L.; Cunningham, K.; Wittmer, J.; et al. Aflatoxin and PAH exposure biomarkers in a US population with a high incidence of hepatocellular carcinoma. Sci. Total Environ. 2010, 408, 6027–6031. [Google Scholar] [CrossRef]
  198. Polychronaki, N.; Turner, P.C.; Mykkänen, H.; Gong, Y.; Amra, H.; Abdel-Wahhab, M.; El-Nezami, H. Determinants of aflatoxin M1 in breast milk in a selected group of Egyptian mothers. Food Addit. Contam. 2006, 23, 700–708. [Google Scholar] [CrossRef]
  199. Gürbay, A.; Sabuncuoğlu, S.A.; Girgin, G.; Şahin, G.; Yiğit, Ş.; Yurdakök, M.; Tekinalp, G. Exposure of newborns to aflatoxin M1 and B1 from mothers’ breast milk in Ankara, Turkey. Food Chem. Toxicol. 2010, 48, 314–319. [Google Scholar] [CrossRef]
  200. Abdulrazzaq, Y.M.; Osman, N.; Yousif, Z.M.; Al-Falahi, S. Aflatoxin M1 in breast-milk of UAE women. Ann. Trop. Pediatr. 2003, 23, 173–179. [Google Scholar] [CrossRef] [PubMed]
  201. Patriarca, M.; Barlow, N.; Cross, A.; Hill, S.; Robson, A.; Taylor, A.; Tyson, J. Atomic spectrometry update: Review of advances in the analysis of clinical and biological materials, foods and beverages. J. Anal. At. Spectrom. 2021, 36, 452–511. [Google Scholar] [CrossRef]
  202. Sanchis, A.; Bosch-Orea, C.; Salvador, J.-P.; Marco, M.-P.; Farré, M. Development and validation of a multianalyte immunoassay for the quantification of environmental pollutants in seawater samples from the Catalonia coastal area. Anal. Bioanal. Chem. 2019, 411, 5897–5907. [Google Scholar] [CrossRef] [PubMed]
  203. Devalloir, Q. Effects of Multi-Stressors (Pollution, Nutritional Quality) on the Immunocompetence of the Wood Mouse. Ph.D. Thesis, Université Bourgogne Franche-Comté, Besançon, France, 2023. Available online: https://theses.hal.science/tel-04598579 (accessed on 29 April 2025).
  204. Afzal, A.; Mahreen, N. Emerging insights into the impacts of heavy metals exposure on health, reproductive and productive performance of livestock. Front. Pharmacol. 2024, 15, 1375137. [Google Scholar] [CrossRef]
  205. Kraft, S.; Buchenauer, L.; Polte, T. Mold, mycotoxins and a dysregulated immune system: A combination of concern? Int. J. Mol. Sci. 2021, 22, 12269. [Google Scholar] [CrossRef]
  206. Gao, Y.; Meng, L.; Liu, H.; Wang, J.; Zheng, N. The compromised intestinal barrier induced by mycotoxins. Toxins 2020, 12, 619. [Google Scholar] [CrossRef]
  207. Gonkowski, S.; Gajęcka, M.; Makowska, K. Mycotoxins and the enteric nervous system. Toxins 2020, 12, 461. [Google Scholar] [CrossRef]
  208. Huang, Q.; Jiang, K.; Tang, Z.; Fan, K.; Meng, J.; Nie, D.; Zhao, Z.; Wu, Y.; Han, Z. Exposure assessment of multiple mycotoxins and cumulative health risk assessment: A biomonitoring-based study in the Yangtze River Delta, China. Toxins 2021, 13, 103. [Google Scholar] [CrossRef]
  209. Kyei, N.N.; Cramer, B.; Humpf, H.-U.; Degen, G.H.; Ali, N.; Gabrysch, S. Assessment of multiple mycotoxin exposure and its association with food consumption: A human biomonitoring study in a pregnant cohort in rural Bangladesh. Arch. Toxicol. 2022, 96, 2123–2138. [Google Scholar] [CrossRef]
  210. Gong, Y.Y.; Watson, S.; Routledge, M.N. Aflatoxin exposure and associated human health effects, a review of epidemiological studies. Food Saf. 2016, 4, 14–27. [Google Scholar] [CrossRef]
