Next Article in Journal
Non-Invasive Analysis of Bulk and Surface Remodeling of Non-Woven PLLA and Fiber-Sponge PLLA/Chitosan Scaffolds in Cell Culture Environment
Previous Article in Journal
Nickel Phosphine Complexes: Synthesis, Characterization, and Behavior in the Polymerization of 1,3-Butadiene
Previous Article in Special Issue
Unlocking the Therapeutic Potential of Ellagitannins: A Comprehensive Review of Key Representatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cytotoxic and Antimicrobial Activity of the Ageratina Genus

by
Sarai Rojas-Jiménez
1,2,
David Osvaldo Salinas-Sánchez
3,4,*,
Verónica Rodríguez-López
5,
Roberta Salinas-Marín
1,
Dante Avilés-Montes
6,
César Sotelo-Leyva
7,
Rodolfo Figueroa-Brito
8,
Genoveva Bustos Rivera-Bahena
2,
Rodolfo Abarca-Vargas
9,
Dulce María Arias-Ataide
4 and
María Guadalupe Valladares-Cisneros
2,*
1
Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico
2
Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma del Estado de Morelos (UAEM), Av. Universidad 1001, Col. Chamilpa, Cuernavaca 62209, Morelos, Mexico
3
Centro de Investigación en Biodiversidad y Conservación, Universidad Autónoma del Estado de Morelos (UAEM), Av. Universidad 1001, Col. Chamilpa, Cuernavaca 62209, Morelos, Mexico
4
Escuela de Estudios Superiores Jicarero (EESJ), Universidad Autónoma del Estado de Morelos (UAEM), Carretera Galeana-Tequesquitengo s/n, Comunidad El Jicarero, Jojutla 62915, Morelos, Mexico
5
Facultad de Farmacia, Universidad Autónoma del Estado de Morelos (UAEM), Av. Universidad 1001, Col. Chamilpa, Cuernavaca 62209, Morelos, Mexico
6
Facultad de Ciencias Biológicas, Universidad Autónoma del Estado de Morelos (UAEM), Av. Universidad 1001, Col. Chamilpa, Cuernavaca 62209, Morelos, Mexico
7
Facultad de Ciencias Químico-Biológicas (FCQB), Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39090, Guerrero, Mexico
8
Centro de Desarrollo de Productos Bióticos (CEPROBI-IPN), Yautepec 62730, Morelos, Mexico
9
Facultad de Medicina, Universidad Autónoma del Estado de Morelos (UAEM), Leñeros, Esquina Iztazzíhuatl s/n, Col. Volcanes, Cuernavaca 62350, Morelos, Mexico
*
Authors to whom correspondence should be addressed.
Molecules 2025, 30(23), 4656; https://doi.org/10.3390/molecules30234656
Submission received: 28 October 2025 / Revised: 30 November 2025 / Accepted: 1 December 2025 / Published: 4 December 2025

Abstract

Medicinal plants have long been used for therapeutic purposes in many cultures. They represent sources of important bioactive compounds, often of pharmacological significance. Ageratina Spach is the largest genus in Mexico and is characterised by its traditional use in the treatment of cancer and infections of the skin, blood, and intestines. Different species of Ageratina have been biologically evaluated at the extract and compound levels, and their chemical contents have been purified and characterised. Following a PRISMA meta-analysis, 29 scientific reports were selected and analysed. Tables of different Ageratina species were integrated to compare their cytotoxic and antimicrobial activity at the extract and compound levels. Twelve pure and isolated natural compounds were tested for cytotoxic activity against several cell lines from lung, colon, and breast cancer, cervical carcinoma, hepatocarcinoma, promyelocytic leukaemia, and histiocytic lymphoma. Forty-one pure and isolated natural compounds were evaluated for antimicrobial activity against a wide spectrum of microorganisms, including Gram-positive and Gram-negative bacteria, yeast, fungi, parasites and viruses. Ageratina Spach contains cytotoxic and antimicrobial substances with broad chemical profiles. In addition to being a plant with active compounds, it could be useful for future rational drug design.

1. Introduction

Cancer is a disease in which the cell cycle is deregulated by DNA damage, leading to uncontrolled cell growth and the potential for metastasis to different parts of the body. The most common treatment for cancer is chemotherapy, which produces side effects and can result in resistance to the compounds used [1]. Cancer is a prevalent disease worldwide. In 2022, there were already almost 20 million new cases globally, and by 2050, this number is projected to increase to approximately 33 million [2]. Global cancer incidence is a significant public health issue, and projections show a continued increase in cases and deaths, driven by factors like population growth and ageing [3,4]. On the other hand, antimicrobial-resistant infections caused by bacteria, fungi, parasites, and viruses are continually increasing worldwide, making it urgent to prevent and treat them effectively [5,6,7].
Worldwide research on medicinal plants is needed to develop new treatments for diseases. The World Health Organization (WHO) recognises the use of traditional medicine for therapeutic purposes, as it is the oldest known healthcare system, especially in developing countries [8]. Medicinal plants are an important source of natural products for treating different health problems. As research on medicinal plants has increased, about 80% of chemotherapeutic drugs have been derived from plants [9].

Ethnobotany

Ageratina Spach is a native New World genus in the tribe Eupatorieae of the Asteraceae family [10]. This genus comprises 316 accepted species of Ageratina. They are distributed from the United States to South America; Mexico has the largest number of species. However, some species of Ageratina have been introduced into some countries in Europe, Africa, Asia, and Oceania [11]. Ageratina spp. are perennial herbs, shrubs, or small trees less than 10 m high. The florets are white or lavender, ranging from a few (about 10) to many per head (50 or more), with tubular corollas and poorly defined throats (Figure 1); apically, they include five lobes that are generally well defined, pubescent, or glabrous [12].
Species of the genus Ageratina grow in warm, humid climates with abundant rainfall, because these plants absorb water through their roots, retaining it in their stems, and benefiting from the humidity of their surroundings. Consequently, their distribution and dispersal are primarily in mountainous areas, near rivers, streams, or ravines [13]. Studies of some Ageratina species have demonstrated their ability to develop in a wide variety of soils with different pH levels, as well as tolerating some salinity and low nutrient levels. This has allowed species of this genus to be ecologically adaptable and widely distributed [14,15].
In Mexico, Ageratina spp. are widely used in traditional Mexican medicine, commonly known as ‘axihuitl’, which means water root. Axihuitl is very commonly used to treat numerous diseases, including persistent gastric ulcers, bacterial and fungal infections, and cancer. It is also employed to treat women after childbirth, fungal infections of the feet, and for heat treatment [16,17]. This review provides a current overview of scientific studies on Ageratina species, offering comprehensive and critical information on their chemistry and pharmacological activities worldwide from 2010 to 2025.

2. Results

The PRISMA guidelines [18] were used to ensure transparency and completeness of the systematic review process (Figure 2).
Identification: The researchers conducted a literature review across several databases from 2010 to 2025. The records identified 200 relevant studies on Scopus and 266 on PubMed. Additionally, a manual search using Google Scholar found 708 studies. In addition to the automated database search to ensure that no relevant studies were overlooked (286), 80 studies were removed using the term ‘biological evaluation’; they were marked as ineligible. Screening: After removing duplicates, 200 articles remained. After reviewing the titles and abstracts, 144 articles were excluded, leaving 56 for full-text screening. Eligibility: The remaining articles were read in full and assessed. Five studies were excluded as they were reviews. Then, 51 reports were assessed, and 22 were excluded because they included different activities, and some others were from different evaluations.
Inclusion: Finally, 29 papers were included in this systematic review, all of which reported cytotoxic and antimicrobial activities. These articles were analysed in detail to answer the study’s research question.
Table 1 shows the different species of the Ageratina genus identified by the PRISMA meta-analysis and included in this review. The species are presented in alphabetical order.
Table 2 shows the cytotoxic activity of the organic and aqueous extracts obtained from different Ageratina species.
Ageratina gracilis is a unique species that has lower IC50 values at the extract level; the inflorescent petrol extract was active against HT29 cells (IC50 = 12.67 + 1.13 μg/mL) and the leaf petrol extract was active against HT29 (IC50 = 11.20 + 1.20 μg/mL); SiHa (IC50 = 12.91 + 0.92 μg/mL), and MDA-MB-231 cells (IC50 = 14.72 + 0.69 μg/mL) [36].
Table 3 shows the cytotoxic activity of the diverse pure isolated natural compounds from the different Ageratina species.
The pure natural compounds isolated from Ageratina Spach and bio-evaluated in an in vitro anticancer assay are shown in Figure 3.
According to the IC50 values established to distinguish an active cytotoxic substance [5], the compounds with good cytotoxic activity were from Ageratina adenophora. A new tricyclic cadinene (1) was isolated and moderately active against A549 (IC50 = 11.45 + 0.69 μM) and SMMC-7721 cells (IC50 = 9.96 + 1.45 μM). Also, (+)-(5R,7S,9R,10S)-2-oxocadinan-3,6(11)-dien-12,7-olide (2) was isolated, which was moderately active against A549 (IC50 = 9.85 + 0.88 μM), SMMC-7721 (IC50 = 13.44 + 2.32 μM), and MDA-MB-231 cells (IC50 = 12.72 + 1.58 μM). Cadinene norsesquiterpenoid (3) was isolated from A. adenophora and was moderately active against SMMC-7721 cells (IC50 = 10.28 + 1.67 μM) [27]. 9-oxo-10,11-dehydro-ageraphorone (Euptox A) (4) was cytotoxically active against HeLa (IC50 = 0.55 + 0.05 mg/mL), Caco-2 (IC50 = 1.43 + 0.08 mg/mL), and MCF-7 cells (IC50 = 1.63 + 0.08 mg/mL) [23]. Quercetin 3,7-dimethylether (11), isolated from Ageratina dictyoneura, was strongly active against U-937 cells (IC50 = 7.0 + 0.5 μM), and 8β-hydroxy-β-cyclocostunolide (12), isolated from Ageratina illita, was also strongly active against U-937 cells (IC50 = 6.0 + 0.7 μM) [34].
Table 4 shows the antimicrobial activity of the organic and aqueous extracts from Ageratina evaluated in vitro against different microorganisms: Gram-positive and Gram-negative cocci and bacilli, and yeast, fungi, protozoa, and viruses.
According to the minimal inhibitory concentration (MIC) values for distinguishing an active antimicrobial substance [48], we observed that the essential oil from Ageratina pentlandiana leaves was active against S. aureus (MIC = 11.9 + 0.1 μL/mL), Bacillus subtilis (22.7 + 0.3 μL/mL), Escherichia coli (57.7 + 0.1 μL/mL), and Salmonella thyphimurium (41.6 + 0.1 μL/mL) [45].
Table 5 shows the antimicrobial activity of the compounds isolated from species of Ageratina Spach and evaluated in vitro against different microorganisms: Gram-positive and Gram-negative cocci and bacilli, yeast, fungi, protozoa, and viruses.
The pure natural compounds isolated from Ageratina Spach and bio-evaluated in vitro antimicrobial assays are shown in Figure 4.
Deltoidin A (24) from Ageratina deltoide was active against E. coli (MIC = 16 μg/mL) [32]. (8S)-10-Benzoyloxy-8,9-epoxy-6-hydroxythymol isobutyrate (26) was active against Entamoeba histolytica (IC50 = 1.6 μM). 10-Benzoyloxy-8,9-epoxy-6-acetyloxythymol isobutyrate (27) was active against E. histolytica (IC50 = 0.84 μM) and Giardia lamblia (IC50 = 24.4 μM) [35]. (−)-(5S,9S,10S,13S)-labd-7-en-15-oic acid (34) was active against Staphylococcus aureus (MIC = 0.78 mg/mL) and Bacillus subtilis (MIC = 0.15 mg/mL); (−)-(5S,9S,10S,13Z)-labda-7,13-dien-15-oic acid (35) was active against S. aureus (MIC = 1.0 mg/mL). (+)-(5S,8R,9R,10S,13R)-8-hydroxylabdan-15-oic acid (36) was active for S. aureus; all of these compounds were isolated from Ageratina jocotepecana [41]. Encecalin (37) was active against Fusarium oxysporum, producing a 10 mm inhibition zone. 7-hydroxy-dehydrotremetone (38) produced an inhibition zone for Colletotrichum gloesporoides (17.28 mm), Colletotrichum musae (17.24 mm), Rhizoctonia solani (16.40 mm), F. oxysporum (13.90 mm), and Alternaria alternata (14.36 mm). All of these compounds were isolated from Ageratina adenophora [25]. All compounds isolated from Ageratina cylindrica were active for E. histolytica and G. lamblia [30].