  211. Sumon, A.H.; Islam, F.; Mohanto, N.C.; Kathak, R.R.; Molla, N.H.; Rana, S.; Degen, G.H.; Ali, N. The presence of Aflatoxin M1 in milk and milk products in Bangladesh. Toxins 2021, 13, 440. [Google Scholar] [CrossRef]
  212. Ritieni, A.; Santini, A.; Mussap, M.; Ferracane, R.; Bosco, P.; Gazzolo, D.; Galvano, F. Simultaneous determination of mycotoxins in biological fluids by LC-MS/MS. Front. Biosci. 2010, 2, 151–158. [Google Scholar] [CrossRef]
  213. Omar, S.S. Incidence of aflatoxin M1 in human and animal milk in Jordan. J. Toxicol. Environ. Health Part A 2012, 75, 1404–1409. [Google Scholar] [CrossRef] [PubMed]
  214. Kościelecka, K.; Kuć, A.; Kubik-Machura, D.; Męcik-Kronenberg, T.; Włodarek, J.; Radko, L. Endocrine effect of some mycotoxins on humans: A clinical review of the ways to mitigate the action of mycotoxins. Toxins 2023, 15, 515. [Google Scholar] [CrossRef] [PubMed]
  215. Gong, Y.Y.; Cardwell, K.; Hounsa, A.; Egal, S.; Turner, P.C.; Hall, A.J.; Wild, C.P. Dietary aflatoxin exposure and impaired growth in young children from Benin and Togo: Cross-sectional study. Br. Med. J. 2002, 325, 20–21. [Google Scholar] [CrossRef] [PubMed]
  216. Kensler, T.W.; Eaton, D.L. 65 Years on—Aflatoxin Biomarkers Blossoming: Whither Next? Toxins 2024, 16, 496. [Google Scholar] [CrossRef]
  217. Al-Jaal, B.A.; Jaganjac, M.; Barcaru, A.; Horvatovich, P.; Latiff, A. Aflatoxin, fumonisin, ochratoxin, zearalenone and deoxynivalenol biomarkers in human biological fluids: A systematic literature review, 2001–2018. Food Chem. Toxicol. 2019, 129, 211–228. [Google Scholar] [CrossRef]
  218. Davwar, P.; David, P.; Imoh, L.; Duguru, M.; Zawaya, K.; Tsok, Y.; Sagay, A.; Okeke, E. Aflatoxin exposure in a population of HIV patients at risk of hepatocellular carcinoma, North-Central, Nigeria. Afr. Health Sci. 2023, 23, 81–87. [Google Scholar] [CrossRef]
  219. Jolly, P.E.; Akinyemiju, T.F.; Sakhuja, S.; Sheth, R. Association of aflatoxin B1 levels with mean CD4 cell count and uptake of ART among HIV infected patients: A prospective study. PLoS ONE 2022, 17, e0260873. [Google Scholar] [CrossRef]
  220. Madeen, E.P.; Maldarelli, F.; Groopman, J.D. Environmental pollutants, mucosal barriers, and pathogen susceptibility; the case for aflatoxin b1 as a risk factor for hiv transmission and pathogenesis. Pathogens 2021, 10, 1229. [Google Scholar] [CrossRef]
  221. Bekker, L.-G.; Beyrer, C.; Mgodi, N.; Lewin, S.R.; Delany-Moretlwe, S.; Taiwo, B.; Masters, M.C.; Lazarus, J.V. HIV infection. Nat. Rev. Dis. Primers 2023, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  222. Mutua, F.K.; Grace, D.; Watts, C. Food Safety Investments in East Africa; International Livestock Research Institute: Nairobi, Kenya, 2021; p. 12. [Google Scholar]
  223. Rasouli, H.; Nayeri, F.D.; Khodarahmi, R. May phytophenolics alleviate Aflatoxins-induced health challenges? A holistic insight on current landscape and future prospects. Front. Nutr. 2022, 9, 981984. [Google Scholar] [CrossRef] [PubMed]
  224. Dabuo, B.; Xorlali, N.; Amoliga, N.T.; Atibodu, Z.K.; Newman, P.M.; Mohammed, A.; Ali, R.A.; Abudu, A. Aspergillus and Aspergillosis in People with Chronic Diseases. 2023. Available online: https://www.intechopen.com/chapters/87304 (accessed on 29 April 2025).