3. Discussion

The study of medicinal plants through their organic or aqueous extracts and their pure natural products continues to be of great importance, because they afford a wide range of compounds with pharmacological activity against significant diseases, such as cancer and microbial, fungal, parasitic, and viral infections, among others. According to the WHO, these diseases are the leading causes of mortality worldwide [5,48]. For this purpose, it is of the utmost importance to conduct reviews of scientific studies that provide an overview of the extracts and molecules isolated from plants used in traditional medicine, such as those of the Ageratina Spach. In this review, 29 scientific papers were selected through a PRISMA meta-analysis from 2010 to 2025, covering the anticancer and antimicrobial effects of 47 compounds isolated from 16 species of the Ageratina genus.
The criteria for cytotoxic activity of a plant as a promising crude extract for the purification of active compounds, according to the American National Cancer Institute (NCI), USA, are an IC50 < 20 μg/mL [48,49,50,51] and an IC50 < 4 μg/mL (or <10 μM) for an moderately and high active cytotoxic natural pure compound, respectively [52]. The cytotoxic activity at extract level of the inflorescent petrol extract from A. gracilis had lower IC50 values against SiHa (IC50 = 21.19 ± 1.25 μg/mL) and HT29 cells (IC50 = 12.67 ± 1.13 μg/mL) and the petrol extract of the leaves from the same species was active against SiHa (IC50 = 12.91 ± 0.92 μg/mL), HT29 (IC50 = 11.20 ± 1.20 μg/mL), and MDA-MB-231 cells (IC50 = 14.72 ± 0.69 μg/mL) [36]. Those indicate that nonpolar extract from A. gracilis contains interesting cytotoxic active compounds.
A variability of active pure compounds isolated from Ageratina Spach, according to their chemical structure, were terpenes (mono- and sesqui-), cinnamic acids, flavonoids, and their glucosides. Twelve pure natural compounds isolated from Ageratina species were evaluated against different human cancer cell lines, including breast, lung, hepatic, uterine, colon, skin, prostatic, leukaemia, and lymphoma. The results showed that, according to the IC50 values established from NCI, the active cytotoxic pure natural products were a new tricyclic cadinene (1) isolated from A. Adenophora, which was active against A549 (IC50 = 11.45 ± 0.69 μM) and SMMC-7721 cells (IC50 = 9.96 ± 1.45 μM). The (+)-(5R,7S,9R,10S)-2-oxocadinan-3,6(11)-dien-12,7-olide (2) was also isolated from A. Adenophora, and was active against A549 (IC50 = 9.85 ± 0.88 μM), and MDA-MB-231 cells (IC50 = 12.72 ± 1.58 μM). Cadinene norsesquiterpenoid (3) was also isolated from A. Adenophora and was active only against SMMC-7721 cells (IC50 = 10.28 ± 1.67 μM) [27]. On the other hand, Quercetin 3,7-dimethylether (11), isolated from A. dictyoneura, was active against U-937 cells (IC50 = 7.0 ± 0.5 μM) and 8β-hydroxy-β-cyclocostunolide (12), isolated from A. illita, was also active against U-937 cells (IC50 = 6.0 ± 0.7 μM) [34].
Compounds 1, 2, and 3 are sesquiterpene lactones isolated from A. adenophora. Compounds 1 and 2 were reported to be active for A549 cells. Compounds 1 and 3 were reported to be active for human hepatocellular carcinoma (SMMC-7721), and compound 2 was active for breast cancer (MDA-MB-231). Likewise, compound 12 isolated from A. dictyoneura is also a lactone, and it was active against histiocytic lymphoma (U937 cells) like the flavonoid 11 isolated from A. illita. Lactones consistently induce programmed cell death in lung, breast, liver, and leukaemia cancer cell lines [53,54,55]. And flavonoids induce apoptosis, affecting the cell cycle and modulating crucial signalling pathways [55,56]. The molecular mechanisms for cytotoxic activity from lactones and flavonoids are associated with alterations in the balance of Bcl-2 family proteins (increasing pro-apoptotic Bax and decreasing anti-apoptotic Bcl-2) and the activation of caspases-9 and -3 [57,58,59,60]. Other observations are that lactones target crucial signalling cascades, vital for cancer cell survival and progression. A key mechanism involves suppressing the NK-kB pathway, which is often overactive in cancer cells and regulates genes for survival, proliferation, and inflammation [61,62,63].
Likewise, plants contain abundant compounds, some of which have antimicrobial properties, providing them with protection against aggressors, especially microorganisms. Since the earliest times, plants have been used by various communities as medicine to treat a wide range of diseases, including infections. Multiple studies on medicinal plants have been conducted because they constitute a potential source of interesting compounds that warrant investigation for their antimicrobial activity and possess chemical structures that support further study and design to enhance the effects of new medicines [64]. The criteria for selecting organic or aqueous plant extracts and pure natural compounds with potential antimicrobial activity are based on the MIC, a key criterion used by many scientists to select antimicrobial botanicals. Plant extracts with MICs ≤ 100 μg/mL reveal significant antimicrobial activity. However, pure natural compounds with MICs < 10 μg/mL (or <25 μM) indicate strong antimicrobial activity, a characteristic of many pure natural compounds, and suggest that they may be considered potential candidates for developing a novel low-toxicity antitumor agent [65,66,67].
According to the MIC values for distinguishing active antimicrobial substances [64,68], the antimicrobial activities of Ageratina species against fungi, yeast, protozoa, and viruses have been observed in a few pure natural compounds. Macranthoin F (20) and Macranthoin G (21) were active against S. enterica, with MICs of 14.7 and 7.4 μM, respectively; those compounds were isolated from A. adenophora [24]. On the other hand, (8S)-10-Benzoyloxy-8,9-epoxy-6-hydroxythymol isobutyrate (26) and 10-Benzoyloxy-8,9-epoxy-6-acetyloxythymol isobutyrate (27) isolated from Ageratina glabrata were bioactive against E. histolytica, IC50 = 1.6 and 0.84 μM, respectively [35]. These important findings allow us to observe the activity of these natural compounds against opportunistic enteric pathogens, many of which are now multi-resistant [69,70].
Quinic acid derivatives such as caffeoylquinic acid ester and compounds 20 and 21 employ multi-targeted mechanisms against S. enterica, primarily by interfering with bacterial virulence, disrupting cell membrane integrity, and modulating host immune responses [71,72]. Quinic acid and its derivatives increase the permeability of the microorganism membrane and disrupt the cell wall structure, leading to leakage of intracellular components and cell death [73].
The high lipophilicity of monoterpenes (such as thymol and carvacrol) and their derivatives, like compounds 26 and 27, is a multi-targeted approach that disrupts membrane integrity against Entamoeba histolytica, which is the primary mechanism of action. They readily integrate into the lipid bilayer of the parasite’s membrane and increase permeability, causing microorganisms to die by depolarization and a critical ionic disruption in homeostasis and enzymatic functions, which are necessary for survival. The compounds interact with and may cause oxidation of vital parasite proteins, including signalling molecules and oxidoreductases, thereby triggering parasite death [74,75,76].
The results indicate that natural compounds isolated from Ageratina Speech exhibit low activity against microorganisms associated with important infections such as amoebiasis, as well as giardiasis, which cause chronic diarrhoea [77,78,79]. However, it is important to continue to carry out investigations into medicinal plants used in traditional medicine against methicillin-resistant S. aureus and fungi, parasites, and viruses, because they contain valuable compounds that could exhibit effective activity against currently resistant pathogens [80,81,82].