  225. Gurunathan, S.; Lee, A.R.; Kim, J.H. Antifungal effect of nanoparticles against COVID-19 linked black fungus: A perspective on biomedical applications. Int. J. Mol. Sci. 2022, 23, 12526. [Google Scholar] [CrossRef] [PubMed]
  226. Malvandi, A.M.; Shahba, S.; Mehrzad, J.; Lombardi, G. Metabolic disruption by naturally occurring mycotoxins in circulation: A focus on vascular and bone homeostasis dysfunction. Front. Nutr. 2022, 9, 915681. [Google Scholar] [CrossRef]
  227. Ye, D.; Hao, Z.; Tang, S.; Velkov, T.; Dai, C. Aflatoxin exposure-caused male reproductive toxicity: Molecular mechanisms, detoxification, and future directions. Biomolecules 2024, 14, 1460. [Google Scholar] [CrossRef]
  228. Kannan, K.; George, J.A.; Sahadevan, R.; Kothari, M.; Sadhukhan, S. Insights into one drug, multi-target aspects of polyphenols for diabetes management: In vitro, in vivo, and clinical evidence. Phytochem. Rev. 2024, 1–49. [Google Scholar] [CrossRef]
  229. Liu, Y.; Hao, C.; Li, L.; Zhang, H.; Zha, W.; Ma, L.; Chen, L.; Gan, J. The role of oxidative stress in the development and therapeutic intervention of hepatocellular carcinoma. Curr. Cancer Drug Targets 2023, 23, 792–804. [Google Scholar] [CrossRef]
  230. Hasan, M.I.; Hossain, M.A.; Bhuiyan, P.; Miah, M.S.; Rahman, M.H. A system biology approach to determine therapeutic targets by identifying molecular mechanisms and key pathways for type 2 diabetes that are linked to the development of tuberculosis and rheumatoid arthritis. Life Sci. 2022, 297, 120483. [Google Scholar] [CrossRef]
  231. Thenuwara, G.; Javed, B.; Singh, B.; Byrne, H.J.; Tian, F. Sex-and Gender-Specific Considerations in Mycotoxin Screening: Assessing Differential Exposure, Health Impacts, and Mitigation Strategies. Microbiol. Res. 2024, 15, 2455–2492. [Google Scholar] [CrossRef]
  232. McGlynn, K.A.; Petrick, J.L.; El-Serag, H.B. Epidemiology of hepatocellular carcinoma. Hepatology 2021, 73, 4–13. [Google Scholar] [CrossRef]
  233. Saini, A.; Kumar, M.; Bhatt, S.; Saini, V.; Malik, A. Cancer causes and treatments. Int. J. Pharm. Sci. Res. 2020, 11, 3121–3134. [Google Scholar]
  234. Santacroce, M.P.; Conversano, M.; Casalino, E.; Lai, O.; Zizzadoro, C.; Centoducati, G.; Crescenzo, G. Aflatoxins in aquatic species: Metabolism, toxicity and perspectives. Rev. Fish Biol. Fish. 2008, 18, 99–130. [Google Scholar] [CrossRef]
  235. Marchese, S.; Polo, A.; Ariano, A.; Velotto, S.; Costantini, S.; Severino, L. Aflatoxin B1 and M1: Biological properties and their involvement in cancer development. Toxins 2018, 10, 214. [Google Scholar] [CrossRef] [PubMed]
  236. Mary, V.S.; Theumer, M.G.; Arias, S.L.; Rubinstein, H.R. Reactive oxygen species sources and biomolecular oxidative damage induced by aflatoxin B1 and fumonisin B1 in rat spleen mononuclear cells. Toxicology 2012, 302, 299–307. [Google Scholar] [CrossRef] [PubMed]
  237. Iqbal, M.J.; Kabeer, A.; Abbas, Z.; Siddiqui, H.A.; Calina, D.; Sharifi-Rad, J.; Cho, W.C. Interplay of oxidative stress, cellular communication and signaling pathways in cancer. Cell Commun. Signal. 2024, 22, 7. [Google Scholar] [CrossRef] [PubMed]
  238. Dai, C.; Tian, E.; Hao, Z.; Tang, S.; Wang, Z.; Sharma, G.; Jiang, H.; Shen, J. Aflatoxin B1 toxicity and protective effects of curcumin: Molecular mechanisms and clinical implications. Antioxidants 2022, 11, 2031. [Google Scholar] [CrossRef]
  239. Ráduly, Z.; Szabó, L.; Madar, A.; Pócsi, I.; Csernoch, L. Toxicological and medical aspects of Aspergillus-derived mycotoxins entering the feed and food chain. Front. Microbiol. 2020, 10, 2908. [Google Scholar] [CrossRef]
  240. Popescu, R.G.; Rădulescu, A.L.; Georgescu, S.E.; Dinischiotu, A. Aflatoxins in feed: Types, metabolism, health consequences in swine and mitigation strategies. Toxins 2022, 14, 853. [Google Scholar] [CrossRef]
  241. He, L.; He, T.; Farrar, S.; Ji, L.; Liu, T.; Ma, X. Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell. Physiol. Biochem. 2017, 44, 532–553. [Google Scholar] [CrossRef]
  242. Sadiq, I.Z. Free radicals and oxidative stress: Signaling mechanisms, redox basis for human diseases, and cell cycle regulation. Curr. Mol. Med. 2023, 23, 13–35. [Google Scholar] [CrossRef]
  243. Malik, S.; Krishnaswamy, K.; Mustapha, A. Hazard analysis and risk-based preventive controls (HARPC): Current food safety and quality standards for complementary foods. Foods 2021, 10, 2199. [Google Scholar] [CrossRef]
  244. Mishra, R.; Madhav, S.; Dhaka, R.K.; Garg, P. (Eds.) Biomarkers in Environmental and Human Health Biomonitoring: An Integrated Perspective; Elsevier: Amsterdam, The Netherlands, 2024; pp. 150–151. [Google Scholar]
  245. Dropulic, L.K.; Lederman, H.M. Overview of infections in the immunocompromised host. Diagn. Microbiol. Immunocompromised Host 2016, 4. [Google Scholar] [CrossRef]
  246. Omar, S.S.; Haddad, M.A.; Parisi, S. Validation of HPLC and Enzyme-Linked Immunosorbent Assay (ELISA) techniques for detection and quantification of Aflatoxins in different food samples. Foods 2020, 9, 661. [Google Scholar] [CrossRef]
  247. Baratta, F.; Pastori, D.; Angelico, F.; Balla, A.; Paganini, A.M.; Cocomello, N.; Ferro, D.; Violi, F.; Sanyal, A.J.; Del Ben, M. Nonalcoholic fatty liver disease and fibrosis associated with increased risk of cardiovascular events in a prospective study. Clin. Gastroenterol. Hepatol. 2020, 18, 2324–2331.e4. [Google Scholar] [CrossRef]
  248. Wenndt, A.; Mutua, F.; Grace, D.; Thomas, L.F.; Lambertini, E. Quantitative assessment of aflatoxin exposure and hepatocellular carcinoma (HCC) risk associated with consumption of select Nigerian staple foods. Front. Sustain. Food Syst. 2023, 7, 1128540. [Google Scholar] [CrossRef]
  249. Visser, M.E.; Schoonees, A.; Ezekiel, C.N.; Randall, N.P.; Naude, C.E. Agricultural and nutritional education interventions for reducing aflatoxin exposure to improve infant and child growth in low-and middle-income countries. Cochrane Database Syst. Rev. 2020, 2020, CD013376. [Google Scholar] [CrossRef]
  250. Wild, C.P.; Miller, J.D.; Groopman, J.D. Intervention Strategies to Reduce Human Exposure to Aflatoxins and Fumonisins. Mycotoxin Control in Low-and Middle-Income Countries; International Agency for Research on Cancer (IARC): Lyon, Paris, 2015; pp. 103–112. [Google Scholar]