4. Materials and Methods

A search for scientific studies on different species of Ageratina Spach was conducted across the Springer®, PubMed®, ScienceDirect®, and Google Scholar® online databases. The searching methodology used in this work is as follows:
Information sources: The search is conducted across multiple electronic databases, including Scopus, PubMed, and Google Scholar.
Search terms: A detailed search string will be constructed using a combination of keywords, Boolean operators, and medical subject headings (MeSH). For example: (‘cytotoxic activity’ AND ‘antimicrobial activity’ AND ‘Ageratina’), (‘anticancer activity’ OR ‘antifungal’ OR ‘antiviral’ OR ‘Ageratina’).
Search limitations: Searches are limited by date to the timeframe 2010 to 2025.
Additional sources: The reference lists of included studies and relevant review articles will be hand-searched to find additional studies.
Population/Intervention: Studies that investigate the cytotoxic or antimicrobial activity of specific Ageratina species.
Outcome: Quantitative data on cytotoxic effects (e.g., IC50 values) and antimicrobial effects (e.g., minimum inhibitory concentration—MIC, or zone of inhibition).
Study design: Eligible study designs typically include in vitro and, if relevant, in vivo studies, depending on the review’s scope.
Selection process: The screening process will involve two independent reviewers to reduce bias.
Title and abstract screening: Reviewers will screen titles and abstracts based on the eligibility criteria.
Full-text review: The full text of potentially eligible studies will be retrieved and assessed.
Documentation: The study selection process will be summarised in a PRISMA 2020 flow diagram, clearly indicating the number of studies identified, screened, and ultimately included or excluded.
Data extraction form: A standardised, pre-designed data extraction form will be used to ensure consistency.
Study characteristics: Author, year of publication, study design, and location.
Compound details: Name, source (plant, plant extract, isolated compound), concentration tested, and other relevant properties.
Cytotoxic data: Cell line tested, duration of exposure, and quantitative measures such as IC50 values.
Antimicrobial data: Pathogen tested (e.g., bacterium, fungus), assay method, and quantitative measures such as MIC or zone of inhibition.
Cytotoxicity: Quantitative pooling of IC50 values may be possible for studies using the same cell line and compound class.
Antimicrobial activity: Pooled estimates for MIC values or zones of inhibition may be calculated for studies focusing on the same pathogen and compound.

5. Future Perspectives

A few of the globally medicinal plants have been scientifically investigated. An infinite number of bioactive compounds, chemical and pharmacological, are unexplored and underexploited in medicinal plants. Traditional Latin America’s medicine, particularly traditional Mexican medicine [83], is an important natural source for the discovery of cytotoxic and resistance compounds that could become useful therapeutic tools [84,85,86].
However, the interactions between medicinal plant extracts and anticancer or antimicrobial agents can be either favourable, such as synergism, or harmful, as in antagonism [87]. Therefore, further studies are required, especially in vivo studies and research on the toxicity of these products, for them to be recognised as biomedical agents.
This work shows the considerable potential of complex natural compound mixtures from the Ageratina species, and suggests future investigations involving precise chemical and scientific explorations, because this species has adequate in vitro cytotoxic and antimicrobial potential. In the future, this should be followed by in vitro testing with other cancer cells or in vivo testing in infected animal models to determine the preclinical relevance of such compounds and to establish a valid correlation with the in vitro efficacy results [88]. New studies should include molecular docking, structural chemical modifications, or synthesis of derivatives to improve pharmacokinetics and pharmacodynamics, as well as chemical structure–activity relationship analysis or green synthesis of nanoparticles [81,89].
Advanced techniques of biotechnological, genomics, proteomics, and metabolomics are nowadays applied to the research of medicinal plants and contribute to the advancement of alternative natural antimicrobials, characterisation of their interactions, and elucidation of the mechanisms of natural compounds’ action [90,91,92].
Likewise, further studies are required for extracts and individual isolated natural compounds. We especially suggest conducting toxicity assays and toxic in vivo studies of these natural compounds in order for them to be recognised as biomedical agents [93].

6. Conclusions

In summary, this work compiles the studies on Ageratina Spach published from 2010 to 2025; these medicinal plants are a natural source of compounds with potent antimicrobial activity and moderate cytotoxicity. Sixteen species of Ageratina have been studied; however, A. adenophora is the predominant species and has received more research attention. Additionally, 47 compounds included in this review showed chemical structures such as terpenes (mono- and sesqui-), cinnamic acids, flavonoids, and their glucosides, all of which are natural products that exhibit antimicrobial activity.
Medicinal plants have been extensively studied as a potential source of compounds used in the treatment of various human diseases, including cancer, antibiotic-resistant microorganisms, parasites, and viruses. Public health is becoming a global issue; it is crucial to evaluate the potential of plant-derived natural products and assess how well they can combat both current and emerging pathogens.

Author Contributions

Conceptualization, M.G.V.-C., D.O.S.-S. and S.R.-J.; Investigation, M.G.V.-C., D.O.S.-S., S.R.-J., D.A.-M., V.R.-L., R.A.-V. and R.S.-M.; Methodology, S.R.-J., R.F.-B., R.A.-V. and G.B.R.-B.; Resources, M.G.V.-C., D.O.S.-S., D.A.-M., C.S.-L. and D.M.A.-A.; Supervision, M.G.V.-C. and D.O.S.-S.; Validation, M.G.V.-C., D.O.S.-S., S.R.-J., D.A.-M. and C.S.-L.; Writing—original draft, M.G.V.-C., D.O.S.-S. and S.R.-J.; Writing—review and editing, M.G.V.-C., D.O.S.-S., V.R.-L., R.S.-M. and D.A.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article as no new datasets were created or analysed in this study. All data used were obtained from publicly available literature accessed via the Springer®, PubMed®, ScienceDirect®, and Google Scholar® online databases, as described in the Methods Section.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
A549Lung cancer
APAerial parts
ATTCAmerican Type Culture Collection
BioModBiological model
BaxBcl-2-associated X protein
Bcl-2B-cell lymphoma 2
CCLCancer cell line
CC5050% Cytotoxic Concentration
Caco-2Colon cancer
Calu-1Epidermoid carcinoma of the lung
DNADeoxyribonucleic acid
DCMDichloromethane
DfDiffusion
Df discDiffusion disc
DIZDiameter of Inhibitory Zone
EC5050% Effective Concentration
EtOAcEthyl acetate
EO-HdestEssential oil by Hydro distillation
EOEssential Oil
FFlowers
FSFlowering stage
IC50Fifth inhibitory concentration
H-EtOAcHexane-Ethyl acetate
HCT-116Adherent cell of colon cancer
HCT-15Colorectal carcinoma
HeLaCervical carcinoma
HepG2Hepatocarcinoma
HT29Colorectal adenocarcinoma
HL-60Promyelocytic leukaemia
HSV-1Herpes simplex virus type 1
HSV-2Herpes simplex virus type 1
IInflorescence
IC50Half-maximal Inhibitory Concentration
KBEpithelial carcinoma
LLeaves
LC50Fifth Lethal Concentration;
L&FLeaves and flowers
L&SLeaves and stems
mMeters
MICMinimal inhibitory concentration
MBCMinimal bactericide concentration
MDA-MB-231Human breast cancer
MCF-7Breast cancer
MRSAMethicillin-resistant Staphylococcus aureus
MTT3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide
MTS3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-H-tetrazolium
MTT/PMSTetrazolium salt/phenazine methosulfate
μdmicrodilution
NANitazoxanide assay
NK-kBNuclear Factor kappa-light-chain-enhancer of activated B cells
OVCAROvarian adenocarcinoma
PC-3Prostatic adenocarcinoma
PP/ESPart of the plant/extraction solvent
RRoot(s)
SStems
SiHaUterine squamous cell carcinoma
SCASuspension cell assay
SMMC-7721uman hepatocellular carcinoma
SW480Human colon carcinoma
4T1Breast cancer
U-937Histiocytic lymphoma
UISOMerkel carcinoma cell
VIICEVirus inhibition induced cytopathic effect
VSVaginal suppositories
VStVegetative stage
Verokidney-derived epithelial cell
WHOWorld Health Organization