  251. Smith, G.I.; Shankaran, M.; Yoshino, M.; Schweitzer, G.G.; Chondronikola, M.; Beals, J.W.; Okunade, A.L.; Patterson, B.W.; Nyangau, E.; Field, T.; et al. Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J. Clin. Investig. 2020, 130, 1453–1460. [Google Scholar] [CrossRef]
  252. Jones, D.L.; Morgan, K.E.; Martinez, P.C.; Rodriguez, V.J.; Vazquez, A.; Raccamarich, P.D.; Alcaide, M.L. COVID-19 burden and risk among people with HIV/AIDS. J. Acquir. Immune Defic. Syndr. 2021, 87, 869–874. [Google Scholar] [CrossRef]
  253. Lee, M.; Quinn, R.; Pradhan, K.; Fedorov, K.; Levitz, D.; Fromowitz, A.; Thakkar, A.; Shapiro, L.C.; Kabarriti, R.; Ruiz, R.E.; et al. Impact of COVID-19 on case fatality rate of patients with cancer during the Omicron wave. Cancer Cell 2022, 40, 343–345. [Google Scholar] [CrossRef]
  254. Liu, Y.; Wu, F. Global burden of aflatoxin-induced hepatocellular carcinoma: A risk assessment. Environ. Health Perspect. 2010, 118, 818–824. [Google Scholar] [CrossRef]
  255. Mupunga, I. A Comparative Study of Natural Contamination with Aflatoxins and Fumonisins in Selected Food Commodities from Botswana and Zimbabwe. University of South Africa. 2013. Available online: https://www.researchgate.net/publication/313888038_A_comparative_study_of_natural_contamination_with_aflatoxins_and_fumonisins_in_selected_food_commodities_from_Botswana_and_Zimbabwe (accessed on 29 April 2025).
Figure 1. Various AF exposure routes in humans.
Figure 1. Various AF exposure routes in humans.
Toxins 17 00414 g001
Figure 2. Biomarkers for AFs in biological samples.
Figure 2. Biomarkers for AFs in biological samples.
Toxins 17 00414 g002
Figure 3. AF biomonitoring in immunocompromised individuals.
Figure 3. AF biomonitoring in immunocompromised individuals.
Toxins 17 00414 g003
Table 1. Dietary exposure to aflatoxin.
Table 1. Dietary exposure to aflatoxin.
Food CommodityType of AflatoxinAnalytical MethodSample Size% ContaminationConcentration Range (ppb)References
MaizeAFsHPLC20080LOD-40.31[27]
GroundnutAFsHPLC180100LOD-162.40[27]
Breakfast cerealAFB1HPLC-FD2669LOD-0.13[58]
maizeAFsHPLC80052LOD-1369[59]
Maize fermented doughAFsHPLC3297LOD-310[60]
MaizeAFsHPLC-10020–355[60]
MaizeAFB1LC-MS/MS114471–149[61]
GingerAFsHPLC, ELISA1001003.63–411.1[62]
Processed maizeAFB1HPLC17650-[63]
Home-brewed beerAFsHPLC/TLC2928200–400[64]
GroundnutAFsELISA7593LOD-43.23[65]
Dried beefAFB1LC-MS/MS108663.91–295.41[66]
UHT MilkAFM1HPLC1154.50.013–0.219[67]
Dairy productsAFM1HPLC15645.50.015–7.350[67]
Rice and maizeAFsELISA32741.75–173.3[68]
CowpeaAFsLC-MS/MS81-LOD-209[69]
SorghumAFsELISA201004.80–42.60[70]
MilletAFsELISA201004.80–45.60[70]
Yam flourAFsELISA201005.0–39.45[70]
GarriAFsELISA201002.60–55.40[70]
RiceAFsELISA621002.10–248.20[70]
MilkAFM1HPLC-FD37256.1LOD-345.8[71]
MaizeAFsHPLC-FD180571.3–91.4[72]
MaizeAFs 30005–72LOD-76.2[73]
Milk and dairy productsAFM1ELISA1601000.137–0.319[55]
%—Percentage; AFs—total AFs; AFB1—aflatoxin B1; AFM1—aflatoxin M1; HPLC—High-Pressure Liquid Chromatography; HPLC-FD—High-Performance Liquid Chromatography with Fluorescence Detection; LC-MS/MS—Liquid Chromatography–Tandem Mass Spectrometry; ELISA—Enzyme-Linked Immunosorbent Assay; TLC—Thin-Layer Chromatography; LOD—Limit of Detection.