References

  1. Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of multidrug resistance in cancer chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef]
  2. Garg, P.; Malhotra, J.; Kulkarni, P.; Horne, D.; Salgia, R.; Singhal, S.S. Emerging therapeutic strategies to overcome drug resistance in cancer cells. Cancers 2024, 16, 2478. [Google Scholar] [CrossRef] [PubMed]
  3. Bizuayehu, H.M.; Ahmed, K.Y.; Kibret, G.D.; Dadi, A.F.; Belachew, S.A.; Bagade, T.; Tegegne, T.K.; Venchiarutti, R.L.; Kibret, K.T.; Hailegebireal, A.H.; et al. Global disparities of cancer and its projected burden in 2050. JAMA Netw. Open 2024, 7, e2443198. [Google Scholar] [CrossRef] [PubMed]
  4. Soerjomataram, I.; Bray, F. Planning for tomorrow: Global cancer incidence and the role of prevention 2020–2070. Nat. Rev. Clin. Oncol. 2021, 18, 663–672. [Google Scholar] [CrossRef] [PubMed]
  5. WHO. Noncommunicable Diseases Progress Monitor 2025; World Health Organization: Geneva, Switzerland, 2025; Available online: https://www.who.int/publications/i/item/9789240105775 (accessed on 19 October 2025).
  6. Oliveira, M.; Antunes, W.; Mota, S.; Madureira-Carvalho, Á.; Dinis-Oliveira, R.J.; Dias da Silva, D. An overview of the recent advances in antimicrobial resistance. Microorganisms 2024, 12, 1920. [Google Scholar] [CrossRef]
  7. Urban-Chmiel, R.; Marek, A.; Stępień-Pyśniak, D.; Wieczorek, K.; Dec, M.; Nowaczek, A.; Osek, J. Antibiotic resistance in bacteria—A review. Antibiotics 2022, 11, 1079. [Google Scholar] [CrossRef]
  8. Aarland, R.C.; Peralta-Gómez, S.; Sánchez-Morales, C.; Parra-Bustamante, F.; Villa-Hernández, J.M.; de León-Sánchez, F.D.; Pérez-Flores, L.J.; Rivera-Cabrera, F.; Mendoza-Espinoza, J.A. A pharmacological and phytochemical study of medicinal plants used in Mexican folk medicine. Indian J. Tradit. Knowl. 2015, 14, 550–557. [Google Scholar]
  9. Pirintsos, S.; Panagiotopoulos, A.; Bariotakis, M.; Daskalakis, V.; Lionis, C.; Sourvinos, G.; Karakasiliotis, I.; Kampa, M.; Castanas, E. From traditional ethnopharmacology to modern natural drug discovery: A methodology discussion and specific examples. Molecules 2022, 27, 4060. [Google Scholar] [CrossRef]
  10. King, R.M.; Robinson, H.E. The genera of the Eupatorieae (Asteraceae). In Monographs in Systematic Botany from the Missouri Botanical Garden; Missouri Botanical Garden: St. Louis, MO, USA, 1987; pp. 1–581. [Google Scholar]
  11. Royal Botanic Gardens. Ageratina . Available online: https://powo.science.kew.org/taxon/urn:lsid:ipni.org:names:30003158-2 (accessed on 29 September 2022).
  12. Clewell, A.F.; Wooten, J.W. A revision of Ageratina (Compositae: Eupatorieae) from eastern North America. Brittonia 1971, 23, 123–143. [Google Scholar] [CrossRef]
  13. Zhang, X.; Wang, Y.; Peng, P.; Wang, G.; Zhao, G.; Zhou, Y.; Tang, Z. Mapping the Distribution and Dispersal Risks of the Alien Invasive Plant Ageratina adenophora in China. Diversity 2022, 14, 915. [Google Scholar] [CrossRef]
  14. Xian, X.; Zhao, H.; Wang, R.; Zhang, H.; Chen, B.; Huang, H.; Liu, W.; Wan, F. Predicting the potential geographical distribution of Ageratina adenophora in China using equilibrium occurrence data and ensemble model. Front. Ecol. Evol. 2022, 10, 973371. [Google Scholar] [CrossRef]
  15. Kim, E.; Choi, J.; Song, W. Introduction and Spread of the Invasive Alien Species Ageratina altissima in a Disturbed Forest Ecosystem. Sustainability 2021, 13, 6152. [Google Scholar] [CrossRef]
  16. Flores, A.G.; Enríquez, M.I.A.; González, J.B.C.; Miranda, D.M.V.; Bahena, C.Y.M.; Moreno, J.M.P. Plantas útiles de los patios de Santo Domingo, Ocotitlán, Tepoztlán, Morelos, México [Useful Plants of the Playgrounds of Santo Domingo, Ocotitlán, Tepoztlán, Morelos, México]. Trop. Subtrop. Agroecosyst. 2020, 23, 50. [Google Scholar]
  17. Monroy, C.; Castillo, P. Plantas Medicinales Utilizadas en el Estado de Morelos; Universidad Autónoma del Estado de Morelos: Cuernavaca, Mexico, 2000; pp. 23–24. [Google Scholar]
  18. Crompton, H.; Burke, D. Artificial intelligence in higher education: The state of the field. Int. J. Educ. Technol. High. Educ. 2023, 20, 22. [Google Scholar] [CrossRef]
  19. Chaiwaree, S.; Pongpaibul, Y.; Thammasit, P. Anti-dermatophyte activity of the aqueous extracts of Thai medicinal plants. Braz. J. Biol. 2022, 82, e254291. [Google Scholar] [CrossRef]
  20. Manandhar, S.; Luitel, S.; Dahal, R.K. In vitro antimicrobial activity of some medicinal plants against human pathogenic bacteria. J. Trop. Med. 2019, 2019, 1895340. [Google Scholar] [CrossRef]
  21. Chanu, K.D.; Sharma, N.; Kshetrimayum, V.; Chaudhary, S.K.; Ghosh, S.; Haldar, P.K.; Mukherjee, P.K. Ageratina adenophora (Spreng.) King & H. Rob. Standardized leaf extract as an antidiabetic agent for type 2 diabetes: An in vitro and in vivo evaluation. Front. Pharmacol. 2023, 14, 1178904. [Google Scholar] [CrossRef]
  22. Mani, S.; Natesan, K.; Shivaji, K.; Balasubramanian, M.G.; Ponnusamy, P. Cytotoxic effect induced apoptosis in lung cancer cell line on Ageratina adenophora leaf extract. Biocatal. Agric. Biotechnol. 2019, 22, 101381. [Google Scholar] [CrossRef]
  23. André, R.; Catarro, J.; Freitas, D.; Pacheco, R.; Oliveira, M.C.; Serralheiro, M.L.; Falé, P.L. Action of euptox A from Ageratina adenophora juice on human cell lines: A top-down study using FTIR spectroscopy and protein profiling. Toxicol. Vitr. 2019, 57, 217–225. [Google Scholar] [CrossRef]
  24. Zhang, M.; Liu, W.X.; Zheng, M.F.; Xu, Q.L.; Wan, F.H.; Wang, J.; Lei, T.; Zhou, Z.Y.; Tan, J.W. Bioactive quinic acid derivatives from Ageratina adenophora. Molecules 2013, 18, 14096–14104. [Google Scholar] [CrossRef]
  25. Zheng, G.; Luo, S.; Li, S.; Hua, J.; Li, W.; Li, S. Specialized metabolites from Ageratina adenophora and their inhibitory activities against pathogenic fungi. Phytochemistry 2018, 148, 57–62. [Google Scholar] [CrossRef]
  26. Luo, J.H.; Gu, W.J.; Zhang, E.B.; Huang, Y.; Zhang, Y.H.; Yang, Y.X.; Geng, H. Three new cadinene-type sesquiterpenoids from the aerial parts of Ageratina adenophora. Nat. Prod. Res. 2023, 39, 2320–2324. [Google Scholar] [CrossRef] [PubMed]
  27. Dong, L.M.; Zhang, M.; Xu, Q.L.; Zhang, Q.; Luo, B.; Luo, Q.W.; Liu, W.B.; Tan, J.W. Two new thymol derivatives from the roots of Ageratina adenophora. Molecules 2017, 22, 592. [Google Scholar] [CrossRef] [PubMed]
  28. Luo, B.; Dong, L.M.; Xu, Q.L.; Zhang, X.; Zhang, Q.; Liu, W.B.; Tan, J.W. A new monoterpene and a new sesquiterpene from the roots of Ageratina adenophora. Phytochem. Lett. 2018, 24, 67–70. [Google Scholar] [CrossRef]
  29. Bustos-Brito, C.; Sanchez-Castellanos, M.; Esquivel, B.; Calderon, J.S.; Calzada, F.; Yepez-Mulia, L.; Hernández-Barragán, A.; Joseph-Nathan, P.; Cuevas, G.; Quijano, L. Structure, absolute configuration, and antidiarrheal activity of a thymol derivative from Ageratina cylindrica. J. Nat. Prod. 2014, 77, 358–363. [Google Scholar] [CrossRef]
  30. Bustos-Brito, C.; Sánchez-Castellanos, M.; Esquivel, B.; Calderón, J.; Calzada, F.; Yépez-Muilia, L.; Joseph-Nathan, P.; Cuevas, G.; Quijano, L. ent-Kaurene Glycosides from Ageratina cylindrica. J. Nat. Prod. 2015, 78, 2580–2587. [Google Scholar] [CrossRef]
  31. Bustos-Brito, C.; Esquivel, B.; Calzada, F.; Yepez-Mulia, L.; Calderón, J.S.; Porras-Ramirez, J.; Quijano, L. Further thymol derivatives from Ageratina cylindrica. Chem. Biodivers. 2016, 13, 1281–1289. [Google Scholar] [CrossRef]
  32. Arciniegas, A.; Pérez-Castorena, A.L.; Meléndez-Aguirre, M.; Ávila, J.G.; García-Bores, A.M.; Villaseñor, J.L.; Romo de Vivar, A. Chemical composition and antimicrobial activity of Ageratina deltoidea. Chem. Biodivers. 2018, 15, e1700529. [Google Scholar] [CrossRef]
  33. Valarezo, E.; Jaramillo-Jaramillo, E.; Carrión-Campoverde, A.; Morocho, V.; Jaramillo-Fierro, X.; Cartuche, L.; Meneses, M.A. A Study of the Essential Oil Isolated from Ageratina dendroides (Spreng.) RM King & H. Rob.: Chemical Composition, Enantiomeric Distribution, and Antimicrobial, Antioxidant, and Anticholinesterase Activities. Plants 2023, 12, 2796. [Google Scholar] [CrossRef]
  34. Eiroa, J.L.; Triana, J.; Pérez, F.J.; Castillo, Q.A.; Brouard, I.; Quintana, J.; Estévez, F.; León, F. Secondary metabolites from two Hispaniola Ageratina species and their cytotoxic activity. Med. Chem. Res. 2018, 7, 1792–1799. [Google Scholar] [CrossRef]
  35. Bustos-Brito, C.; Vázquez-Heredia, V.J.; Calzada, F.; Yépez-Mulia, L.; Calderón, J.S.; Hernández-Ortega, S.; Esquivel, B.; García-Hernández, N.; Quijano, L. Antidiarrheal thymol derivatives from Ageratina glabrata. Structure and absolute configuration of 10-benzoyloxy-8, 9-epoxy-6-hydroxythymol isobutyrate. Molecules 2016, 21, 1132. [Google Scholar] [CrossRef] [PubMed]
  36. Méndez-Callejas, G.; Rodríguez-Mayusa, J.; Riveros-Quiroga, M.; Mahete-Pinilla, K.; Torrenegra-Guerrero, R. Antiproliferative activity of chloroformic fractions from leaves and inflorescences of Ageratina gracilis. Emir. J. Food Agric. 2017, 29, 59–68. [Google Scholar] [CrossRef]
  37. del Barrio, G.S.I.; García, T.; Roque, A.; Álvarez, A.L.; Calderón, J.S.; Parra, F. Antiviral activity of Ageratina havanensis and major chemical compounds from the most active fraction. Braz. J. Pharmacogn. 2011, 21, 915–920. [Google Scholar] [CrossRef]
  38. García, T.H.; Da Rocha, C.Q.; Dias, M.J.; Pino, L.L.; Del Barrio, G.; Roque, A.; Pérez, C.E.; dos Santos, L.C.; Spengler, I.; Vilegas, W. Comparison of the qualitative chemical composition of extracts from Ageratina havanensis collected in two different phenological stages by FIA-ESI-IT-MSn and UPLC/ESI-MSn: Antiviral activity. Nat. Prod. Commun. 2017, 12, 1934578X1701200110. [Google Scholar] [CrossRef]
  39. García, T.H.; Rocha, C.Q.D.; Delgado-Roche, L.; Rodeiro, I.; Ávila, Y.; Hernández, I.; Cuellar, C.; Lopes, M.T.P.; Vilegas, W.; Auriemma, G.; et al. Influence of the phenological state of in the antioxidant potential and chemical composition of Ageratina havanensis. effects on the p-glycoprotein function. Molecules 2020, 25, 2134. [Google Scholar] [CrossRef]
  40. Torres-Barajas, L.; Rojas-Vera, J.; Morales-Méndez, A.; Rojas-Fermín, L.; Lucena, M.; Buitrago, A. Chemical composition and evaluation of antibacterial activity of essential oils of Ageratina jahnii and Ageratina pichinchensis collected in Mérida, Venezuela. Bol. Latinoam. Caribe Plantas Med. Aromát. 2013, 12, 92–98. [Google Scholar]
  41. Garcia-Sanchez, E.; Ramírez-López, C.B.; Talavera-Alemán, A.; Leon-Hernandez, A.; Martinez-Munoz, R.E.; Martínez-Pacheco, M.M.; Gómez-Hurtado, M.A.; Cerda-García-Rojas, C.M.; Joseph-Nathan, P.; del Río, R.E. Absolute configuration of (13 R)-and (13 S)-labdane diterpenes coexisting in Ageratina jocotepecana. J. Nat. Prod. 2014, 77, 1005–1012. [Google Scholar] [CrossRef]
  42. Caballero-Gallardo, K.; Quintero-Rincón, P.; Stashenko, E.E.; Olivero-Verbel, J. Chemical composition and in vitro bioactivities of hydroalcoholic extracts from Turnera pumilea, Hyptis lantanifolia, and Ageratina popayanensis cultivated in Colombia. Sci. Rep. 2025, 15, 31189. [Google Scholar] [CrossRef]
  43. Romero-Cerecero, O.; Islas-Garduño, A.L.; Zamilpa, A.; Tortoriello, J. Effectiveness of Ageratina pichinchensis Extract in Patients with Vulvovaginal Candidiasis. A Randomized, Double-Blind, and Controlled Pilot Study. Phytother. Res. 2017, 31, 885–890. [Google Scholar] [CrossRef]
  44. Romero-Cerecero, O.; Zamilpa-Álvarez, A.; Ramos-Mora, A.; Alonso-Cortés, D.; Jiménez-Ferrer, J.E.; Huerta-Reyes, M.E.; Tortoriello, J. Effect on the wound healing process and in vitro cell proliferation by the medicinal Mexican plant Ageratina pichinchensis. Planta Medica 2011, 77, 979–983. [Google Scholar] [CrossRef]
  45. Solís-Quispe, L.; Pino, J.A.; Falco, A.S.; Tomaylla-Cruz, C.; Quispe-Tonccochi, E.G.; Solís-Quispe, J.A.; Aragón-Alencastre, L.J.; Solís-Quispe, A. Chemical composition and antibacterial activities of essential oil from Ageratina pentlandiana (DC.) RM King, H. Rob. leaves grown in the Peruvian Andes. J. Essent. Oil Res. 2019, 31, 409–413. [Google Scholar] [CrossRef]
  46. Carrillo-Hormaza, L.; Mora, C.; Alvarez, R.; Alzate, F.; Osorio, E. Chemical composition and antibacterial activity against Enterobacter cloacae of essential oils from Asteraceae species growing in the Páramos of Colombia. Ind. Crops Prod. 2015, 77, 108–115. [Google Scholar] [CrossRef]
  47. Castañeda, J.S.; Suta-Velásquez, M.; Mateus, J.; Pardo-Rodriguez, D.; Puerta, C.J.; Cuéllar, A.; Robles, J.; Cuervo, C. Preliminary chemical characterization of ethanolic extracts from Colombian plants with promising anti-Trypanosoma cruzi activity. Exp. Parasitol. 2021, 223, 108079. [Google Scholar] [CrossRef] [PubMed]
  48. Alonso-Castro, A.J.; Villarreal, M.L.; Salazar-Olivo, L.A.; Gomez-Sanchez, M.; Dominguez, F.; Garcia-Carranca, A. Mexican medicinal plants used for cancer treatment: Pharmacological, phytochemical and ethnobotanical studies. J. Ethnopharmacol. 2011, 133, 945–972. [Google Scholar] [CrossRef]
  49. Suffness, M.; Pezzuto, J.M. Assays related to cancer drug discovery. In Methods in Plant Biochemistry: Assays for Bioactivity; Academic Press: Cambridge, MA, USA, 1990; Volume 6, pp. 71–133. [Google Scholar]
  50. Canga, I.; Vita, P.; Oliveira, A.I.; Castro, M.Á.; Pinho, C. In vitro cytotoxic activity of African plants: A review. Molecules 2022, 27, 4989. [Google Scholar] [CrossRef]
  51. Merrouni, I.A.; Elachouri, M. Anticancer medicinal plants used by Moroccan people: Ethnobotanical, preclinical, phytochemical and clinical evidence. J. Ethnopharmacol. 2021, 266, 113435. [Google Scholar] [CrossRef]
  52. Ibrahim, R.S.; Seif El-Din, A.A.; Abu-Serie, M.; Abd El Rahman, N.M.; El-Demellawy, M.; Metwally, A.M. Investigation of In-Vitro Cytotoxic and Potential Anticancer Activities of Flavonoidal Aglycones from Egyptian Propolis. Rec. Pharm. Biomed. Sci. 2017, 2, 13–20. [Google Scholar] [CrossRef]
  53. Li, J.; Li, X.; Liu, H. Sesquiterpene lactones and cancer: New insight into antitumor and anti-inflammatory effects of parthenolide-derived Dimethylaminomicheliolide and Micheliolide. Front. Pharmacol. 2025, 16, 1551115. [Google Scholar] [CrossRef]
  54. Hsu, C.-Y.; Rajabi, S.; Hamzeloo-Moghadam, M.; Kumar, A.; Maresca, M.; Ghildiyal, P. Sesquiterpene lactones as emerging biomolecules to cease cancer by targeting apoptosis. Front. Pharmacol. 2024, 15, 1371002. [Google Scholar] [CrossRef]
  55. Monasterio, A.; Urdaci, M.C.; Pinchuk, I.V.; Lopez-Moratalla, N.; Martinez-Irujo, J.J. Flavonoids induce apoptosis in human leukemia U937 cells through caspase-and caspase-calpain-dependent pathways. Nutr. Cancer 2004, 50, 90–100. [Google Scholar] [CrossRef]
  56. Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as anticancer agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, Y.; Li, J.; Xia, L. Plant-derived natural products and combination therapy in liver cancer. Front. Oncol. 2023, 13, 1116532. [Google Scholar] [CrossRef] [PubMed]
  58. Rajabi, S.; Maresca, M.; Yumashev, A.V.; Choopani, R.; Hajimehdipoor, H. The most competent plant-derived natural products for targeting apoptosis in cancer therapy. Biomolecules 2021, 11, 534. [Google Scholar] [CrossRef] [PubMed]
  59. Montané, X.; Kowalczyk, O.; Reig-Vano, B.; Bajek, A.; Roszkowski, K.; Tomczyk, R.; Pawliszak, W.; Giamberini, M.; Mocek-Płóciniak, A.; Tylkowski, B. Current perspectives of the applications of polyphenols and flavonoids in cancer therapy. Molecules 2020, 25, 3342. [Google Scholar] [CrossRef]
  60. Shamas-Din, A.; Kale, J.; Leber, B.; Andrews, D.W. Mechanisms of action of Bcl-2 family proteins. Cold Spring Harb. Perspect. Biol. 2013, 5, a008714. [Google Scholar] [CrossRef]
  61. Teng, C.; Chen, J.W.; Shen, L.S.; Chen, S.; Chen, G.Q. Research advances in natural sesquiterpene lactones: Overcoming cancer drug resistance through modulation of key signalling pathways. Cancer Drug Resist. 2025, 8, 13. [Google Scholar] [CrossRef]
  62. Cao, F.; Chu, C.; Qin, J.J.; Guan, X. Research progress on antitumor mechanisms and molecular targets of Inula sesquiterpene lactones. Chin. Med. 2023, 18, 164. [Google Scholar] [CrossRef]
  63. Chauhan, A.; Islam, A.U.; Prakash, H.; Singh, S. Phytochemicals targeting NF-κB signalling: Potential anti-cancer interventions. J. Pharm. Anal. 2022, 12, 394–405. [Google Scholar] [CrossRef]
  64. Ayoub, I.M.; El-Shazly, M.; Lu, M.C.; Singab, A.N.B. Antimicrobial and cytotoxic activities of the crude extracts of Dietes bicolor leaves, flowers and rhizomes. S. Afr. J. Bot. 2014, 95, 97–101. [Google Scholar] [CrossRef]
  65. Hussain, S.; Liufang, H.; Shah, S.M.; Ali, F.; Khan, S.A.; Shah, F.A.; Li, J.B.; Li, S. Cytotoxic effects of extracts and isolated compounds from Ifloga spicata (forssk.) sch. bip against HepG-2 cancer cell line: Supported by ADMET analysis and molecular docking. Front. Pharmacol. 2022, 13, 986456. [Google Scholar] [CrossRef]
  66. Serwecińska, L. Antimicrobials and antibiotic-resistant bacteria: A risk to the environment and to public health. Water 2020, 12, 3313. [Google Scholar] [CrossRef]
  67. Paulos, B.; Bisrat, D.; Yeshak, M.Y.; Asres, K. Natural Products with Potent Antimycobacterial Activity (2000–2024): A Review. Molecules 2025, 30, 3708. [Google Scholar] [CrossRef] [PubMed]
  68. Gonzalez-Pastor, R.; Carrera-Pacheco, S.E.; Zúñiga-Miranda, J.; Rodríguez-Pólit, C.; Mayorga-Ramos, A.; Guamán, L.P.; Barba-Ostria, C. Current Landscape of Methods to Evaluate Antimicrobial Activity of Natural Extracts. Molecules 2023, 28, 1068. [Google Scholar] [CrossRef] [PubMed]
  69. Kowalska-Krochmal, B.; Dudek-Wicher, R. The minimum inhibitory concentration of antibiotics: Methods, interpretation, clinical relevance. Pathogens 2021, 10, 165. [Google Scholar] [CrossRef]
  70. Fei, L.; YanChun, H.; Lei, W.; Hui, T.; Quan, M.; Biao, L.; YaJun, H.; JunLiang, D.; YaHui, W. Antitumor activity in vitro by 9-oxo-10, 11-dehydroageraphorone extracted from Eupatorium adenophorum. Asian J. Chem. 2014, 26, 7321–7323. [Google Scholar]
  71. Heena; Kaushal, S.; Kaur, V.; Panwar, H.; Sharma, P.; Jangra, R. Isolation of quinic acid from dropped Citrus reticulata Blanco fruits: Its derivatization, antibacterial potential, docking studies, and ADMET profiling. Front. Chem. 2024, 12, 1372560. [Google Scholar] [CrossRef]
  72. Lu, L.; Zhao, Y.; Yi, G.; Li, M.; Liao, L.; Yang, C.; Cho, C.; Zhang, B.; Zhu, J.; Zou, K.; et al. Quinic acid: A potential antibiofilm agent against clinical resistant Pseudomonas aeruginosa. Chin. Med. 2021, 16, 72. [Google Scholar] [CrossRef]
  73. Ercan, L.; Doğru, M. Antioxidant and antimicrobial capacity of quinic acid. Bitlis Eren Üniv. Fen Bilim. Derg. 2022, 11, 1018–1025. [Google Scholar] [CrossRef]
  74. Peter, S.; Sotondoshe, N.; Aderibigbe, B.A. Carvacrol and Thymol Hybrids: Potential Anticancer and Antibacterial Therapeutics. Molecules 2024, 29, 2277. [Google Scholar] [CrossRef]
  75. Khan, S.T.; Khan, M.; Ahmad, J.; Wahab, R.; Abd-Elkader, O.H.; Musarrat, J.; Al-Kedhairy, A.A. Thymol and carvacrol induce autolysis, stress, growth inhibition and reduce the biofilm formation by Streptococcus mutans. AMB Express 2017, 7, 49. [Google Scholar] [CrossRef]
  76. Quintanilla-Licea, R.; Mata-Cárdenas, B.D.; Vargas-Villarreal, J.; Bazaldúa-Rodríguez, A.F.; Ángeles-Hernández, I.K.; Garza-González, J.N.; Hernández-García, M.E. Antiprotozoal activity against Entamoeba histolytica of plants used in northeast Mexican traditional medicine. Bioactive compounds from Lippia graveolens and Ruta chalepensis. Molecules 2014, 19, 21044–21065. [Google Scholar] [CrossRef] [PubMed]
  77. Ranasinghe, S.; Armson, A.; Lymbery, A.J.; Zahedi, A.; Ash, A. Medicinal plants as a source of antiparasitics: An overview of experimental studies. Pathog. Glob. Health 2023, 117, 535–553. [Google Scholar] [CrossRef] [PubMed]
  78. Ullah, F.; Ayaz, M.; Sadiq, A.; Ullah, F.; Hussain, I.; Shahid, M.; Yessimbekov, Z.; Adhikari-Devkota, A.; Devkota, H.P. Potential role of plant extracts and phytochemicals against foodborne pathogens. Appl. Sci. 2020, 10, 4597. [Google Scholar] [CrossRef]
  79. Noites, A.; Borges, I.; Araújo, B.; da Silva, J.C.E.; de Oliveira, N.M.; Machado, J.; Pinto, E. Antimicrobial activity of some medicinal herbs to the treatment of cutaneous and mucocutaneous infections: Preliminary research. Microorganisms 2023, 11, 272. [Google Scholar] [CrossRef]
  80. El-Saadony, M.T.; Saad, A.M.; Mohammed, D.M.; Korma, S.A.; Alshahrani, M.Y.; Ahmed, A.E.; Ibrahim, E.H.; Salem, H.M.; Alkafaas, S.S.; Saif, A.M.; et al. Medicinal plants: Bioactive compounds, biological activities, combating multidrug-resistant microorganisms, and human health benefits—A comprehensive review. Front. Immunol. 2025, 16, 1491777. [Google Scholar] [CrossRef]
  81. Vaou, N.; Stavropoulou, E.; Voidarou, C.; Tsigalou, C.; Bezirtzoglou, E. Towards Advances in Medicinal Plant Antimicrobial Activity: A Review Study on Challenges and Future Perspectives. Microorganisms 2021, 9, 2041. [Google Scholar] [CrossRef]
  82. Perez-Flores, J.G.; Garcia-Curiel, L.; Perez-Escalante, E.; Contreras-Lopez, E.; Aguilar-Lira, G.Y.; Angel-Jijon, C.; Gonzalez-Olivares, L.G.; Baena-Santillan, E.S.; Ocampo-Salinas, I.O.; Guerrero-Solano, J.A.; et al. Plant Antimicrobial Compounds and Their Mechanisms of Action on Spoilage and Pathogenic Bacteria: A Bibliometric Study and Literature Review. Appl. Sci. 2025, 15, 3516. [Google Scholar] [CrossRef]
  83. Alonso-Castro, A.J.; Domínguez, F.; Maldonado-Miranda, J.J.; Castillo-Pérez, L.J.; Carranza-Álvarez, C.; Solano, E.; Isiordia-Espinoza, M.A.; Juárez-Vázquez, M.d.C.; Zapata-Morales, J.R.; Argueta-Fuertes, M.A.; et al. Use of medicinal plants by health professionals in Mexico. J. Ethnopharmacol. 2017, 198, 81–86. [Google Scholar] [CrossRef]
  84. Theodoridis, S.; Drakou, E.G.; Hickler, T.; Thines, M.; Nogues-Bravo, D. Evaluating natural medicinal resources and their exposure to global change. Lancet Planet. Health 2023, 7, e155–e163. [Google Scholar] [CrossRef]
  85. Kangra, K.; Kakkar, S.; Mittal, V.; Kumar, V.; Aggarwal, N.; Chopra, H.; Malik, T.; Garg, V. Incredible use of plant-derived bioactives as anticancer agents. RSC Adv. 2025, 15, 1721–1746. [Google Scholar] [CrossRef]
  86. Porras, G.; Chassagne, F.; Lyles, J.T.; Marquez, L.; Dettweiler, M.; Salam, A.M.; Samarakoon, T.; Shabih, S.; Farrokhi, D.R.; Quave, C.L. Ethnobotany and the role of plant natural products in antibiotic drug discovery. Chem. Rev. 2020, 121, 3495–3560. [Google Scholar] [CrossRef] [PubMed]
  87. Pradeep Prabhu, P.; Mohanty, B.; Lobo, C.L.; Balusamy, S.R.; Shetty, A.; Perumalsamy, H.; Mahadev, M.; Mijakovic, I.; Dubey, A.; Singh, P. Harnessing the nutriceutics in early-stage breast cancer: Mechanisms, combinational therapy, and drug delivery. J. Nanobiotechnol. 2024, 22, 574. [Google Scholar] [CrossRef] [PubMed]
  88. Singh, K.; Gupta, J.K.; Chanchal, D.K.; Shinde, M.G.; Kumar, S.; Jain, D.; Almarhoon, Z.M.; Alshahrani, A.M.; Calina, D.; Sharifi-Rad, J.; et al. Natural products as drug leads: Exploring their potential in drug discovery and development. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2025, 398, 4673–4687. [Google Scholar] [CrossRef] [PubMed]
  89. Ali, S.; Amin, A.; Akhtar, M.S.; Zaman, W. Micro-and Nanoengineered Devices for Rapid Chemotaxonomic Profiling of Medicinal Plants. Nanomaterials 2025, 15, 899. [Google Scholar] [CrossRef]
  90. Happy, K.; Ban, Y.; Mudondo, J.; Haniffadli, A.; Gang, R.; Choi, K.O.; Rahmat, E.; Okello, D.; Komakech, R.; Kang, Y. SMART-HERBALOMICS: An Innovative Multi-Omics Approach to Studying Medicinal Plants Grown in Controlled Systems such as Phytotrons. Phytomedicine 2025, 148, 157303. [Google Scholar] [CrossRef]
  91. Ydyrys, A.; Nurtayeva, M.; Yerkebayeva, M. Advancing Medicinal Plant Research with Artificial Intelligence: Key Applications and Benefits. Nat. Resour. Hum. Health 2025, 5, 740–775. [Google Scholar] [CrossRef]
  92. Suriyaamporn, P.; Pamornpathomkul, B.; Patrojanasophon, P.; Ngawhirunpat, T.; Rojanarata, T.; Opanasopit, P. The artificial intelligence-powered new era in pharmaceutical research and development: A review. AAPS PharmSciTech 2024, 25, 188. [Google Scholar] [CrossRef]
  93. Naveed, M.; Abid, A.; Aziz, T.; Saleem, A.; Hanif, N.; Ali, I.; Alasmari, A.F. Comparative toxicity assessment of fisetin-aided artificial intelligence-assisted drug design targeting epibulbar dermoid through phytochemicals. Open Chem. 2024, 22, 20230197. [Google Scholar] [CrossRef]
Figure 1. Aerial parts characteristics of Ageratina Spach.
Figure 1. Aerial parts characteristics of Ageratina Spach.
Molecules 30 04656 g001
Figure 2. PRISMA meta-analysis flow diagram for systematic review of Ageratina.
Figure 2. PRISMA meta-analysis flow diagram for systematic review of Ageratina.
Molecules 30 04656 g002
Figure 3. Structure of compounds isolated from Ageratina Spach.
Figure 3. Structure of compounds isolated from Ageratina Spach.
Molecules 30 04656 g003
Figure 4. Structure of compounds isolated from Ageratina Spach evaluated as antimicrobials.
Figure 4. Structure of compounds isolated from Ageratina Spach evaluated as antimicrobials.
Molecules 30 04656 g004aMolecules 30 04656 g004b
Table 1. Species of Ageratina analysed in the review, from 2010 to 2025.
Table 1. Species of Ageratina analysed in the review, from 2010 to 2025.
SpeciesCollection PlacePart of the PlantExtract SolventRefs.
A. adenophoraThailandLEthanol[19]
NepalAPMethanol[20]
IndiaLHydroalcoholic[21]
IndiaLMethanol[22]
PortugalLAqueous[23]
ChinaAPEthanol[24]
ChinaRMethanol[25]
ChinaREthanol[26,27,28]
A. cylindricaMéxicoLDCM[29]
MéxicoLAqueous[30]
MéxicoLPetrol[31]
A. deltoideaMexicoAPHexane[32]
A.dendroidesEcuadorL&FEO-Hdest[33]
A. dictyoneuraDominican RepublicAPEthanol[34]
A. glabrataMexicoLDCM[35]
A.gracilisColombiaIEthanol[36]
ColombiaIPetrol
ColombiaLEthanol
ColombiaLPetrol
A.havanensisCubaLEthanol[37]
CubaLEtOAc
CubaLn-butanol
CubaSEthanol
CubaSEtOAc
CubaSn-butanol
CubaSEthanol
CubaL&FEtOAc[38]
CubaL&Fn-butanol
CubaL (FS)Ethanol[39]
CubaL (FS)EtOAc
CubaL (FS)n-butanol
CubaS (FS)Ethanol
CubaS (FS)EtOAc
CubaS (FS)n-butanol
CubaF (FS)Ethanol
CubaF (FS)EtOAc
CubaF (FS)n-butanol
A. illitaDominican RepublicAPEthanol[34]
A.janniVenezuelaLEO-Hdest[40]
A. jocotepecanaMexicoFHexane[41]
MexicoL
A.popayanensisColombiaAPHydroalcoholic[42]
A.pichinchensisMexicoAP(7:3) H-EtOAc[43]
MexicoAPAqueous[44]
MexicoAP(7:3) H-EtOAc
VenezuelaLEO-Hdest[40]
A.pentlandianaPeruLEO-Hdest[45]
A. tinifoliaColombiaAPEO-Hdest[46]
A.vacciniaefoliaColombiaLEthanol[47]
L = leaves, AP = Aerial parts; R = Root(s), S = Steams; F = Flowers; I = inflorescence; L&F = Leaves and flowers, FS = Flowering stage, DCM = Dichloromethane, H-EtOAc = Hexane-Ethyl acetate, EtOAc = Ethyl acetate, EO-Hdest = Essential oil by Hydro distillation.
Table 2. The cytotoxic activity of the extracts obtained from Ageratina Spach.
Table 2. The cytotoxic activity of the extracts obtained from Ageratina Spach.
SpeciePP/ESCCLResult
IC50 (μg/mL)
Ref.
A. adenophoraL/HydroalcoholicHCT-11665.65 ± 2.1[21]
L/MethanolA54950.08 ± 0.14[22]
L/AqueousHeLa950 ± 0.07[23]
Caco-2289 ± 0.12
MCF-7302 ± 0.16
A. havanensis (FS)L/Ethanol4T1381.6 ± 7.5[39]
L/EtOAc252.5 ± 10.1
L/n-butanol302.0 ± 8.0
S/Ethanol228.2 ± 8.7
F/Ethanol263.5 ± 8.2
F/EtOAc259.5 ± 10.6
F/n-butanol315.5 ± 9.9
A. havanensis (VSt)L/Ethanol4T1392.8 ± 6.7
L/EtOAc313.0 ± 12.1
L/n-butanol496.5 ± 6.7
S/Ethanol355.7 ± 7.6
A. gracilisI/EthanolSiHa41.14 ± 1.02[36]
HT2962.33 ± 1.24
A54974.56 ± 0.95
MDA-MB-23162.81 ± 0.37
PC-377.35 ± 1.06
I/PetrolSiHa21.19 ± 1.25
HT2912.67 ± 1.13
A54938.50 ± 1.18
MDA-MB-23126.94 ± 1.05
PC-352.65 ± 0.64
L/EthanolSiHa71.98 ± 1.53
HT2953.05 ± 0.82
A549116.96 ± 1.04
MDA-MB-23158.44 ± 0.78
PC-372.46 ± 0.47
L/PetrolSiHa12.91 ± 0.92
HT2911.20 ± 1.20
A54934.80 ± 1.15
MDA-MB-23114.72 ± 0.69
PC-329.85 ± 1.27
A. pichinchensisL&S/AqueousKB≥20[44]
HCT-15≥20
UISO≥20
OVCAR≥20
L&S/(7:3) H-EtOAcKB≥20
HCT-15≥20
UISO≥20
OVCAR≥20
A. popayanensisAP/HydroalcoholicCalu-1444[42]
HepG2387
A. havanensisL/EthanolVeroCC50 (µg/mL)
2834 ± 448
[37]
L/EtOAcVero1670 ± 0.2
L/n-butanolVero404.9 ± 43.5
S/EthanolVero5685 ± 117
S/EtOAcVero2270 ± 99.9
S/n-butanolVero457.4 ± 28.1
L&F/EtOAcVero1242.8 ± 37.2[38]
PP/ES = Part of the plant/extraction solvent, L = leaves, AP = Aerial parts; S = Steams; F = Flowers; L&F = Leaves and flowers; I = inflorescence; L&S = Leaves and Steams, FS = Flowering stage, VSt = Vegetative stage, H-EtOAc = Hexane-Ethyl acetate, EtOAc = Ethyl acetate, CCL = Cancer cell line; IC50: half-maximal Inhibitory Concentration; CC50: 50% Cytotoxic Concentration; A549: lung cancer; Caco-2: Colon cancer; Calu-1: epidermoid carcinoma of the lung; HCT-116: Adherent cell of colon cancer; HCT-15: colorectal carcinomas; HeLa: cervical carcinoma; HepG2: Hepatocarcinoma; HT29: colorectal adenocarcinoma; KB: Epithelial carcinoma; MCF-7: Breast cancer; MDA-MB-231: Human breast cancer; OVCAR: Ovarian adenocarcinoma; PC-3: Prostatic adenocarcinoma; SiHa: Uterine squamous cell carcinoma; 4T1: breast cancer; UISO: Merkel carcinoma cell; Vero: kidney-derived epithelial cell.
Table 3. The cytotoxic activity of the compounds obtained from Ageratina Spach.
Table 3. The cytotoxic activity of the compounds obtained from Ageratina Spach.
SpecieCompoundsBioModCCLResult
IC50 μM
Ref.
A. adenophoraNew tricyclic cadinene (1)MTSHL-6024.06 ± 2.21[26]
A54911.45 ± 0.69
SMMC-77219.96 ± 1.45
MDA-MB-23116.35 ± 3.32
SW48028.75 ± 3.93
(+)-(5R,7S,9R,10S)-2-oxocadinan-3,6(11)-dien-12,7-olide (2)HL-6035.73 ± 1.51
A5499.85 ± 0.88
SMMC-772113.44 ± 2.32
MDA-MB-23112.72 ± 1.58
SW48026.03 ± 2.91
Cadinene norsesquiterpenoid (3)HL-6042.85 ± 1.35
A54921.82 ± 0.65
SMMC-772110.28 ± 1.67
MDA-MB-23130.42 ± 2.24
SW48023.65 ± 1.49
9-oxo-10,11-dehydro-ageraphorone (Euptox A) (4)HeLa(mg/mL)
0.55 ± 0.05
[23]
Caco-21.43 ± 0.08
MCF-71.63 ± 0.08
7,9-diisobutyryloxy-8-ethoxythymol (5)MTTA549IC50 (μM)
>100
[27]
HeLa>100
HepG2>100
7-acetoxy-8-methoxy-9-isobutyryloxythymol (6)A549>100
HeLa>100
HepG2>100
7,9-diisobutyryloxy-8-methoxythymol (7)A549>100
HeLa>100
HepG2>100
9-oxoageraphorone (8)A549>100
HeLa>100
HepG2>100
(−)-isochaminic acid (9)A54932.37 ± 3.75
HeLa25.64 ± 2.34
HepG241.87 ± 6.53
(1α,6α)-10-hydroxy-3-carene-2-one (10)A54930.65 ± 3.87
HeLa18.36 ± 1.72
HepG239.44 ± 3.61
A. dictyoneuraQuercetin 3,7-dimethylether (11)U-9377.0 ± 0.5[34]
A. illita(8R)-8-hydroxy-β-cyclocostunolide (12)6.0 ± 0.7
BioMod = Biological model, CCL = Cancer cell line, MTT: 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide; MTS: 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-H-tetrazolium; IC50: half-maximal Inhibitory Concentration; HepG2: Hepatocarcinoma HL-60: promyelocytic leukaemia; SMMC-7721: human hepatocellular carcinoma; SW480: Human colon carcinoma; U-937: Histiocytic lymphoma.
Table 4. The antimicrobial activity of the extracts derived from Ageratina Spach.
Table 4. The antimicrobial activity of the extracts derived from Ageratina Spach.
SpeciePP/ESMethodMicroorganismResultRef.
A. adenophoraAP/MethanolμdMRSAMIC (mg/mL)
125
[20]
S. aureus25
AP/EOμdE. coliMIC (μg/mL)
>4000
[33]
P. aeruginosa>4000
E. faecium>4000
E. faecalis>4000
S. aureus>4000
AP/EOμdS. enterica subsp. enterica serovar Thypimurium>4000
A. janniL/EODf discS. aureusMIC (mg/mL)
49.5
[40]
E. faecalis49.5
A.pentlandianaL/EOμdS. aureusMIC (μL/mL)
11.9 + 0.1
[45]
B. subtilis22.7 ± 0.3
E. coli57.7 ± 0.1
S. thyphimurium41.6 ± 0.1
MBCS. aureus11.9 + 0.1
B. subtilis22.7 ± 0.1
E. coli64.8 ± 0.3
S. thyphimurium50.0 ± 0.2
A. pichinchensisL/EODf discS. aureusMIC (mg/mL)
104
[40]
E. faecalis104
A.tinifoliaAP/EOμdE. cloacae
clinical sample
>5[46]
E. cloacae
ATCC
>5
A. havanensisL/EthanolSCAHSV-1EC50 (μg/mL)
809.9 ± 59.6
[37]
HSV-21050 ± 42.9
L/EtOAcHSV-1311.6 ± 10.1
HSV-2>450
L/n-butanolHSV-1>225
HSV-2128.1 ± 22.8
S/EthanolHSV-1>450
HSV-22614 ± 158
S/EtOAcHSV-1>450
HSV-2>450
S/n-butanolHSV-1240.9 ± 5.7
HSV-2145.4 ± 22.1
L&F/EtOAcVIICEHSV-1463.4 ± 12.5[38]
HSV-2>200
A. adenophoraL/EthanolDfT. mentagrophytesMIC (mg/mL)
0.04
[19]
T. rubrum<0.0025
A. dendroidesAP/EOμdA. nigerMIC (μg/mL)
>4000
[33]
AP/EOμdC. albicans>4000
A. pichinchensisAP/(7:3) H-EtOAcVSC. albicansDecrease %
81.2
[43]
A. vacciniaefoliaL/EthanolMTTT. cruziEC50 (μg/mL)
256
[47]
L = leaves; AP = Aerial parts; S = Steams; H-EtOAc = Hexane-Ethyl acetate; EtOAc = Ethyl acetate; EO = Essential Oil, μd = microdilution; Df = diffusion; Df disc = diffusion disc; SCA = Suspension cell assay; VIICE = Virus inhibition induced cytopathic effect; VS = Vaginal suppositories (in vivo assay, patients with vulvovaginitis associated with C. albicans applied vaginally one suppository diary for 6 days); MIC: Minimal inhibitory concentration; MBC: Minimal bactericide concentration; ATCC: American Type Culture Collection; EC50: 50% Effective Concentration; Microorganisms: Aspergillus niger, Bacillus subtilis, Candida albicans, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2), Pseudomonas aeruginosa, Salmonella enterica subsp. enterica serovar, Salmonella thyphimurium, Staphylococcus aureus, Methicillin-resistant Staphylococcus aureus (MRSA), Tinea pedis, Trichophyton mentagrophytes, Trichophyton rubrum, Trypanosome cruzi.
Table 5. The antimicrobial activity of the compounds obtained from Ageratina Spach.
Table 5. The antimicrobial activity of the compounds obtained from Ageratina Spach.
SpecieCompoundsMethodMicroorganism AssayResult
MIC (µg/mL)
Ref.
A. adenophora2α-methoxyl-3β-methyl-6-(acetyl-O-methyl)-2,3-dihydrobenzofuran (13)μdS. aureus25[28]
B. cereus50
B. subtilis25
E. coli>100
1,6-dihydroxy-1-isopropyl-4,7-dimethyl-3,4-dihydronaphthalen-2(1H)-one (14)S. aureus>100
B. cereus>100
B. subtilis>100
E. coli>100
Eupatorenone (15)S. aureus12.5
B. cereus25
B. subtilis25
E. coli>100
3-hydroxymuurola-4,7(11)-dien-8-one (16)S. aureus25
B. cereus12.5
B. subtilis25
E. coli>100
9-oxoageraphorone (8)S. aureus>100
B. cereus>100
B. subtilis>100
E. coli>100
(4R,5S)-4-hydroxy-5-isopropyl-2-methyl-2-cyclohexehone (17)S. aureus>100
B. cereus>100
B. subtilis>100
E. coli>100
7,9-diisobutyryloxy-8-ethoxythymol (5)μdS. aureus>200[27]
B. thuringiensis>200
B. subtilis125
E. coli>200
S. dysenteriae>200
7-acetoxy-8-methoxy-9-isobutyryloxythymol (6)S. aureus125
B. thuringiensis62.5
B. subtilis62.5
E. coli>200
S. dysenteriae>200
7,9-di-isobutyryloxy-8-methoxythymol (7)S. aureus>200
B. thuringiensis125
B. subtilis125
E. coli>200
E. dysenteriae>200
9-oxoageraphorone (8)S. aureus>200
B. thuringiensis>200
B. subtilis>200
E. coli>200
S. dysenteriae>200
(−)-isochaminic acid (9)S. aureus31.3
B. thuringiensis31.3
B. subtilis15.6
E. coli62.5
S. dysenteriae62.5
(1α,6α)-10-hydroxy-3-carene-2-one (10)S. aureus15.6
B. thuringiensis31.3
B. subtilis15.6
E. coli62.5
S. dysenteriae62.5
5-O-trans-o-coumaroylquinic acid methyl ester (18)μdS. aureusMIC (µM)
88.8
[24]
B. thuringiensis88.8
E. coli88.8
S. enterica88.8
S. dysenteriae177.6
Chlorogenic acid methyl ester (19)S. aureus84.8
B. thuringiensis84.8
E. coli84.8
S. enterica84.8
S. dysenteriae169.8
Macranthoin F (20)S. aureus29.4
B. thuringiensis59.0
E. coli59.0
S. enterica14.7
S. dysenteriae117.9
Macranthoin G (21)S. aureus59.0
B. thuringiensis59.0
E. coli59.0
S. enterica7.4
S. dysenteriae117.9
A. deltoideaGrandiflorenic acid (22)μdS. aureusMIC (µg/mL)
31
[32]
Kaurenoic acid (23)S. aureus31
Deltoidin A (24)E. coli16
8β-angeloyloxyelemacronquistianthus acid (25)S. aureus125
E. coli125
A. glabrata(8S)-10-Benzoyloxy-8,9-epoxy-6-hydroxythymol isobutyrate (26)MTT/PMSE. histolyticaIC50 (µM)
1.6
[35]
G. lamblia36.9
10-Benzoyloxy-8,9-epoxy-6-acetyloxythymol isobutyrate (27)E. histolytica0.84
G. lamblia24.2
10-Benzoyloxy-8,9-epoxy-6-methoxythymol isobutyrate (28)E. histolytica169.6
G. lamblia191.2
10-Benzoyloxy-8,9-epoxythymol isobutyrate (29)E. histolytica25.9
G. lamblia48.3
10-Benzoyloxy-8,9-dehydro-6-hydroxythymol isobutyrate (30)E. histolytica61.2
G. lamblia68.0
10-Benzoyloxy-6,8-dihydroxy-9-isobutyryloxythymol (31)E. histolytica45.6
G. lamblia60.7
Pectolinaringenin (32)E. histolytica43.6
G. lamblia68.7
(8S)-8,9-epoxy-6-hydroxy-10-benzoyloxy-7-oxothymol isobutyrate (33)E. histolytica184.9
G. lamblia167.4
A. jocotepecana(−)-(5S,9S,10S,13S)-labd-7-en-15-oic acid (34)μdS. aureusMIC (mg/mL)
0.78
[41]
B. subtilis0.15
(−)-(5S,9S,10S,13Z)-labda-7,13-dien-15-oic acid (35)S. aureus1.00
B. subtilis10.00
(+)-(5S,8R,9R,10S,13R)-8-hydroxylabdan-15-oic acid (36)S. aureus2.34
B. subtilis1.56
A. adenophoraEncecalin (37)Df disc
(50 μg/disc)
F. oxysporum f. sp. niveumDIZ (mm)
10.00 ± 0.15
[25]
7-hydroxy-dehydrotremetone (38)C. gloeosporioides17.28 ± 0.46
C. musae17.24 ± 0.52
R. solani16.40 ± 0.81
F. oxysporum f. sp. niveum13.90 ± 1.05
A. alternata14.36 ± 0.68
A. cylindricaCylindrinol B (39)NAE. histolyticaIC50 (µM)
287.5
[31]
G. lamblia226.2
Cylindrinol C (40)E. histolytica237.0
G. lamblia251.9
Cylindrinol D (41)E. histolytica86.9
G. lamblia134.0
Cylindrinol E (42)E. histolytica210.2
G. lamblia164.5
Cylindrinol F (43)E. histolytica213.2
G. lamblia151.1
ent-15β-(β-L-fucosyloxy)kaur-16-en-19-oic acid β-Dglucopyranosyl ester (44)E. histolytica43.3[30]
G. lamblia41.9
ent-15β-(4-acetoxy-β-L-fucosyloxy) kaur-16-en-19-oic acid β-D-glucopyranosyl ester (45)E. histolytica49.5
G. lamblia69.5
ent-15β-(3-acetoxy-β-L-fucosyloxy) kaur-16-en-19-oic acid β-D-glucopyranosyl ester (46)E. histolytica52.7
G. lamblia48.9
ent-15β-(β-L-fucosyloxy)kaur-16-en-19-oic acid (47)E. histolytica73.5
G. lamblia98.5
(8S)-8,9-epoxy-6-hydroxy-10-benzoyloxy-7-oxothymol isobutyrate (33)E. histolytica184.9[29]
G. lamblia167.4
MIC = Minimal inhibitory concentration; μd = microdilution; Df disc = Diffusion disc; DIZ = Diameter of Inhibitory Zone; MTT/PMS = Tetrazolium salt/phenazine methosulfate; IC50 = Fifth inhibitory concentration; NA = Nitazoxanide assay; Microorganisms: Alternaria alternata; Bacillus cereus; Bacillus subtilis; Bacillus thuringiensis; Colletotrichum gloeosporioides; Colletotrichum musae; Entamoeba histolytica; Escherichia coli; Fusarium oxysporum f. sp. niveum; Giardia lamblia; Salmonella enterica; Shigella dysenteriae; Staphylococcus aureus; Rhizoctonia solani.
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

Rojas-Jiménez, S.; Salinas-Sánchez, D.O.; Rodríguez-López, V.; Salinas-Marín, R.; Avilés-Montes, D.; Sotelo-Leyva, C.; Figueroa-Brito, R.; Bustos Rivera-Bahena, G.; Abarca-Vargas, R.; Arias-Ataide, D.M.; et al. Cytotoxic and Antimicrobial Activity of the Ageratina Genus. Molecules 2025, 30, 4656. https://doi.org/10.3390/molecules30234656

AMA Style

Rojas-Jiménez S, Salinas-Sánchez DO, Rodríguez-López V, Salinas-Marín R, Avilés-Montes D, Sotelo-Leyva C, Figueroa-Brito R, Bustos Rivera-Bahena G, Abarca-Vargas R, Arias-Ataide DM, et al. Cytotoxic and Antimicrobial Activity of the Ageratina Genus. Molecules. 2025; 30(23):4656. https://doi.org/10.3390/molecules30234656

Chicago/Turabian Style

Rojas-Jiménez, Sarai, David Osvaldo Salinas-Sánchez, Verónica Rodríguez-López, Roberta Salinas-Marín, Dante Avilés-Montes, César Sotelo-Leyva, Rodolfo Figueroa-Brito, Genoveva Bustos Rivera-Bahena, Rodolfo Abarca-Vargas, Dulce María Arias-Ataide, and et al. 2025. "Cytotoxic and Antimicrobial Activity of the Ageratina Genus" Molecules 30, no. 23: 4656. https://doi.org/10.3390/molecules30234656

APA Style

Rojas-Jiménez, S., Salinas-Sánchez, D. O., Rodríguez-López, V., Salinas-Marín, R., Avilés-Montes, D., Sotelo-Leyva, C., Figueroa-Brito, R., Bustos Rivera-Bahena, G., Abarca-Vargas, R., Arias-Ataide, D. M., & Valladares-Cisneros, M. G. (2025). Cytotoxic and Antimicrobial Activity of the Ageratina Genus. Molecules, 30(23), 4656. https://doi.org/10.3390/molecules30234656

Article Metrics

Back to TopTop