Table 2. Environmental and occupational exposure to AF.
Table 2. Environmental and occupational exposure to AF.
EnvironmentAnalytical MethodSample Size% ContaminationContamination Range (ppb)References
Sugar production factoryELISA15-6–11[101]
Sugar and paper-making factoryELISA181565.9–10.4[102]
Warehouses for green coffee,
black pepper, and cocoa
beans
HPLC44-LOD-0.023[103]
Farms handling grainsHPLC24- [104]
Animal feed productionELISA4520LOD-8[105]
Waste industryELISA411002.5–25.9[106]
Poultry productionELISA31591–4.23[107]
Textile industry 5833 [108]
Swine productionHPLC-MS/MS2516 [92]
Waste managementELISA41--[109]
Feed mill workersHPLC2810073.4–189.2[110]
Table 4. AF biomonitoring and its effects on diabetes, hepatitis, HIV, and cancer.
Table 4. AF biomonitoring and its effects on diabetes, hepatitis, HIV, and cancer.
ConditionPopulationBiomarkers AssessedType of AFAF Levels
ppb
Method UsedReferences
Diabetes15–55Glucose levels, Glycosylated, Heamoglobin (HbA1c)AFM11.86Elisa kits, Microplate Elisa readers, Spectrophotometric methods[23]
Pregnant women
HepatitisChildren, age 5–12Liver enzymes (ALT, AST)AFM115Serum Biochemical Tests, Enzyme Immunoassays, Liver Function Tests[251]
HIVAdults with HIV-positiveCD4 countAFB110Flow Cytometry, Immunoassays, Laboratory Blood Tests[252]
CancerCancer patients, various typesTumor markersAFB125Serum Biochemical Tests, ELISA[253]
HIVHIVHIV Viral LoadAFB1400 ng/kg body weight/daySerum Biochemical Tests[220]
CancerCancer patients in European population AFB110 ng/kg body weight per dayMargin of Exposure (MOE) approach
CancerHepatocellular Carcinoma in sub-Saharan AfricaHCCAFB15 to 500Quantitative cancer risk assessment[254]
CancerHepatocellular carcinoma in Southern AfricaHCCAFB1Peanut butter (6.8–250.)
Peanut (6.6–622.1)
(HPLC)[255]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fagbohun, T.R.; Nji, Q.N.; Okechukwu, V.O.; Adelusi, O.A.; Nyathi, L.A.; Awong, P.; Njobeh, P.B. Aflatoxin Exposure in Immunocompromised Patients: Current State and Future Perspectives. Toxins 2025, 17, 414. https://doi.org/10.3390/toxins17080414

AMA Style

Fagbohun TR, Nji QN, Okechukwu VO, Adelusi OA, Nyathi LA, Awong P, Njobeh PB. Aflatoxin Exposure in Immunocompromised Patients: Current State and Future Perspectives. Toxins. 2025; 17(8):414. https://doi.org/10.3390/toxins17080414

Chicago/Turabian Style

Fagbohun, Temitope R., Queenta Ngum Nji, Viola O. Okechukwu, Oluwasola A. Adelusi, Lungani A. Nyathi, Patience Awong, and Patrick B. Njobeh. 2025. "Aflatoxin Exposure in Immunocompromised Patients: Current State and Future Perspectives" Toxins 17, no. 8: 414. https://doi.org/10.3390/toxins17080414

APA Style

Fagbohun, T. R., Nji, Q. N., Okechukwu, V. O., Adelusi, O. A., Nyathi, L. A., Awong, P., & Njobeh, P. B. (2025). Aflatoxin Exposure in Immunocompromised Patients: Current State and Future Perspectives. Toxins, 17(8), 414. https://doi.org/10.3390/toxins17080414

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop