0% found this document useful (0 votes)
16 views51 pages

Molecules 30 00851 v2

This review focuses on Lantana camara L., a medicinal plant known for its diverse biological activities and phytochemical properties. It highlights the plant's use in traditional medicine, particularly in treating various ailments, and summarizes its bioactive compounds, including triterpenes and flavonoids. The review aims to provide an updated synthesis of existing literature on L. camara, encouraging further research into its medicinal potential.

Uploaded by

ariufjf
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
16 views51 pages

Molecules 30 00851 v2

This review focuses on Lantana camara L., a medicinal plant known for its diverse biological activities and phytochemical properties. It highlights the plant's use in traditional medicine, particularly in treating various ailments, and summarizes its bioactive compounds, including triterpenes and flavonoids. The review aims to provide an updated synthesis of existing literature on L. camara, encouraging further research into its medicinal potential.

Uploaded by

ariufjf
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 51

Review

Ethnobotany, Phytochemistry, and Biological Activity of Extracts


and Non-Volatile Compounds from Lantana camara L. and
Semisynthetic Derivatives—An Updated Review
Jorge Ramírez 1,* , Chabaco Armijos 1 , Nelson Espinosa-Ortega 2 , Leydy Nathaly Castillo 1 and Giovanni Vidari 3

1 Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Técnica Particular de Loja,
Loja 1101608, Ecuador; cparmijos@utpl.edu.ec (C.A.); lncastillo@utpl.edu.ec (L.N.C.)
2 UTPL-Alumni, Barrio San Cayetano Alto, Calle Marcelino Champagnat, Loja 1101608, Ecuador;
njespinosa1@utpl.edu.ec
3 Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil 44001, Iraq;
vidari@unipv.it
* Correspondence: jyramirez@utpl.edu.ec

Abstract: Lantana camara L., commonly known as pigeon berry, is a herbaceous plant of
growing scientific interest due to the high medicinal value. In fact, despite being catego-
rized as an invasive species, it has been used for a long time to treat different diseases
thanks to the many biological activities. Triterpenes, flavonoids, phenylpropanoids, and
iridoid glycosides are the bioactive compounds naturally occurring in L. camara that have
demonstrated anticancer, antifilarial, nematocidal, antibacterial, insecticidal, antileishma-
nial, antifungal, anti-inflammatory, and antioxidant properties. The aim of this review is
to update the information concerning the chemistry and biological activity of L. camara
extracts and their constituents, including semisynthetic derivatives, revising the literature
until June 2024. We believe that the data reported in this review clearly demonstrate the
importance of the plant as a promising source of medicines and will therefore stimulate
further investigations.

Academic Editor: H. P. Vasantha Keywords: Verbenaceae; Lantana camara; ethnobotany; phytochemistry; biological activity
Rupasinghe

Received: 8 January 2025


Revised: 6 February 2025
Accepted: 7 February 2025 1. Introduction
Published: 12 February 2025
According to the latest Angiosperm Phylogeny Group classification (APG IV), the
Citation: Ramírez, J.; Armijos, C.; genus Lantana L. is one of the 32 genera belonging to the family Verbenaceae J. St. Hill., in
Espinosa-Ortega, N.; Castillo, L.N.;
the order Lamiales [1]. This genus comprises 133 species with accepted names according
Vidari, G. Ethnobotany, Phytochemistry,
to the December 2023 WFO classification [2] (latest access: 31 May 2024). However, this
and Biological Activity of Extracts and
Non-Volatile Compounds from
number may change in the future due to the description of new species or the segregation
Lantana camara L. and Semisynthetic into other genera [3]. Moreover, the taxonomic classification of the genus is difficult, since
Derivatives—An Updated Review. species are not stable and widespread hybridization occurs, while morphological characters
Molecules 2025, 30, 851. https:// vary with age.
doi.org/10.3390/molecules30040851
A recent phylogenetic study of the tribe Lantaneae Endl. stated that the genus
Copyright: © 2025 by the authors. Lantana L. is not monophyletic and placed the species Lantana camara L. into the section
Licensee MDPI, Basel, Switzerland. Lantana sect. Lantana together with L. horrida Kunth, L. depressa Small, L. leonardiorum
This article is an open access article
Moldenke, L. sabrida Sol., and L. strigocamara R.W. Sanders [4].
distributed under the terms and
L. camara L., which is the most widespread species of the genus, is an evergreen aro-
conditions of the Creative Commons
Attribution (CC BY) license
matic spiny hairy shrub, usually 0.5–3 m high (Figure 1), bearing flowers of different colors,
(https://creativecommons.org/ from red to pink, white, yellow, orange, and violet. The stems and branches are sometimes
licenses/by/4.0/). armed with prickles or spines; the leaves are opposite, simple with large petioles, and oval

Molecules 2025, 30, 851 https://doi.org/10.3390/molecules30040851


L. camara L., which is the most widespread species of the genus, is an evergreen
aromatic spiny hairy shrub, usually 0.5–3 m high (Figure 1), bearing flowers of different
Molecules 2025, 30, 851 2 of 51
colors, from red to pink, white, yellow, orange, and violet. The stems and branches are
sometimes armed with prickles or spines; the leaves are opposite, simple with large
petioles, and oval blades, which are rugged and hairy and have a bluntly toothed margin.
blades, which are rugged and hairy and have a bluntly toothed margin. The plant is known
The plant ispopular
by different known names,
by different popular
such as pigeonnames, such
berry [5], as red
wild pigeon
sageberry [5], wild angel
[6], cuasquito, red sagelip,
[6], cuasquito, angel lip, flowered sage, black sage, shrub verbena, white
flowered sage, black sage, shrub verbena, white sage, and wild sage [7]. It is native to tropi- sage, and wild
sage
cal and[7].subtropical
It is nativeCentral
to tropical
and and
South subtropical
America, fromCentral anditSouth
where America, from
was introduced where
to other coun-it
was
tries,introduced to other
and it has spread allcountries, and it[8]
over the world has spread
(Figure 2).all
L. over
camaratheis world [8] (Figure
considered 2). L.
an invasive
camara is considered an invasive obnoxious weed of pastures, orchards,
obnoxious weed of pastures, orchards, and forest areas, as well as a cultivated ornamental and forest areas,
as
or well
gardenas ahedge
cultivated
plantornamental
[9–11]. The orpoisonous
garden hedge plant [9–11].
properties of theThe poisonous
plant have been properties
known
of
for a long time, especially to livestock; on the other hand, toxicity to humans fromhand,
the plant have been known for a long time, especially to livestock; on the other fruit
toxicity to humans from fruit ingestion has also been reported.
ingestion has also been reported. Due to the plant cosmopolitan distribution and the innate Due to the plant
cosmopolitan
ability to produce distribution
hybrids,and somethe innate ability
varieties to produce
and subspecies arehybrids,
known [12], someandvarieties
have beenand
subspeciesinare
proposed known [12],
a taxonomic and have
revision been proposed
of Lantana in a taxonomic
L. sect. Lantana revisionbecause
[13]. However, of Lantana L.
of the
sect. Lantana [13]. However, because of the intrinsic taxonomic complexity
intrinsic taxonomic complexity [14], in this review, all of the subspecies and varieties have [14], in this
review,
not beenall of the separately,
treated subspecies but andhave
varieties
beenhave not been treated
incorporated separately,
into a single butL.have
species, camarabeen L.
incorporated
sensu lato. into a single species, L. camara L. sensu lato.

Molecules 2025, 30, x FOR PEER REVIEW 3 of 61

Figure 1. Lantana camara L.: (A): entire plant;


(A) entire plant; (B)
(B):flowers;
flowers;(C)
(C):fruits
fruits(photos
(photos
byby the
the authors).
authors).

Figure2.2.Worldwide
Figure Worldwidedistribution
distributionofofLantana
Lantanacamara
camaraL.L.[11].
[11].

L. camara is one of the most important herbal medicines in the world. For example, it
is well known in the Ayurvedic medicinal system with the Sanskrit names of Chaturangi
and Vanacchedi. Different parts of the plant are used as traditional remedies for the
treatment of various human ailments, such as itches, cuts, ulcers, swellings, bilious fever,
catarrh, asthma and bronchitis, eczema, chicken pox, tetanus, malaria, tumors,
Molecules 2025, 30, 851 3 of 51

L. camara is one of the most important herbal medicines in the world. For example,
it is well known in the Ayurvedic medicinal system with the Sanskrit names of Chatu-
rangi and Vanacchedi. Different parts of the plant are used as traditional remedies for
the treatment of various human ailments, such as itches, cuts, ulcers, swellings, bilious
fever, catarrh, asthma and bronchitis, eczema, chicken pox, tetanus, malaria, tumors, stom-
achache, toothache, headache, scabies, leprosy, rheumatism, and as an antiseptic agent to
treat wounds [4,5,15,16]. The essential oil has shown antibacterial, antifungal, cytotoxic,
and mosquito-repellent effects. L. camara has been found to display a variety of biologi-
cal properties, including antiarthritic, anti-aspergillus, antibacterial, anticancer, cardioac-
tive, anti-fertility, antifilarial, hepatoprotective, anti-hyperglycemic, anti-hyperlipidemic,
anti-inflammatory, insecticidal, antimicrobial, antimutagenic, anxiolytic, nematocidal, an-
tioxidant, anti-proliferative, anti-protozoal, antipyretic, antithrombin, antitumor, antiul-
cerogenic, antiurolithiasis, antiviral, and wound-healing properties. Moreover, the plant
extracts have been reported to inhibit the enzymes acetylcholinesterase, alpha amylase,
carboxylesterase, cyclooxygenase-2, inducible nitric oxide synthase (iNOS), glutathione-S-
transferase (GST), 5-lipoxygenase (5-LOX), protein kinase C, and xanthine oxidase [17,18].
Phytochemical studies conducted by different research groups have led to the isolation of
essential oils, various steroids, terpenoids, saponins, iridoids, flavonoids, phenylethanoids,
naphthoquinones, coumarins, polyphenols and other phenolics, and alkaloids [17–23].
Interestingly, the genus Lantana is free of diterpenoids [17].
The information concerning the phytochemistry and the biological activi-
ties of L. camara L. published until March 2000 has been condensed in previous
reviews [17,18,21,24]. Moreover, recent studies on ecological aspects, chemical constituents,
semisynthetic derivatives [25–27], and the biological and pharmacological activities of
L. camara have been reviewed [17,18,20–23,28–35]. However, these publications, although
dealing with different aspects of the plant, are largely incomplete. Therefore, the purpose
of this review is to update and complete the information about L. camara L. to serve as a
starting point for further investigations of the plant.

2. Research Strategies and Literature Sources


To prepare this review, the literature from 14 March 2000 until 10 June 2024 has been
retrieved from the following databases: Pub-Med (https://pubmed.ncbi.nlm.nih.gov/,
accessed on 10 June 2024), Google Scholar (https://scholar.google.com/, accessed on
9 March 2024), Scopus (https://www.scopus.com/, accessed on 9 March 2024), MDPI (https:
//www.mdpi.com/, accessed on 9 March 2024), NIH (https://www.nih.gov/, accessed
on 9 March 2024), Elsevier (https://www.elsevier.com/, accessed on 9 March 2024), Scielo
(https://scielo.org/es/, accessed on 9 March 2024), and Bio One (https://bioone.org/,
accessed on 9 March 2024). The most relevant papers dedicated to the phytochemistry and
the in vitro and in vivo biological effects of L. camara extracts, isolated chemical compounds,
and semisynthetic derivatives were initially considered. Subsequently, among the more
than 1600 articles published on L. camara, the manager software Mendeley Desktop software
version 1.19.8 was used led us to select and review the research papers mainly dedicated
to the above-mentioned topics. Moreover, all duplicated articles and gray sources were
removed. After this first selection, a total of approximately 200 articles directly related to
the topics of the present review were further reduced to 176 based on the relevance of the
information provided by each of them. The systematic search of databases for relevant
articles published on L. camara to compile this review is shown in Figure 3.
approximately 200 articles directly related to the topics of the present review were further
reduced to 176 based on the relevance of the information provided by each of them. The
Molecules 2025, 30, 851 systematic search of databases for relevant articles published on L. camara to compile this4 of 51
review is shown in Figure 3.

Figure 3. Flowchart for the search process and selection of the studies considered for the review.
Figure 3. Flowchart for the search process and selection of the studies considered for the review.
3. Results
3. Results
3.1. Ethnobotany
In Table 1 we have reported the distribution of L. camara L. in different regions and
3.1. Ethnobotany
countries
In Table 1ofwe
thehave
world, together
reported thewith the local of
distribution vernacular
L. camaranames, the part regions
L. in different of the plant
and used
and the
countries preparation
of the methods
world, together of the
with traditional remedies.names, the part of the plant used
local vernacular
and the preparation
Table methods
1. Distribution of traditional
and traditional uses ofremedies.
Lantana camara L. in various continents and countries of
the world.
Table 1. Distribution and traditional uses of Lantana camara L. in various continents and countries
of the world. Part of Preparation
Region Country Vernacular Name Traditional Uses Ref.
the Plant Method
Congo Vernacular Part of the Preparation
Region Country (Bouenza Name
Lantana
Leaves Decoction
Traditional Antidiarrheal
Uses Ref. [36]
Plant
(Kunyi) Method
Department)
Congo Lantana
Africa Leaves
Mwamuganga Decoction Antidiarrheal [36]
(Bouenza (Kunyi) (Mashi),
Democratic
Mavi ya kuku
Republic of Congo Leaves Decoction Antimalarial [37]
(Swahili),
(Bukavu and Uvira)
Makereshe
(Nande)

Africa Democratic Republic To treat cough by the


Decoction;
of Congo Nsudi nsudi Leaves and Ntându and Ndibu
administered [38]
(Kisantu and (Kikongo) fruits ethnic groups;
by rectal route
Mbanza-Ngungu) to treat hemorrhoids
Ethiopia
(Libo-Kemkem NR Leaves Infusion Antidiarrheal [39]
District)
Ethiopia
To treat skin infections,
(Mana Angetu NR Leaves Decoction [40]
gonorrhea, and “evil eye”
District)
Molecules 2025, 30, 851 5 of 51

Table 1. Cont.

Part of the Preparation


Region Country Vernacular Name Traditional Uses Ref.
Plant Method
Ethiopia Yewof kollo
Stems Infusion Antidiarrheal [41]
(Wonago Woreda) (Amharic)
Ethiopia Michi-charo To treat “Michi”, a type of
Leaves Topical [42]
(Sheko District) (Sheko) febrile illness
Galabert,
Decoction,
La Réunion Corbeille d’or Leaves Antimalarial [43]
infusion
(French)
Soussous, Malinké, Guerzé,
Guinea
Tagani Konon, and Manon ethnic
(Low, middle, upper, and Leaves Decoction [44]
(NR) groups use the plant to treat
forest ecological zones)
infectious diseases
Crushed; Kikuyu ethnic group uses
Rûîthiki,
Kenya directly applied the plant to treat common
Mûkenia Leaves [45,46]
(Central Province) to the ear to cold by inhaling
(Kikuyu)
treat otitis crushed leaves
Kenya
Mûkenia
(Embu and Leaves Decoction Antimalarial [47]
(Kikuyu)
Mbeere Districts)
Kenya
Nyabend winy Leaves and
(Formerly: Bondo District, Decoction To treat cough [48]
(Luo) roots
Africa now Siaya County)
Digos, Durumas, and
Kenya Mjsasa
Leaves Decoction Kambas ethnic groups use [49]
(Msambweni District) (Digo)
the plant as an antimalarial
Kenya
Leaves and Burnt for Used as a
(Rusinga Island NR [50]
seeds fumigation mosquito repellent
and Rambira)
Madagascar Kalabera To treat cough,
Aerial parts Decoction [51]
(Antsiranana) (NR) hypertension, and fever
Nigeria Wild sage Burnt for
Leaves Used as an insect repellent [52]
(Ibadan) (English) fumigation
Burnt for
Uganda Kapanga Used as a mosquito
Leaves fumigation; [53]
(Budiope County) (Lusoga) repellent; antimalarial
decoction
Kapanga
Uganda (Lusoga), Burnt for Used as a housefly and
Leaves [54]
(Budondo Subcounty) Tickberry fumigation insect repellent
(English)
Uganda Used to treat ringworms,
Leaves and
(Otwal and Ngai NR Maceration cataracts, snake bites, [55]
roots
Subcounties) and epilepsy
China Luo-ya-min Burnt for Used as a
Leaves [56]
(Xishuangbanna) (Chinese) fumigation mosquito repellent
Burnt for
India Bhoot-phool Used as an insect
Bark fumigation; [57]
(Assam) (Hindi) repellent; antimalarial
decoction
India Decoction,
NR Leaves Antimalarial [58]
Asia (Dharmapuri District) infusion
India Puttu Decoction, To treat several skin and
Leaves [59]
(Jharkhand State) (NR) pounded respiratory diseases
India Antiseptic, antimalarial,
NR Leaves Decoction [60]
(NR) and antirheumatic
Philippines Gainis Leaves and Burnt for
Used as an insect repellent [61]
(Paroc) (NR) stems fumigation
Molecules 2025, 30, 851 6 of 51

Table 1. Cont.

Part of the Preparation


Region Country Vernacular Name Traditional Uses Ref.
Plant Method
Van Kieu ethnic group uses
the plant alone or associated
Vietnam Thục Klay with roots of Mangifera
Roots Decoction [62]
(Hướng Hóa District) (NR) indica and barks of Erythrina
Asia variegata to treat abdominal
pain and diarrhea
Flowers,
Yemen Burnt for
NR leaves, and Used as an insect repellent [63]
(Hajjah District) fumigation
seeds
Alfombrilla,
To treat scorpion and insect
Mexico Gobernadora, Leaves and
Decoction stings; antidiarrheal [64]
(Querétaro) Ororuz stems
and antiparasitic
(Spanish)
North and Jøtskuy
Central Mexico (Zoque), Antidiarrheal, antiparasitic,
America Stems Decoction [65]
(Chiapas) Cinco negritos and antirheumatic
(Spanish)
Mexico Cinco negritos
Aerial parts Decoction Antidiarrheal [66]
(Puebla) (Spanish)
To treat respiratory diseases;
Brazil Cambará Decoction,
Leaves antipyretic and [67]
(Minas Gerais) (Tupi) infusion
antirheumatic
The Saramaccan Marron
ethnic group uses the plant
South Suriname
NR Leaves Decoction for the anti-inflammatory, [68]
America (Pikin Slee)
antiparasitic, and
depurative properties
Colombia
Venturosa Decoction;
(Antioquia Stems To treat snake bites [69]
(Spanish) steam bath
Department)
The France Mille-fleurs Decoction,
Flowers To treat flu syndrome [70]
Caribbean (Guadeloupe) (French) infusion
NR: not reported.

3.2. Phytochemistry
A total of 168 compounds have been described with different names in the considered
period. They include both specialized metabolites isolated from non-volatile fractions
of L. camara as well as semisynthetic derivatives. The distribution pattern of these com-
pounds includes steroids and triterpenoids (75.6%), flavonoids (14.3%), fatty acids and
other miscellaneous compounds (8.9%), and iridoid glucosides (1.2%).

Triterpenoids and Steroids


Steroids and pentacyclic triterpenoids are the predominant constituents isolated in the
indicated period from the non-volatile fractions of L. camara or obtained by semisynthesis.
Steroids (Table 2) are only a few and include common phytosterols such as stigmasterol (1),
β-sitosterol (3), and campesterol (5), in addition to the rare spirostane saponin yamogenin
II (6), which has a unique aglycone moiety. One hundred and twenty-one triterpenoids
(Table 3) have been described in the years considered in this review. Their molecular
structures belong to only five families, i.e., the protostane, euphane, lupane, oleanane, and
ursane ones. The last two skeletons are by far the most common. Alisol A (7) is the only
protostane isolated from L. camara, while euphanes are represented by eight triterpenoids
(8–15). The structures of most of them are characterized by a D7 double bond, an oxidized α-
substituent at C-4, and a γ-lactone E ring that is trans-fused to the cyclopentane D ring and
bears an unsaturated homoprenyl chain. The small lupane family (16–18) includes the rare
Molecules 2025, 30, 851 7 of 51

lantabetulic acid (17), which is characterized by an ether β-bridge connecting C-3 with C-25,
and the highly bioactive betulinic acid (18). A total of 78 oleanane triterpenoids (19–96),
including 34 synthetic ones, and 31 ursane derivatives (97–127), including 6 synthetic ones,
have been described in the considered period. The two skeletons differ from the position of
the methyl groups C-29 and C-30, which are positioned on the quaternary carbon C-20 in
the oleanane compounds, while they are trans-oriented on the tertiary carbons C-19 and
C-20 in the ursane derivatives.
The great variety of oleanane triterpenoids from L. camara derive from a combination
of differently placed double bonds and different oxygenated groups that decorate the basic
skeleton. A β-COOH, as in compound 30, or a β-COOMe group, as in 33, is usually linked
to C-17, with cis-orientation to βH-18. When a carboxylic group is absent at C-17, a D17(18)
double bond occurs, as in triterpenoid 49; a D12(13) double bond is usually present, as in
22, while, very rarely, a double bond occurs between C-11 and C-12, as in 21, or between
C-1 and C-2, as in 78. One compound (54) containing a 9(11),12(13)-diene system has also
been isolated. A carbonyl group is usually present at C-3, as in 26, or at C-11, as in 20; in
one compound, 40, a CO occurs at C-22. An acetal system formed by a β-epoxide bridge
between C-25 and C-3 and an α-OH (or, very rarely, an α-alkoxy) group at C-3 is frequently
present in oleanane structures, as in 29 or 40. A few compounds are known to contain a
lactone ring formed by a β-oxygen atom at C-13 bonded to a β-CO group at C-17, as in 43.
One example (34) of a triterpenoid bearing a β-epoxy ring at C-21/C-22 has been isolated
from L. camara. A free βOH group usually occurs at C-3, as in 31, at C-22, as in 26, and at
C-24, as in 28; very rarely, an OH is present at C-2, as in 78, or at C-19, as in 25, at C-7, as in
23, C-9, as in 64, and C-12, as in 43. One 3-O-acyl (compound 38) and one 3-O-β-D-glucosyl
derivative (84) have been isolated. The 22-βOH is usually esterified with an acyl group,
e.g., an acetyl, as in 42, a propanoyl, as in 46, a butanoyl, as in 50, an isobutanoyl, as in
51, an angelyl [(Z)-2-methylbut-2-enoyl], as in 48, and a senecioyl (3-methylbut-2-enoyl)
residue, as in 49; rarer are the esters with (S)-2-mehylbutanoic acid, as the triterpenoid 66,
and (S)-3-hydroxy-2-methylidenebutanoic acid, as 69.
Most of these structural characteristics are shared by the ursane triterpenoids due
to the close biosynthetic origin of the oleanane and ursane families. A unique ursane
triterpenoid is the 3-O-β-D-glucosyl derivative 127, in which stearic acid is esterified to the
4-OH group of a glucosyl moiety.
The flavonoids (Table 4) are represented by 19 flavones (128–146), which are mainly
apigenin and luteolin derivatives. A semisynthetic derivative (144) is included. Three rare
O-methyl flavonols (147–149) and two isoflavones, i.e., 5,7-dihydroxy-6,3′ ,4′ -trimethoxy
isoflavone (150) and triglycoside 151, have also been isolated. The two iridoids 152 and
153 (Table 5) are the 1-O glucosides of the common aglycones genipin and 4a-OH genipin.
The fatty acids 154–162 (Table 6) include common saturated and unsaturated long-chain
homologues from C14 to C32 . Finally, the small group of miscellaneous metabolites 163–168
(Table 7) includes the toxic cyanogenic glucoside linamarin (164), the common aliphatic
alcohols phytol (165), and triacontane-1-ol (167).
Compounds isolated from L. camara and semisynthetic derivatives are listed in the
following Tables 2–7. The compounds with the same molecular skeleton are grouped and
are then listed by increasing molecular formulae. Compounds with the same molecular
formula are listed in alphabetic order.
Molecules 2025, 30, 851 8 of 51

Table 2. Steroids isolated from non-volatile fractions of Lantana camara.

Molecular Molecular Skeleton


Nº Compound Name Part of the Plant/Solvent Reference
Formula Weight Type
Stigmasterol Leaves/methanol
(1) C29 H48 O 412.702 Stigmastane [71]
(Stigmasta-5,22-dien-3β-ol) Stems/methanol
7-Oxo-β-sitosterol Stems/methanol
(2) C29 H48 O2 428.701 Stigmastane [19,71]
3β-Hydroxy-stigmast-5-en-7-one Roots/chloroform
Aerial parts/petroleum ether
Aerial parts/96% ethanol
Fruits/chloroform
β-Sitosterol Leaves/methanol
(3) Stigmast-5-en-3β-ol C29 H50 O 414.718 Stigmastane Stems/95% ethanol, methanol [19,72–74]
(Figure 4) Roots/chloroform
Leaves, stems,
and roots/
petroleum ether
β-Sitosterol 3-O-β-D-glucopyranoside Aerial parts/methanol
(4) 3-O-β-D-Glucopyranosyl-stigmast-5-en- C35 H60 O6 576.859 Stigmastane Leaves/methanol [15,71,74–77]
3β-ol Stems/95% ethanol, methanol
Campesterol Leaves/methanol
(5) C28 H48 O 400.691 Campestane [71]
Campest-5-en-3β-ol Stems/methanol
Yamogenin II
(25S)-Spirostan-5-ene-3β,21-diol-3-O-α-
L-rhamnopyranosyl-(1′′ 33→2′ )-[α-L-
(6) C45 H72 O17 885.054 Spirostane Leaves/methanol [78,79]
rhamnopyranosyl-(1′′′ →4′ )]-β-D-
glucopyranoside
(Figure 4)
Alisol A
(7) (8α,9β,11β,14β,23S,24R)-11,23,24,25- 490.725 Protostane Roots/chloroform [19]
C30 H50 O5
tetrahydroxy-protost-13(17)-en-3-one
(Figure 4)
Lantrieuphpene B
(8) C31 H44 O5 496.688 Euphane Aerial parts/methanol [80]
(Figure 4)
Lantrieuphpene C
(9) C31 H46 O5 498.704 Euphane Aerial parts/methanol [80]
(Figure 4)
Euphane monolactone A
(10) C32 H46 O6 526.714 Euphane Leaves/acetonitrile [72]
(Figure 4)
Euphane monolactone B
(11) C32 H46 O7 542.713 Euphane Leaves/acetonitrile [72]
(Figure 4)
Lantrieuphpene D
(12) C32 H48 O6 528.730 Euphane Aerial parts/methanol [80]
(Figure 4)
Lantrieuphpene A
(13) C33 H46 O7 554.724 Euphane Aerial parts/methanol [80]
(Figure 4)
Euphane monolactone C
(14) C40 H58 O14 762.890 Euphane Leaves/acetonitrile [72,80]
(Figure 4)
Euphane monolactone D
(15) C42 H60 O15 804.927 Euphane Leaves/acetonitrile [72]
(Figure 4)
Leaves/methanol
Betulonic acid
Stems/methanol
(16) 3-oxo-lup-20(29)-en-28-oic acid C30 H46 O3 454.695 Lupane [61,71,79]
Leaves and stems/petroleum
(Figure 5)
ether
Lantabetulic acid
3,25-β-epoxy-3α-hydroxy-lup-20 Leaves and stems/petroleum
(17) C30 H46 O4 470.694 Lupane [81]
(29)-en-28-oic acid ether
(Figure 5)
Aerial parts/methanol
Leaves/methanol
Betulinic acid [71,72,78,79,
(18) C30 H48 O3 456.711 Lupane Stems/methanol
3β-hydroxy-lup-20 (29)-en-28-oic acid 82]
Leaves and stems/
petroleum ether
Molecules 2025, 30, 851 9 of 51

Table 3. Triterpenoids isolated from non-volatile fractions of Lantana camara and semisynthetic
derivatives.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
Camaradienone
3,25-β-epoxy-3α-hydroxy-28-nor-
(19) C29 H42 O3 438.652 Oleanane Aerial parts/methanol [15]
oleana-12,17-dien-11-one
(Figure 5)
Lantanoic acid
3,25-β-epoxy-3α-hydroxy-11-oxo-olean-
(20) C30 H44 O5 484.677 Oleanane Aerial parts/methanol [83]
12-en-28-oic acid
(Figure 5)
3β-Hydroxy-olean-11-en-28,13-β-olide
(21) C30 H46 O3 454.695 Oleanane HD * [74]
(Figure 5)
Aerial parts/ethanol, methanol
Leaves/ethanol
Leaves and stems/methanol,
Oleanonic acid
petroleum ether [15,71,73–
(22) 3-Oxo-olean-12-en-28-oic acid C30 H46 O3 454.695 Oleanane
Stems/ethanol, methanol 79,81–88]
(Figure 5)
Roots/ethyl acetate, methanol,
n-hexane–ethyl acetate–methanol
(1:1:1)
Camarin
(23) 7α-hydroxy-3-oxo-olean-12-en-28-oic acid C30 H46 O4 470.694 Oleanane Aerial parts/methanol [89,90]
(Figure 5)
4-epi-Hederagonic acid
Aerial parts/ethanol
24-hydroxy-3-oxo-olean-12-en-28-
(24) C30 H46 O4 470.694 Oleanane Leaves and stems/ [84]
oic acid
petroleum ether
(Figure 5)
19α-Hydroxy-oleanonic acid
(S)-19α-hydroxy-3-oxo-olean-12-en-
(25) C30 H46 O4 470.694 Oleanane Aerial parts/methanol [80]
28-oic acid
(Figure 5)
Aerial parts/ethanol
22β-Hydroxy-oleanonic acid
HD
22β-hydroxy-3-oxo-olean-12-en-
(26) C30 H46 O4 470.694 Oleanane Leaves/acetonitrile [75,84,91–97]
28-oic acid
Leaves and stems/petroleum
(Figure 5)
ether
Aerial parts/methanol
HD
Lantanolic acid
Leaves/chloroform, methanol, [15,75,77,79,
3,25-β-epoxy-3α-hydroxy-olean-12-en-
(27) C30 H46 O4 470.694 Oleanane petroleum ether 82,84,89,90,
28-oic acid
Leaves and stems/ 92,98]
(Figure 5)
petroleum ether
Roots/ethanol
22β-Hydroxy-4-epi-hederagonic acid
(28) C30 H46 O5 486.693 Oleanane Aerial parts/ethanol [84]
(Figure 5)
Lantacamaric acid A
3,25-β-epoxy-3α,24-dihydroxy-olean-12-
(29) C30 H46 O5 486.693 Oleanane Leaves and stems/methanol [85]
en-28-oic acid
(Figure 5)
Lantaninilic acid
Aerial parts/methanol
3,25-β-epoxy-3α,22β-dihydroxy-olean- [75,82,83,85,
(30) C30 H46 O5 486.693 Oleanane HD *
12-en-28-oic acid 89,90]
Leaves and stems/methanol
(Figure 5)
Aerial parts/ethanol, methanol
Leaves/methanol
Leaves and stems/
petroleum ether [15,19,74–
Oleanolic acid
Stems/ethanol, methanol 77,79,80,82,
(31) 3β-hydroxy-olean-12-en-28-oic acid C30 H48 O3 456.711 Oleanane
Roots/ethanol, ethyl acetate, 84,86,87,90,
(Figure 6)
(52.5% methanol/47.5% ethyl 91,98–102]
acetate), (60% chloroform/
40% methanol), n-hexane–ethyl
acetate–methanol (1:1:1)
Molecules 2025, 30, 851 10 of 51

Table 3. Cont.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
22β-Hydroxy-oleanolic acid
Aerial parts/ethanol
3β,22β-dihydroxy-olean-12-en-
(32) C30 H48 O4 472.710 Oleanane HD * [84,95,96]
28-oic acid
Roots/ethanol
(Figure 6)
Methyl lantanoate
methyl 3,25-β-epoxy-3α-hydroxy-11-oxo-
(33) C31 H46 O5 498.704 Oleanane HD * [83]
olean-12-en-28-oate
(Figure 6)
21,22-β-Epoxy-3β-hydroxy-olean-12-en-
28-oic acid,
isolated as Roots/n-hexane–ethyl
(34) C31 H48 O4 484.721 Oleanane [74]
Methyl 21,22-β-epoxy-3β-hydroxy- acetate–methanol (1:1:1)
olean-12-en-28-oate
(Figure 6)
Methyl 22β-hydroxy-oleanonate
methyl 22β-hydroxy-3-oxo-
(35) C31 H48 O4 484.721 Oleanane HD * [93,94]
olean-12-en-28-oate
(Figure 6)
Methyl lantaninilate
methyl 3,25-β-epoxy-3α,22β-dihydroxy-
(36) C31 H48 O5 500.720 Oleanane HD * [75]
olean-12-en-28-oate
(Figure 6)
22β-Acetyloxy-oleanonic acid
22β-acetyloxy-3-oxo-olean-12-en-
(37) C32 H48 O5 512.731 Oleanane HD * [94]
28-oic acid
(Figure 6)
Lantanone
3β-acetyloxy-11-oxo-olean-12-en-
(38) C32 H48 O5 512.731 Oleanane Aerial parts/ethanol [101]
28-oic acid
(Figure 6)
Methyl 3,25-β-epoxy-3α-methoxy-22-
(39) oxo-olean-12-en-28-oate C32 H48 O5 512.731 Oleanane HD * [103]
(Figure 6)
22β-Acetyloxy-4-epi-hederagonic acid
(40) C32 H48 O6 528.730 Oleanane Aerial parts/ethanol [104]
(Figure 6)
Lancamarinic acid
22β-acetyloxy-3,25-β-epoxy-3α-
(41) C32 H48 O6 528.730 Oleanane Aerial parts/methanol [105]
hydroxy-olean-12-en-28-oic acid
(Figure 6)
Lancamarolide
22β-acetyloxy-3,25-β-epoxy-3α,12α-
(42) C32 H48 O7 544.729 Oleanane Aerial parts/methanol [82]
dihydroxyolean-28,13-β-olide
(Figure 6)
Lancamaric acid
3,25-β-epoxy-3α-ethoxy-olean-12-en-28-
(43) C32 H50 O4 498.748 Oleanane Aerial parts/methanol [85]
oic acid
(Figure 6)
Aerial parts/methanol
Oleanolic acid 3-O-acetate
HD * [74,77,102,
(44) 3β-acetyloxy-olean-12-en-28-oic acid C32 H50 O4 498.748 Oleanane
Roots/n-hexane–ethyl 106]
(Figure 6)
acetate–methanol (1:1:1)
Methyl 22β-acetyloxy-oleanonate
methyl 22β-acetyloxy-3-oxo-olean-12-en-
(45) C33 H50 O5 526.758 Oleanane HD * [94]
28-oate
(Figure 6)
22β-Propanoyloxy-oleanonic acid
22β-propanoyloxy-3-oxo-olean-12-en-
(46) C33 H50 O5 526.758 Oleanane HD * [94]
28-oic acid
(Figure 6)
Molecules 2025, 30, 851 11 of 51

Table 3. Cont.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
Methyl 22-O-acetyl-lantaninilate
methyl 22β-acetyloxy-3,25-β-epoxy-3α-
(47) C33 H50 O6 542.757 Oleanane HD * [103,107]
hydroxy-olean-12-en-28-oate
(Figure 6)
Lantadienone
22β-angelyloxy-3,25-β-epoxy-3α-hydroxy-
(48) C34 H48 O5 536.753 Oleanane Aerial parts/methanol [15]
28-nor-oleana-12,17-dien-11-one
(Figure 7)
Lantigdienone
3,25-β-epoxy-3α-hydroxy-11-oxo-22β-
(49) C34 H48 O5 536.753 Oleanane Aerial parts/methanol [108]
senecioyloxy-28-nor-olean-12,17-diene
(Figure 7)
22β-Butanoyloxy-oleanonic acid
22β-butanoyloxy-3-oxo-olean-12-en-28-
(50) C34 H52 O5 540.785 Oleanane HD * [94]
oic acid
(Figure 7)
Lantadene D Aerial parts/ethanol
22β-isobutyryloxy-3-oxo-olean-12-en-28- HD *
(51) C34 H52 O5 540.785 Oleanane [19,84,94,97]
oic acid Leaves/acetonitrile, methanol,
(Figure 7) petroleum ether
Methyl 22β-propanoyloxy-oleanonate
methyl 22β-propanoyloxy-3-oxo-olean-
(52) C34 H52 O5 540.785 Oleanane HD * [92]
12-en-28-oate
(Figure 7)
24-Hydroxylantadene D
22β-isobutyryloxy-24-hydroxy-3-oxo-
(53) C34 H52 O6 556.784 Oleanane Aerial parts/ethanol [84]
olean-12-en-28-oic acid
(Figure 7)
Lantrigloylic acid
3,25-β-epoxy-3α-hydroxy-22β-
(54) senecioyloxy-olea-9 (11),12-dien- C35 H50 O6 566.779 Oleanane Aerial parts/methanol [90]
28-oic acid
(Figure 7)
Camangeloyl acid
3,25-β-epoxy-3α-hydroxy-22β-[(Z)-2-
[15,77,83,89,
(55) methyl-2-butenoyloxy]-11-oxo-olean-12- C35 H50 O7 582.778 Oleanane Aerial parts/methanol
106]
en-28-oic
(Figure 7)
Camarinin
3,25-β-epoxy-3α-hydroxy-22β-
[83,84,89,90,
(56) (3-methyl-2-butenoyloxy)- C35 H50 O7 582.778 Oleanane Aerial parts/methanol
107]
11-oxo-olean-12-en-28-oic
(Figure 7)
Lantadene A nitrile
22β-angelyloxy-28-ciano-3-oxo-olean-
(57) C35 H51 NO3 533.797 Oleanane HD * [93]
12-ene
(Figure 7)
Lantadene A acyl chloride
28-chloro-22β-angelyloxy-3,28-dioxo-
(58) C35 H51 ClO4 571.239 Oleanane HD * [93]
olean-12-ene
(Figure 7)
22β-Angelyloxy-3-oxo-olean-28,13-
(59) β-olide C35 H52 O5 552.796 Oleanane HD * [108]
(Figure 7)
Molecules 2025, 30, 851 12 of 51

Table 3. Cont.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
Aerial parts/methanol, ethanol
Leaves/acetone, acetonitrile,
ethanol, ethyl acetate, methanol,
Lantadene A
petroleum ether, methanol–water
(Rehmannic acid)
(70:30)
(60) 22β-angelyloxy-3-oxo-olean-12-en-28- C35 H52 O5 552.796 Oleanane [108]
Leaves and stems/methanol,
oic acid
petroleum ether
(Figure 7)
Roots/n-hexane–ethyl
acetate–methanol (1:1:1); ethanol
Stems/ethanol, methanol
Aerial parts/ethanol,
dichloromethane, methanol,
petroleum ether
Leaves/acetonitrile, 96% ethanol,
Lantadene B
ethyl acetate [15,19,79,82,
3-oxo-22β-senecioyloxy-
(61) C35 H52 O5 552.796 Oleanane Leaves/methanol, 84,91,95,96,
olean-12-en-28-oic acid
(70% methanol/30% water) 98,109–116]
(Figure 7)
Leaves and stems/
petroleum ether
Stems/methanol
Roots/ethanol
Camaric acid Aerial parts/
3,25-β-epoxy-3α-hydroxy-22β-[(Z)-2- dichloromethane, methanol [15,74,77,80,
(62) methyl-2-butenoyloxy]-olean-12-en-28- C35 H52 O6 568.795 Oleanane Leaves and stems/methanol 82,84,90,98,
oic acid Roots/n-hexane–ethyl 99,117,118]
(Figure 7) acetate–methanol (1:1:1)
3,25-β-Epoxy-3α-hydroxy-22β-[(E)-2-
methyl-2-butenoyloxy]-olean-12-en-28-
(63) C35 H52 O6 568.795 Oleanane Aerial parts/ethanol [84]
oic acid
(Figure 7)
9-Hydroxy-lantadene A
22β-angelyloxy-9-hydroxy-3-oxo-olean-
(64) C35 H52 O6 568.795 Oleanane Leaves/ethyl acetate, methanol [119,120]
12- en-28-oic acid
(Figure 7)
24-Hydroxylantadene B
≡ 24-Hydroxy-22β-senecioyloxy-
Aerial parts/ethanol
oleanonic acid
(65) C35 H52 O6 568.795 Oleanane Leaves/ethyl acetate [84,99,116]
24-hydroxy-3-oxo-22β-senecioyloxy-
Leaves and stems/methanol
olean-12-en-28-oic acid
(Figure 7)
24-Hydroxy lantadene X
24-hydroxy-3-oxo-22β-[(E)-2-methylbut-
(66) C35 H52 O6 568.795 Oleanane Aerial parts/ethanol [84]
2-enoyloxy]- olean-12-en-28-oic acid
(Figure 7)
Icterogenin
≡ 24-Hydroxy-lantadene A Aerial parts/ethanol, methanol
≡ 24-Hydroxy-22β-angelyloxy- Leaves/acetone, ethanol [19,79–
(67) oleanonic acid C35 H52 O6 568.795 Oleanane Leaves/ethyl acetate, methanol 82,84,99,109,
24-hydroxy-3-oxo-22β-[(Z)-2-methylbut- Leaves and stems/methanol, 116,118,121]
2-enoyloxy]- olean-12-en-28-oic acid petroleum ether
(Figure 7)
Aerial parts/dichloromethane,
Lantanilic acid ethanol, methanol [15,19,72,77,
3β,25-β-epoxy-3α-hydroxy-22β- Leaves/ethanol, ethyl acetate, 79,81,84,85,
(68) C35 H52 O6 568.795 Oleanane
senecioyloxy-olean-12-en-28-oic acid methanol, petroleum ether 98–100,109,
(Figure 7) Leaves and stems/methanol 116,122,123]
Roots/chloroform
Molecules 2025, 30, 851 13 of 51

Table 3. Cont.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
Camarolic acid
3,25-β-epoxy-3α-hydroxy-22β-[(S)-3-
hydroxy-2-methylidenebutanoyloxy]
(69) C35 H52 O7 584.794 Oleanane Aerial parts/methanol [82,90]
olean-
12-en-28-oic acid
(Figure 7)
Lantacamaric acid B
3,25-β-epoxy-3α,24-dihydroxy-22β-
(70) C35 H52 O7 584.794 Oleanane Leaves and stems/methanol [85]
senecioyloxy-olean-12-en-28-oic acid
(Figure 7)
Lantadene A amide
28-amino-22β-angelyloxy-3,28-dioxo-
(71) C35 H53 NO4 551.812 Oleanane HD * [93]
olean-12-ene
(Figure 7)
HD *
Lantadene C
Leaves/acetonitrile, ethyl
22β-[(S)-2-methylbutanoyloxy]-3-oxo- [91,94,109,
(72) C35 H54 O5 554.812 Oleanane acetate, methanol
olean-12-en-28-oic acid 116,124]
Leaves and stems/
(Figure 7)
petroleum ether
Methyl 22β-butanoyloxy-oleanonate
methyl 22β-butanoyloxy-3-oxo-olean-12-
(73) C35 H54 O5 554.812 Oleanane HD * [94]
en-28-oate
(Figure 7)
Methyl 22β-isobutyryloxy-oleanonate
methyl 22β-isobutyryloxy-3-oxo-olean-
(74) C35 H54 O5 554.812 Oleanane HD * [94]
12-en-28-oate
(Figure 7)
Reduced lantadene A Aerial parts/ethanol
22β-angelyloxy-3β-hydroxy-olean-12- HD *
(75) C35 H54 O5 554.812 Oleanane [84,91,95]
en-28-oic acid Leaves/methanol, acetonitrile
(Figure 8) Roots/ethanol
Reduced lantadene B Aerial parts/ethanol
3β-hydroxy-22β-senecioyloxy-olean-12- HD *
(76) C35 H54 O5 554.812 Oleanane [84,91,95]
en-28-oic acid Leaves/acetonitrile, methanol
(Figure 8) Roots/ethanol
Reduced lantadene C
3β-hydroxy-22β-[2-methylbutanoyloxy]-
(77) C35 H56 O5 554.812 Oleanane Aerial parts/ethanol [84]
olean-12-en-28-oic acid
(Figure 8)
Methyl 22β-angelyloxy-2-hydroxy-3-
oxo-olean-1,12-diene-
(78) C36 H52 O6 580.806 Oleanane HD * [124]
28-oate
(Figure 8)
Lancamarinin
methyl 3,25-β-epoxy-3α-hydroxy-11-oxo-
HD *
(79) 22β-senecioyloxy-olean-12-en- C36 H52 O7 596.805 Oleanane [105]
Aerial parts/methanol
28-oate
(Figure 8)
Methyl camangeloylate
methyl 3,25-β-epoxy-3α-hydroxy-22β-
(80) [(Z)-2′ -methyl-2′ -butenoyloxy]-11-oxo- C36 H52 O7 596.805 Oleanane HD * [77]
olean-12-en-28-oate
(Figure 8)
Lantadene A
methyl ester HD *
(81) methyl 22β-angelyloxy-3-oxo-olean-12- C36 H54 O5 566.823 Oleanane Leaves and stems/ [93,124]
en-28-oate petroleum ether
(Figure 8)
Methyl 22-β-angelyloxy-lantanolate
methyl 22β-angelyloxy-3,25-β-epoxy-3α-
(82) C36 H54 O6 582.822 Oleanane HD * [82]
hydroxy-olean-12-en-28-oate
(Figure 8)
Molecules 2025, 30, 851 14 of 51

Table 3. Cont.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
Methyl camarolate
methyl 3,25-β-epoxy-3α-hydroxy-
22β-[(S)-3-hydroxy-2-
(83) C36 H54 O7 598.821 Oleanane HD * [90]
methylidenebutanoyloxy}
olean-12-en-28-oate
(Figure 8)
3-O-β-D-Glucosyl oleanolic acid
3-O-β-D-glucopyranosyloxy-olean-12-
(84) C36 H58 O8 618.852 Oleanane Leaves/methanol [125,126]
en-28-oic acid
(Figure 8)
22β-Benzoyloxy-oleanonic acid
22β-benzoyloxy-3-oxo-olean-12-en-28-
(85) C37 H50 O5 574.802 Oleanane HD * [94]
oic acid
(Figure 8)
Methyl 22β-benzoyloxy-oleanonate
methyl 22β-benzoyloxy-3-oxo-olean-12-
(86) C38 H52 O5 588.829 Oleanane HD * [94]
en-28-oate
(Figure 8)
3β-(2-Acetyloxybenzoyloxy)-22β-
(87) hydroxy-olean-12-en-28-oic acid C39 H54 O7 634.854 Oleanane HD * [96]
(Figure 8)
3β-[(R,S)-2-(4-
Isobutylphenyl)propanoyloxy]-22β-
(88) C43 H64 O5 660.980 Oleanane HD * [96]
hydroxy-olean-12-en-28-oic acid
(Figure 8)
3β-{2-[2-(2,6-
Dichlorophenylamino)phenyl]acetyloxy}-
(89) C44 H57 Cl2 NO5 750.842 Oleanane HD * [96]
22β-hydroxy-olean-12-en-28-oic acid
(Figure 8)
3β-[(+)-(S)-2-(6-Methoxy-2-
naphthyl)propanoyloxy]-22β-hydroxy-
(90) C44 H60 O6 684.958 Oleanane HD * [96]
olean-12-en-28-oic acid
(Figure 8)
3β-[(R,S)-2-(3-
Benzoylphenyl)propanoyloxy]-22β-
(91) C46 H60 O6 708.980 Oleanane HD * [96]
hydroxy-olean-12-en-28-oic acid
(Figure 9)
3β,22β-Di-(2-acetyloxybenzoyloxy)-
(92) olean-12-en-28-oic acid C48 H60 O10 796.998 Oleanane HD * [96]
(Figure 9)
3β,22β-Di-[(R.S)-2-(4-
isobutylphenyl)propanoyloxy]-olean-
(93) C56 H80 O6 849.250 Oleanane HD * [96]
12-en-28-oic acid
(Figure 9)
3β,22β-Di-{2-[2-(2,6-
dichlorophenylamino)phenyl]acetyloxy}- C58 H66 Cl4 N2
(94) 1028.974 Oleanane HD * [96]
olean-12-en-28-oic acid O6
(Figure 9)
3β,22β-Di-[(+)-(S)-2-(6-methoxy-2-
naphthyl)propanoyloxy]-olean-12-en-
(95) C58 H72 O8 897.206 Oleanane HD * [96]
28-oic acid
(Figure 9)
3β,22β-Di-[(R,S)-2-(3-
benzoylphenyl)propanoyloxy]-olean-
(96) C62 H72 O8 945.250 Oleanane HD * [96]
12-en-28-oic acid
(Figure 9)
Camarolide
(97) 3-oxo-urs-11-en-28,13-β-olide C30 H44 O3 452.679 Ursane Aerial parts/methanol [85]
(Figure 10)
3,24-Dioxo-urs-12-en-28-oic acid
(98) C30 H44 O4 468.678 Ursane Leaves/solvent not reported [127]
(Figure 10)
Molecules 2025, 30, 851 15 of 51

Table 3. Cont.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
Camaranoic acid
3,25-β-epoxy-3α-hydroxy-11-oxo-urs-12-
(99) C30 H44 O5 484.677 Ursane Aerial parts/methanol [81,83,118]
en-28-oic acid
(Figure 10)
Ursonic acid Aerial parts/methanol
(100) 3-oxo-urs-12-en-28-oic acid C30 H46 O3 454.695 Ursane Leaves and stems/ [81,83–85]
(Figure 10) petroleum ether
11α-Hydroxy-3-oxo-urs-12-en-28-oic
(101) acid C30 H46 O4 470.694 Ursane Aerial parts/methanol [82]
(Figure 10)
Aerial parts/methanol
Lantic acid
Leaves/chloroform
3,25-β-epoxy-3α-hydroxy-urs-12-en-28-
(102) C30 H46 O4 470.694 Ursane Leaves and [75,83,98]
oic acid
stems/dichloromethane–
(Figure 10)
methanol (1:1), petroleum ether
11-Oxo-β-boswellic acid
(103) 3α-hydroxy-11-oxo-urs-12-en-24-oic acid C30 H46 O4 470.694 Ursane Leaves/ethyl acetate [120]
(Figure 10)
Aerial parts/ethanol, methanol
Pomonic acid
Roots/
(104) 19α-hydroxy-3-oxo-urs-12-en-28-oic acid C30 H46 O4 470.694 Ursane [74,77,80,84]
n-hexane–ethyl acetate–methanol
(Figure 10)
(1:1:1)
Lantoic acid
3,25-β-epoxy-3α,22β-dihydroxy-urs-12- Aerial parts/methanol
(105) C30 H46 O5 486.693 Ursane [81,84,90,118]
en-28-oic acid Leaves/petroleum ether
(Figure 10)
Ursolic acid
3β-hydroxy-urs-12-en-28-oic acid; Aerial parts/methanol [19,79,89,90,
(106) C30 H48 O3 456.711 Ursane
urs-12-en-3β-ol-28-oic acid Leaves/methanol 121,125]
(Figure 10)
Pomolic acid Aerial parts/methanol
(107) 3β,19α-dihydroxy-urs-12-en-28-oic acid C30 H48 O4 472.710 Ursane Stems/methanol [84,89,90,98]
(Figure 10) Roots/chloroform, ethanol
α-Amyrin Aerial parts/96% ethanol,
(108) C30 H50 O 426.729 Ursane [73,74]
urs-12-en-3β-ol petroleum ether
Roots/
3β,19α-Dihydroxy-ursan-28-oic acid
(109) C30 H50 O4 474.726 Ursane n-hexane–ethyl acetate–methanol [74]
(Figure 10)
(1:1:1)
Methyl camaranoate
methyl 3,25-β-epoxy-3α-hydroxy-11-oxo-
(110) C31 H46 O5 498.704 Ursane HD * [83]
urs-12-en-28-oate
(Figure 10)
Ursoxy acid
3,25-β-epoxy-3α-methoxy-urs-12-en-28-
(111) C31 H48 O4 484.721 Ursane Aerial parts/methanol [106]
oic acid
(Figure 10)
Methyl 25-hydroxy-3-deoxy-ursen-12-
(112) en-28-oate C31 H50 O3 470.738 Ursane HD * [128]
(Figure 10)
Methyl 3β,19α-dihydroxy ursan-28-oate
(113) C31 H52 O4 488.753 Ursane HD * [74]
(Figure 10)
Aerial parts/methanol
Camarinic acid
Leaves/chloroform
22β-acetyloxy-3,25-β-epoxy-3α-
(114) C32 H48 O6 528.730 Ursane Leaves and [16,83,98,109]
hydroxy-12-ursen-28-oic acid
stems/dichloromethane–
(Figure 10)
methanol (1:1)
Methyl ursoxylate
methyl 3,25-β-epoxy-3α-methoxy-urs-12- Aerial parts/methanol
(115) C32 H50 O4 498.748 Ursane [106]
en-28-oate HD *
(Figure 10)
Molecules 2025, 30, 851 16 of 51

Table 3. Cont.

Molecular Molecular Skeleton


Nº Compound Part of the Plant/Solvent Reference
Formula Weight Type
Ursethoxy acid
3,25-β-epoxy-3α-ethoxy-urs-12-en-28-
(116) C32 H50 O4 498.748 Ursane Aerial parts/methanol [129]
oic acid
(Figure 10)
Methyl camaralate
methyl 22β-acetoxy-3,25-β-epoxy-3α- Aerial parts/methanol
(117) C33 H50 O6 542.757 Ursane [77,98]
hydroxy-urs-12-en-28-oate HD *
(Figure 10)
Methyl ursethoxylate
methyl 3,25-β-epoxy-3α-ethoxy-urs-12-
(118) C33 H52 O4 512.775 Ursane Aerial parts/methanol [129]
en-28-oate
(Figure 10)
Lantacin
3β,19α-dihydroxy-22β-senecioyloxy-urs-
(119) C35 H54 O6 570.811 Ursane Aerial parts/methanol [84,89,118]
12-en-28-oic acid
(Figure 10)
Lantaiursolic acid
3β-isovaleroyloxy-19α-hydroxy-urs-12-
(120) C35 H56 O5 556.828 Ursane Roots/ethanol [118]
en-28-oic acid
(Figure 10)
Camaracinic acid
22β-angelyloxy-3,25-β-epoxy-3α-
(121) C36 H54 O6 582.822 Ursane Aerial parts/methanol [82]
methoxy-12-ursen-28-oic acid
(Figure 11)
Camaryolic acid
3,25-β-epoxy-3α-methoxy-22β-
(122) C36 H54 O6 582.822 Ursane Aerial parts/methanol [77,82]
senecioyloxy-urs-12-en-28-oic acid
(Figure 11)
Methyl lantacinate
methyl
(123) 3β,19α-dihydroxy-22β-senecioyloxy- C36 H56 O6 584.838 Ursane HD * [84]
urs-12-en-28-oate
(Figure 11)
Methyl camaracinate
methyl 22β-angelyloxy-3,25-β-epoxy-3α-
(124) C37 H56 O6 596.849 Ursane HD * [82]
methoxy-12-ursen-28-oate
(Figure 11)
Methyl camaryolate
methyl 3,25-β-epoxy-3α-methoxy-22β-
(125) C37 H56 O6 596.849 Ursane HD * [82]
senecioyloxy-urs-12-en-28-oate
(Figure 11)
Ursangilic acid
22β-angelyloxy-
(126) 3,25-β-epoxy-3α-ethoxy-urs-12-en-28- C37 H56 O6 596.849 Ursane Aerial parts/methanol [106]
oic acid
(Figure 8)
Urs-12-en-3β-ol-28-oic acid
(127) 3-O-β-D-glucopyranosyl-4′ - C54 H92 O9 885.321 Ursane Leaves/methanol [125,126]
octadecanoate (Figure 11)
* HD: semisynthetic derivative.
Molecules 2025, 30, 851 17 of 51

Table 4. Flavonoids isolated from non-volatile fractions of Lantana camara and semisynthetic derivatives.

Molecular Molecular Flavonoid


N.º Compound Part of the Plant/Solvent Reference
Formula Weight Type
Hispidulin
Leaves/ethanol
(128) 4′ ,5,7-trihydroxy-6-methoxyflavone C16 H12 O6 300.266 Flavone [63,79,84]
Stems/methanol
(Figure 12)
Pectolinarigenin
(129) 5,7-dihydroxy-4′ ,6-dimethoxyflavone C17 H14 O6 314.293 Flavone Leaves/methanol [19,79,84,130]
(Figure 12)
Tricin
4′ ,5,7-trihydroxy-3′ ,5′ -
(130) C17 H14 O7 330.292 Flavone Leaves/methanol [79]
dimethoxyflavone
(Figure 12)
Apigenin 7-O-β-D-galacturonide
7-O-β-D-galacturonyl-4′ ,5,7-
(131) C21 H18 O11 446.364 Flavone Flowers/methanol–water (70:30) [131]
trihydroxyflavone
(Figure 12)
Anthemoside
Apigenin 7-O-β-D-glucopyranoside
(132) 7-O-β-D-glucopyranosyl- C21 H20 O10 432.381 Flavone Flowers/methanol–water (70:30) [132]
4′ ,5,7-trihydroxyflavone
(Figure 12)
Isovitexin
6-C-β-D-glucopyranosyl-4′ ,5,7- Flowers/
(133) C21 H20 O10 432.381 Flavone [132]
trihydroxyflavone methanol–water (70:30)
(Figure 12)
Vitexin
8-C-β-D-glucopyranosyl-4′ ,5,7- Flowers/
(134) C21 H20 O10 432.381 Flavone [132,133]
trihydroxyflavone methanol–water (70:30)
(Figure 12)
Juncein
Luteolin 4′ -O-β-D-glucopyranoside
(135) 4′ -O-β-D-glucopyranosyl-3′ ,4′ ,5,7- C21 H20 O11 448.380 Flavone Flowers/methanol–water (70:30) [132]
tetrahydroxyflavone
(Figure 12)
Luteolin 7-O-β-D-galactopyranoside
7-O-β-D-galactopyranosyl-3′ ,4′ ,5,7-
(136) C21 H20 O11 448.380 Flavone Flowers/methanol–water (70:30) [132]
tetrahydroxyflavone
(Figure 12)
Luteoloside
Luteolin 7-O-β-glucopyranoside
(137) 7-O-β-D-glucopyranosyl-3′ ,4′ ,5,7- C21 H20 O11 448.380 Flavone Flowers/methanol–water (70:30) [132]
tetrahydroxyflavone
(Figure 12)
6-Methoxy scutellarin
7-O-β-glucuronyl-4′ ,5,7-trihydroxy-6-
(138) C22 H20 O12 476.390 Flavone Leaves and stems/methanol [85]
methoxyflavone
(Figure 12)
Linaroside
7-O-β-D-glucopyranosyl-5,7-dihydroxy- Aerial parts/methanol
(139) C23 H24 O11 476.434 Flavone [16,124,134]
4′ ,6-dimethoxyflavone Leaves/methanol
(Figure 13)
Lantanoside
7-O-(6-O-acetyl-β-D-glucopyranosyl)-
(140) C25 H26 O12 518.471 Flavone Aerial parts/methanol [16,134]
5,7-dihydroxy-4′ ,6-dimethoxyflavone
(Figure 13)
Apigenin 7-O-β-D-galacturonyl-
(141) (2′′ →1′′′ )-O-β-D-galacturonide C27 H26 O17 622.488 Flavone Flowers/methanol–water (70:30) [132]
(Figure 13)
Luteolin 7-O-β-D-galacturonyl-
(142) (2′′ →1′′′ )-O-β-D-galacturonide C27 H26 O18 638.487 Flavone Flowers/methanol–water (70:30) [132]
(Figure 13)
Molecules 2025, 30, 851 18 of 51

Table 4. Cont.

Molecular Molecular Flavonoid


N.º Compound Part of the Plant/Solvent Reference
Formula Weight Type
Luteolin 7-O-β-D-glucuronyl-
(143) (2′′ →1′′′ )-O-β-D-glucuronide C27 H26 O18 638.487 Flavone Flowers/methanol–water (70:30) [132]
(Figure 13)
Acetyl lantanoside
7-O-(2,6-O-diacetyl-β-D-
(144) glucopyranosyl)-5,7-dihydroxy-4′ ,6- C27 H28 O13 560.508 Flavone HD * [16,134]
dimethoxyflavone
(Figure 13)
Acacetin-7-O-β-D-rutinoside
7-O-β-D-rutinosyl-5,7-dihydroxy-
(145) C28 H32 O14 592.550 Flavone Leaves/methanol [79]
4′ -methoxyflavone
(Figure 13)
Pectolinarin
7-O-β-D-rutinosyl Aerial parts/ethanol
(146) C29 H34 O15 622.576 Flavone [19,84,133]
-5,7-dihydroxy-4′ ,6-dimethoxyflavone Leaves/ethanol, methanol
(Figure 13)
3,7-O-Dimethylquercetin
(147) 3′ ,4′ ,5-trihydroxy-3,7-dimethoxyflavone C17 H14 O7 330.292 Flavonol Leaves/acetone [127]
(Figure 13)
3,5,7,8-Tetrahydroxy-3′ ,6-
(148) dimethoxyflavone C17 H14 O8 346.291 Flavonol Leaves/methanol [79]
(Figure 13)
6-Methoxykaempferol-7-O-β-D-
glucoside
(149) 7-O-β-D-glucopyranosyl-3,4′ ,5,7- C22 H22 O12 478.406 Flavonol Flowers/95% methanol [135]
tetrahydroxy-6-methoxyflavone
(Figure 13)
5,7-Dihydroxy-6,3′ ,
(150) 4′ -trimethoxy isoflavone C18 H15 O7 343.311 Isoflavone Leaves/methanol [79]
(Figure 13)
Gautin
5,7-dihydroxy-6,3′ ,4′ -trimethoxy
isoflavone-5-O-α-L-rhamnopyranosyl-7-
(151) C34 H42 O19 754.691 Isoflavone Leaves/methanol [79]
O-β-D-arabinopyranosyl-(1′′′ →4′′ )-O-β-
D-xylopyranoside
(Figure 13)
* HD: semisynthetic derivative.

Table 5. Iridoid glucosides isolated from non-volatile fractions of Lantana camara L.

Molecular Molecular
N.º Compound Part of the Plant/Solvent Reference
Formula Weight
Geniposide
methyl (1S,4aS,7aS)-7-(hydroxymethyl)-1- Leaves and stems/
[(2S,3R,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl) methanol
(152) C17 H24 O10 388.369 [91]
oxan-2-yl] oxy-1,4a,5,7a-tetrahydrocyclopenta[c]pyran- Roots/
4-carboxylate ethanol
(Figure 14)
Theviridoside Aerial parts and roots/
methyl (1S,4aR,7aR)-4a-hydroxy-7-(hydroxymethyl)-1- ethanol
[(2S,3R,4S,5S,6R)-3,4,5-trihydroxy-6-(hydroxymethyl) Leaves and stems/
(153) C17 H24 O11 404.368 [91]
oxan-2-yl] methanol
oxy-5,7a-dihydro-1H-cyclopenta[c]pyran-4-carboxylate Roots/
(Figure 14) methanol
Molecules 2025, 30, 851 19 of 51

Table 6. Fatty acids isolated from non-volatile fractions of Lantana camara L.

Molecular Molecular
N.º Compound Part of the Plant/Solvent References
Formula Weight
Myristic acid
(154) C14 H28 O2 228.376 Aerial parts/petroleum ether [73]
tetradecanoic acid
Palmitic acid Aerial parts/methanol/petroleum ether
(155) C16 H32 O2 256.430 [73,77,82]
hexadecanoic acid Stems/ethanol
Linoleic acid
(156) C18 H32 O2 280.452 Aerial parts/petroleum ether [73]
(9Z,12Z)-octadeca-9,12-dienoic acid
Oleic acid
(157) C18 H34 O2 282.468 Aerial parts/petroleum ether [73]
(9Z)-octadec-9-enoic acid
Stearic acid Aerial parts/methanol
(158) C18 H36 O2 284.484 [77,82]
octadecanoic acid Stems/ethanol
Arachidic acid
(159) eicosanoid acid C20 H40 O2 312.538 Aerial parts/petroleum ether [73]
(Figure 14)
Behenic acid
(160) docosanoic acid C22 H44 O2 340.592 Aerial parts/methanol [77,82]
(Figure 14)
Lignoceric acid
(161) tetracosanoic acid C24 H48 O2 368.646 Aerial parts/methanol [106]
(Figure 14)
Lacceroic acid
(162) dotriacontanoic acid C32 H64 O2 480.862 Aerial parts/methanol [106]
(Figure 14)

Table 7. Other compounds isolated from non-volatile fractions of Lantana camara L.

Molecular Molecular
N.º Compound Part of the Plant/Solvent References
Formula Weight
Ethyl-β-D-galactopyranoside Stems/
(163) C8 H16 O6 208.210 [15]
(Figure 14) ethanol
Linamarin
Leaves and stems/
(164) 2-(β-D-Glucopyranosyloxy)-2-methylpropanenitrile C10 H17 NO6 247.247 [99]
methanol
(Figure 14)
Phytol
Leaves and stems/
(165) 3,7,11,15-tetramethyl-2-hexadecen-1-ol C20 H40 O 296.539 [75]
petroleum ether
(Figure 14)
Di-(2-ethylhexyl) phthalate Fruits/
(166) C24 H38 O4 390.564 [73]
(Figure 14) chloroform
Triacontan-1-ol Aerial parts/
(167) C30 H62 O 438.825 [73]
(Figure 14) petroleum ether
Trilinolein
Fruits/
(168) Glyceryl trilinoleate C56 H96 O6 865.37 [73]
chloroform
(Figure 14))

The chemical structures of steroids, triterpenes, flavonoids, iridoid glycosides, fatty


acids, and miscellaneous compounds isolated from L. camara or obtained by semisynthe-
sis are depicted in Figures 4–14. The structures of very well-known compounds have
been omitted.
Molecules 2025, 30, 851 Molecules 2025, 30, x FOR PEER REVIEW 20 of 51 26 of 61

Molecules 2025, 30, x FOR PEER REVIEW 27 of 61

Figure 4. StructuresFigure 4. Structures


of compounds 3, of
andcompounds
6–15. 3, and 6–15.

Figure
Figure 5. Structures of 5. Structures
compounds 16, of
17,compounds 16, 17, and 19–30.
and 19–30.
Molecules 2025, 30,
Molecules 2025, 30, 851
x FOR PEER REVIEW 28
21of 61
of 51

Figure 6.
Figure 6. Structures
Structures of
of compounds
compounds 31–47.
31–47.
Molecules 2025,
Molecules 30, x851
2025, 30, FOR PEER REVIEW 2922ofof 61
51

Figure 7. Structures
Structures of compounds 48–74.
Molecules 2025,30,
Molecules 2025, 30,851
x FOR PEER REVIEW 30
23 of 61
of 51

Figure 8.
Figure 8. Structures
Structures of
of compounds
compounds 75–90.
75–90.
Molecules 2025,
Molecules 2025, 30,
30, 851
x FOR PEER REVIEW 31
24of 61
of 51

Figure 9. Structures of compounds 91–96.


Molecules 2025, 30, 851
x FOR PEER REVIEW 3225of
of 61
51

O
12 O 28 12
11 13 R1 11 R2
H H COOH H COOR3

H H O H
O O HO
R1 24
(97)
(98) R = CHO (99) R1 = O; R2 = H; R3 = H
(100) R = H (102) R1 = H,H; R2 = H; R3 = H
(105) R1 = H,H; R2 = OH; R3 = H
(110) R1 = O; R2 = H; R3 = Me

OH
19
12
R2 O
H COOH H H COOR2

H H H
1
R HO R1
HOOC
24
(101) R1 = O; R2 = OH (103) (104) R1= O; R2 = H
(106) R1 = OH, H; R2 = H (107) R1 = OH, H; R2 = H
(109) R1 = OH, H; R2 = H; 12,13-dihydro
(113) R1 = OH, H;R2 = Me; 12,13-dihydro

12 12 22
OR1
2 HO 25 H COOR2
H COOR H COOMe 28
O H O H
H
R1O HO
1 2
(111 ) R = Me; R = H (112) (114) R1 = Ac; R2 = H
(115) R1 = Me; R2 = Me (117) R1 = Ac; R2 = Me
(116) R1 = Et; R2 = H
(118) R1 = Et; R2 = Me

OH OH

19 19
12 12
OR
H COOH H COOH

H H
HO RO

(119) R = 3-metylbut-2-enoyl (120) R = isovaleryl

Figure
Figure 10.
10. Structures
Structures of
of compounds 97–107 and
compounds 97–107 and 109–120.
109–120.
Molecules
Molecules 2025, 30,
2025, 30,
Molecules 2025, x
x FOR
FOR PEER
30, 851 PEER REVIEW
REVIEW 33
33 of 61
26 of 61
of 51

Figure 11.
Figure 11. Structures
11. Structures of
Structures of compounds
of compounds 121–127.
compounds 121–127.
121–127.
Figure

Figure
Figure 12. of
12. Structures of
Figure 12. compounds
of compounds 128–138.
compounds 128–138.
128–138.
Molecules2025,
Molecules 2025,30,
30,851
x FOR PEER REVIEW 34
27 of 61
of 51

Figure13.
Figure 13. Structures
Structuresof
ofcompounds
compounds139–151.
139–151.
Molecules 2025,30,
Molecules2025, 30,851
x FOR PEER REVIEW 35
28 of 51
61

Figure 14.
Figure 14. Structures
Structures of
of compounds
compounds 152,
152, 153,
153,and
and159–168.
159–168.

3.3. Biological
3.3. Biological Activities
Activities
A great
A great variety
variety of of biological
biological effects
effectsexerted
exertedby byseveral
severalextracts
extractsofofdifferent
different parts
parts ofof
L.
camara
L. camara have
havebeen
been tested
testedin vitro and,
in vitro and,moremore rarely, alsoalso
rarely, in vivo
in vivo(Table
(Table8). The most
8). The relevant
most rele-
vant biological properties included significant anti-inflammatory and analgesic effectsleaf
biological properties included significant anti-inflammatory and analgesic effects of a of
aaqueous extractextract
leaf aqueous [136]; [136];
a moderate antibacterial
a moderate activity
antibacterial of leafof
activity alcoholic and aqueous
leaf alcoholic and aque- ex-
tracts
ous againstagainst
extracts Escherichia coli, Proteus
Escherichia vulgaris,
coli, Proteus and Vibrio
vulgaris, andcholerae [121], and
Vibrio cholerae against
[121], Bacillus
and against
subtilis,subtilis,
Bacillus Klebsiella pneumoniae,
Klebsiella Staphylococcus
pneumoniae, Staphylococcusaureus, and Pseudomonas
aureus, and Pseudomonas aeruginosa [137];
aeruginosa the
[137];
inhibitory properties of a 70% aqueous ethanolic flower extract
the inhibitory properties of a 70% aqueous ethanolic flower extract against the growth against the growth of the
Mycobacterium
of the Mycobacterium tuberculosis H37RVH37RV
tuberculosis strain [138];
strain the antidiabetic
[138]; effects effects
the antidiabetic of an aqueous
of an aque- leaf
extract [125]; the moderate antidiarrheal effects of an ethanol leaf
ous leaf extract [125]; the moderate antidiarrheal effects of an ethanol leaf extract [100]; extract [100]; the signif-
icant
the antioxidant
significant properties
antioxidant of methanol
properties leaf and leaf
of methanol flowerandextracts [139]; the
flower extracts highthe
[139]; in high
vitro
antiparasitic activity of dichloromethane and methanol extracts
in vitro antiparasitic activity of dichloromethane and methanol extracts of the leaves/aerial of the leaves/aerial parts
against
parts Leishmania
against amazonensis
Leishmania promastigotes
amazonensis promastigotes [64,67,109], and dichloromethane
[64,67,109], and dichloromethane and ethyl
and
acetate leaf extracts against the chloroquine-sensitive strains
ethyl acetate leaf extracts against the chloroquine-sensitive strains 3D7 and D10, and the3D7 and D10, and the chlo-
roquine-resistant strain
chloroquine-resistant W2, W2,
strain of Plasmodium
of Plasmodium falciparum [43,58,140];
falciparum the potent
[43,58,140]; in vitro
the potent in nem-
vitro
atocidal activity of a methanol extract of the aerial parts and its
nematocidal activity of a methanol extract of the aerial parts and its partitions against thepartitions against the lar-
vae of of
larvae thethe
root-knot
root-knotnematode
nematodeMeloidogyne
Meloidogyneincognita;
incognita;the thesignificant
significant in in vitro
vitro anti-COVID-
anti-COVID-
19 activity
19 activity of
of 95%
95% ethanol
ethanol extracts
extracts ofof the
the leaves
leaves and and flowers
flowers from
from different
different cultivars
cultivars [103];
[103];
the significant
the significantcancer
cancerreduction
reductionand andincreased
increasedsurvival
survival rate
rate of of
micemice exhibited
exhibited byby a metha-
a methanol
nol leaf
leaf extract
extract [141];[141];
thethe
highhigh in vitro
in vitro cytotoxicity
cytotoxicity ofof differentleaf
different leafextracts
extracts[43,58,142];
[43,58,142]; the the
DNA-protective effects
DNA-protective effects of of an
an aqueous
aqueous leaf leaf extract
extract [143];
[143]; the
thesignificant
significant hepatoprotective
hepatoprotective
effects of
effects ofaamethanol
methanolleaf leafextract
extract[130];
[130]; thethehighhigh insecticidal,
insecticidal, larvicidal,
larvicidal, andand termiticidal
termiticidal ac-
activities of different extracts, especially of the leaves in polar solvents, against several
Molecules 2025, 30, 851 29 of 51

tivities of different extracts, especially of the leaves in polar solvents, against several insects
(mosquitos, moths, termites, weevils, and bugs) [52,59,86,144–153]; the wound-healing
effects of ethanol and water leaf extracts [108,154].
In summary, the alcoholic and aqueous leaf extracts seem to exhibit the highest and
widest biological properties. In our opinion, the most promising biological effects of the
extracts, which have attracted the greatest interest from several research groups, are the
antiparasitic, nematocidal, and insecticidal properties.
Concerning the bioactivities of the compounds isolated from L. camara and the semisyn-
thetic derivatives (Table 9), potent nematocidal effects against Meloidogyne incognita larvae
(mortality > 80%) were shown by different oleanane triterpenoids, such as camaric acid (62),
camarin (23), camarinin (56), lantanilic acid (68), lantanolic acid (27), the ursane triter-
penoids camarinic acid (114), lantacin (119), lantic acid (102), lantoic acid (105), pomolic
acid (107), and ursolic acid (106), and the flavonoids linaroside (139) and lantanoside (140).
Other interesting properties were the in vitro antiparasitic activity (IC50 < 10 µM) to-
wards Leishmania mexicana promastigotes exhibited by camaric acid (62) and lantanilic
acid (68). On the other hand, the in vitro cytotoxicity of most triterpenoids towards dif-
ferent human tumor cell lines was from moderate to very weak (IC50 = 20–80 µM) or null
(IC50 > 100 µM), except for the high activity (IC50 < 10 µM) exhibited by camaric
acid (62), lantacamaric acid B (70), and lantanilic acid (68) towards HL-60 (JCRB0085)
leukemia cells, icterogenin (67) towards colon cancer HCT-116 and lymphocytic leukemia
L1210 cells, lantadene B (61) towards lung carcinoma A549 cells, oleanolic acid (31) towards
drug-resistant human ovarian carcinoma IGROV-1 cells, and oleanonic acid (22) towards
leukemia HL-60 and Ehrlich ascites carcinoma (EAC) cells. However, the in vivo antitumor
activity, measured by the percent mice survival and percent overall papilloma incidence,
was observed only for large doses of the administered compound, such as, for example, the
ester 78. Interesting in vivo antidiabetic effects were exhibited by urs-12-en-3β-ol-28-oic
acid 3-O-β-D-glucopyranosyl-4′ -octadecanoate (127). In vitro high antibacterial activity
against the Mycobacterium tuberculosis strain H37Rv was exhibited by acetyl lantanoside
(144). Powerful in vitro protein tyrosine phosphatase inhibition effects (IC50 < 10 µM) were
determined for camaric acid (62), di(2-ethylhexyl) phthalate (166), 24-hydroxylantadene B
(65), 22β-hydroxy-oleanolic acid (32), 22β-hydroxy-oleanonic acid (25), lantadenes A (71),
B (62) and D (54), lantanilic acid (68), oleanolic acid (31), oleanonic acid (22), and reduced
lantadenes A (75), B (76), and C (77). The in vitro anti-inflammatory activity was de-
termined by measuring the inhibition of the following two inflammatory mechanisms:
22β-hydroxy-oleanonic acid (26) and lantadene A (60) and B (61) strongly inhibited the
TNF-α-induced NF-KB activation (IC50 < 10 µM), but they were ineffective (IC50 > 100 µM)
against cyclooxygenase-2 (COX-2).
L. camara is also known for the toxicity to animals causing hepatotoxicity, photosensiti-
zation, and jaundice. Lantadene A (60) is the main toxic pentacyclic triterpenoid present in
this weed.
Table 8. Biological activities of different extracts of Lantana camara L. a, *.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
The anti-inflammatory activity assay
was carried out using
Significant (p < 0.05)
Analgesic, carrageenan-induced lung edema and
Leaves/water anti-inflammatory and analgesic [136]
anti-inflammatory pleurisy mice. An analgesic effect
activity, and minimal toxic effects.
assay was carried out using the
formalin pain test.
Molecules 2025, 30, 851 30 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
The in vitro antibacterial activity of a
crude extract was screened at
concentrations of 1000 µg/mL and
500 µg/mL against Bacillus cereus var
mycoides (ATCC 11778), B. pumilus
(ATCC 14884), B. subtilis (ATCC 6633), Except for E. coli and P. aeruginosa,
Leaves/
Bordetella bronchiseptica (ATCC 4617), a complete inhibition of bacterial
dichloromethane–methanol [60]
Micrococcus luteus (ATCC 9341), growth was observed at
(1:1)
Staphylococcus aureus (ATCC 29737), both concentrations.
S. epidermidis (ATCC 12228),
Escherichia coli (ATCC 10536),
Klebsiella pneumoniae (ATCC 10031),
Pseudomonas aeruginosa (ATCC 9027),
and Streptococcus faecalis (MTCC 8043).
The in vitro antibacterial activity of a
B. cereus and M. fortuitum: MIC
crude extract was screened against
Leaves, stems, and MBC values > 1000 µg/mL;
Bacillus cereus (ATCC 14579), [44]
and roots/methanol S. aureus: MIC = 250 µg/mL,
Mycobacterium fortuitum (ATCC 6841),
MBC > 1000 µg/mL.
and Staphylococcus aureus (ATCC 6538).
A crude extract was tested in vitro
Inhibition zone
with the disk diffusion method against
Leaves/methanol diameter [121]
Escherichia coli, Proteus vulgaris, and
= 50 mm.
Vibrio cholerae.
MIC values (mg/mL) of a
methanol extract: B. subtilis and
K. pneumoneae = 8; S. aureus and
Crude extracts were tested against
P. aeruginosa = 5.
Bacillus subtilis (ATCC 6059),
MIC values (mg/mL) of an ethanol
Leaves/ Klebsiella pneumoniae,
extract: B. subtilis = 10, [137]
methanol, ethanol, water Staphylococcus aureus (ATCC 6538),
Antibacterial K. pneumoneae = 12, S. aureus = 6.5,
and Pseudomonas aeruginosa
P. aeruginosa = 8.
(ATCC 7221).
MIC values (mg/mL) of an
aqueous extract: B. subtilis and
S. aureus = 8, P. aeruginosa = 10.
A crude extract was tested against the
All concentrations inhibited the
Flowers/ Mycobacterium tuberculosis H37RV
growth of M. tuberculosis H37RV [138]
70% aqueous ethanol strain at different concentrations
from the first to the sixth week.
(25, 50, and 100 µg/mL).
MIC values (µg/mL) determined
Crude extracts were tested against the
for M. smegmatis mc2155
Mycobacterium smegmatis mc2155 strain,
strain = 574 ± 196; M. tuberculosis
Leaves/methanol M. tuberculosis H37Rv strain, and [155]
H37Rv strain = 574 ± 196;
rifampicin-resistant M. tuberculosis
M. tuberculosis TMC-331
TMC-331 strain.
strain = 176 ± 33.
The in vitro antibacterial activity of a
crude extract was tested against E. coli MIC values determined for all
Aerial parts/methanol [156]
(ATCC25922), Klebsiella pneumoniae, tested bacteria = 25 µg/mL.
Pantoea sp., and Shigella flexneri.
A crude extract was tested
Inhibition zone
Leaves/methanol with the disk diffusion method against [128]
diameter = 20 mm.
Helicobacter pylori.
Crude flower and leaf extracts showed
the highest inhibitory effects against
Inhibition area ranging from
B. subtillis.
Leaves and flowers/water, 6 to 9 mm.
Extracts separated by column [157]
methanol, acetone, benzene Inhibition area ranging from
chromatography displayed weaker
3 to 7 mm.
inhibitory effects against B. subtillis
than crude extracts.
Molecules 2025, 30, 851 31 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
The concentration of 1 mg/mL
exhibited the highest anticoagulant
The in vitro anticoagulant activity of
activity; values expressed as
Leaves, flowers, and crude extracts was tested at
Anticoagulant prothrombin time: [158]
roots/70% aqueous ethanol concentrations of 0.125, 0.25, 0.50, and
flowers = 21.7 ± 3 s;
1 mg/mL.
leaves = 16.9 ± 2.4 s;
roots = 21.5 ± 2.8 s.
Blood glucose levels:
dose 1: at the 8th
day = 183.83 ± 4.29 mg/dL; at the
Wistar albino rats (150–200 g);
14th day = 165.50 ± 4.26 mg/dL; at
dose 1 = 250 mg extract/kg body
the 21st day = 136.83 ± 1.99 mg/dL.
Antidiabetic Leaves/water weight and dose 2 = [125]
Dose 2: at the 8th
500 mg extract/kg body weight were
day = 180.50 ± 3.07 mg/dL; at the
administered orally for 21 days.
14th day = 157.83 ± 5.28 mg/dL;
at the 21st
day = 124.67 ± 2.40 mg/dL.
% Intestinal transit:
Group II of male mice (Laca strain;
group II: 34.78 ± 3.52; group III:
20–25 g)
1 ± 0.01; group IV: 26.46 ± 6.82;
received 1% leaf powder for ten days;
group V: 31.74 ± 1.49; group VI:
Leaves/ethanol groups III–VI received a single dose of [100]
38.67 ± 6.60. Mean defecation in
125, 250, 500, and 1000 mg extract/kg
4 h: group III: 9 ± 1.18; group IV:
body weight; subsequently, castor
9 ± 2.06; group V: 1 ± 0.05;
Antidiarrheal oil-induced diarrhea was evaluated.
group VI: total constipation.
Groups III–V of Swiss albino mice
(6–88 weeks; 20–30 g) received a single The most effective dose was
Leaves/80% aqueous dose of 100, 200, and 400 mg 400 mg/kg body weight with a
[159]
methanol extract/kg body weight; subsequently, 67.9% inhibition of diarrhea and an
castor oil-induced diarrhea antidiarrheal index of 87.6.
was evaluated.
The antifungal activity of a crude
extract was tested at concentrations of
Leaves/dicloromethane– 1000 µg/mL and 500 µg/mL against Complete inhibition was observed
[60]
methanol (1:1) Candida albicans (MTCC 10231), at both concentrations.
Aspergillus niger (MTCC 1344), and
Saccharomyces cerevisiae (ATCC 9763).
The in vitro antifungal activity of a
Leaves, stems, and MIC and MBC values >
crude extract was tested against [44]
roots/methanol 1000 µg/mL.
Candida albicans (ATCC 10231).
Antifungal
A crude extract was tested in vitro
Inhibition zone
Leaves/methanol with the disk diffusion method against [79]
diameter = 0.5 mm.
Aspergillus niger and Candida albicans.
% Inhibition of the methanol
Crude extracts were tested against extract: A. fumigatus = 71.4;
Leaves/methanol, water Aspergillus fumigatus and A. flavus = 66.4. [137]
A. flavus. % Inhibition of the water extract:
A. fumigatus = 61.5; A. flavus = 57.8.
Molecules 2025, 30, 851 32 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
Leaves:
total phenols > 100 mg/g
extract; DPPH assay:
IC50 = 16.02 ± 0.04 µg/mL;
xanthine oxidase inhibition assay:
IC50 < 20 µg/mL; Griess–Ilosvay
method: IC50 < 10 ± 2 µg/mL.
Flowers:
total phenols > 100 mg/g
extract; DPPH assay:
IC50 = 28.92 ± 0.19 µg/mL;
Phytochemical analysis: xanthine oxidase inhibition assay:
Folin–Ciocalteu assay; gallic acid and IC50 > 20 µg/mL; Griess–Ilosvay
ascorbic acid as reference standard. method: IC50 < 10 ± 2 µg/mL.
The in vitro antioxidant activity was Fruits:
tested by the following total phenols < 100 mg/g
different methods: extract; DPPH assay:
Leaves, flowers, fruits, roots,
1,1-diphenyl-2-picrylhydrazyl (DPPH) IC50 = 90.11 ± 0.57 µg/mL; [139]
and stems/methanol
radical scavenging assay, with ascorbic xanthine oxidase inhibition assay:
acid as a reference standard; xanthine IC50 > 20 µg/mL; Griess–Ilosvay
oxidase inhibition assay, with method: IC50 > 10 ± 2 µg/mL.
allopurinol as a reference standard; Roots:
Griess–Ilosvay method, with total phenols > 100 mg/g
allopurinol as a reference standard. extract; DPPH assay:
IC50 = 31.52 ± 0.74 µg/mL;
xanthine oxidase inhibition assay:
IC50 > 20 µg/mL; Griess–Ilosvay
method: IC50 < 10 ± 2 µg/mL.
Stems:
total phenols > 100 mg/g
extract; DPPH assay:
IC50 = 46.96 ± 2.51 µg/mL;
xanthine oxidase inhibition assay:
IC50 < 20 µg/mL; Griess–Ilosvay
method: IC50 > 10 ± 2 µg/mL.
Antioxidant
Leaves:
total phenols: 227.10 ± 9.07; Gallic
Acid Equivalents (GAE µg/mg),
22.7%; total flavonoids:
46.55 ± 1.50; quercetin equivalents
(QuercE µg/mg), 4.6%; caffeic acid
(10.75 ± 0.04 mg/g), 1.07%;
quercetin (2.87 ± 0.01 mg/g,
0.28%); TBARS assay: basal
IC50 = 57.69 ± 4.01 µg/mL;
Phytochemical analyses: induced Fe2+ IC50 = 32.48 ± 3.51
Folin–Ciocalteu assay, with gallic acid µg/mL; iron chelation assay:
as a reference standard; aluminum IC50 = 214.20 ± 2.50 µg/mL;
chloride method, with quercetin as a deoxyribose degradation assay:
reference standard; quantification of IC50 = 285.64 ± 20.63 µg/mL;
phenolics and flavonoids by FRAP assay: 8.28 ± 0.07 mM
HPLC-DAD. In vitro antioxidant Fe 2+ /g extract.
Leaves and roots/ethanol [160]
activity was determined by the Roots:
following different methods: total phenols = 211.80 ± 7.94
thiobarbituric acid reactive substances (GAE µg/mg), 21.3%; total
(TBARS) assay with phospholipids; flavonoids = 33.64 ± 1.52 (QuercE
iron chelation assay; deoxyribose µg/mg, 3.3%); caffeic acid
degradation assay; ferric-reducing (8.27 ± 0.01 mg/g, 0.82%), rutin
antioxidant power (FRAP). (5.35 ± 0.03 mg/g, 0.53%);
TBARS assay: basal
IC50 = 168.92 ± 7.36 µg/mL;
induced Fe2+
IC50 = 63.84 ± 4.56 µg/mL;
iron chelation assay:
IC50 = 448.19 ± 4.50 µg/mL;
deoxyribose degradation assay:
IC50 = 276.89 ± 31.26 µg/mL;
FRAP assay: 11.64 ± 0.10 mM
Fe2+ /g extract.
Molecules 2025, 30, 851 33 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
The concentration of lipid peroxides
(LPOs) in the stomach mucosa of
LPO: 29.23 ± 0.35 and
Wister albino rats used in ulcerogenic
27.7 ± 0.50 nmol/g;
Leaves/methanol models was indirectly measured by the [128]
GSH: 181.52 ± 0.83
TBARS assay; the concentration of
and 202.9 ± 1.08 µg/g.
reduced glutathione was
also determined.
Leaves/methanol DPPH assay. IC50 = 74.3 µg/mL. [130]
Total phenolic content:
40.859 ± 0.017 (mg GAE/g
dry sample).
Quantitative analysis of
Total flavonoid content:
phytochemicals: total phenolic content,
53.112 ± 0.199 (mg rutin/g
total flavonoid content, and total
dry sample).
Leaves/methanol tannin content. In vitro antioxidant [137]
Total tannin content: 0.860 ± 0.038
activity (DPPH radical scavenging
(mg/g dry sample).
activity assay and hydroxyl radical
Antioxidant DPPH assay:
scavenging activity assay).
IC50 ≥ 0.2 mg/mL.
Hydroxyl radical assay:
IC50 ≤ 0.2 mg/mL.
Total phenolic content:
Total phenolic content
Leaves/ethyl acetate 2419.6 mg/L GAE. DPPH assay: [161]
and DPPH assay.
IC50 = 36.18 mg/mL.
The antioxidant capacity of
FRAP: 8.17 ± 0.04 mmol/g, DPPH:
Aerial parts/methanol a crude extract was evaluated by the [156]
IC50 = 16.13 ± 0.35 µg/mL.
FRAP and the DPPH assays.
DPPH assay: IC50 = 42.66 µg/mL;
DPPH, metal chelating activity, and metal chelating activity assay:
Leaves/water [162]
FRAP assays. IC50 = 1036.4 µg/mL; FRAP test:
dose-dependent activity.
DPPH: IC50 = 24.80 ± 0.52 µg/mL;
Leaves/methanol DPPH and FRAP assays. [143]
FRAP: IC50 = 21.61 ± 0.26 µg/mL.
Dichlorometane extract:
Leaves and IC50 = 11.7 ± 4.4 µg/mL and IC50 :
Leishmania amazonensis
stems/dichloromethane; 21.8 ± 2.4 µg/mL;
(MHOM/77BR/LTB0016) [64]
dichloromethane–methanol dichloromethane–methanol (1:1)
promastigotes and amastigotes.
(1:1); water and water extracts:
IC50 > 200 µg/mL.
Leishmania amazonensis IC50 = 14 µg/mL and
(MHOM/Br/75/Josefa) isolated IC50 > 250 µg/mL.
promastigotes and L. chagasi
(MHOM/Br/74/PP75) [67]
Leaves/methanol
isolated promastigotes.
Crude extract: IC50 = 21.8 ± 2.4 µM
The in vitro antiparasitic activity of a and IC50 = 42.6 ± 1.9 µM. The
crude extract and 18 fractions obtained most active fractions against
by open-column chromatographic L. amazonensis amastigotes
Aerial
separation were tested against were as follows: FII,
parts/dichloromethane
Antiparasitic Leishmania amazonensis IC50 = 9.1 ± 3.4 µM; FX, [109]
(MHOM/BR/77/LTB0016) IC50 = 7.9 ± 0.3 µM; FXI,
promastigotes and L. mexicana IC50 = 8.0 ± 1.1 µM; FXVI,
isolated amastigotes. IC50 = 8.5 ± 1.7 µM.
Dichloromethane extracts of two
different batches were tested against
the chloroquine-sensitive strain 3D7 3D7: IC50 = 8.7 ± 1 and
and chloroquine-resistant strain W2 of 14.1 ± 8.4 µg/mL; W2:
Plasmodium falciparum, and by the IC50 = 5.7 ± 1.6 and
parasite dehydrogenase lactate essay. 12.2 ± 2.9 µg/mL.
Leaves/dichloromethane [43]
Five female Swiss mice (10 weeks old;
25 ± 2 g), infected by
Plasmodium berghei NK173, received a
single dose of 50 mg extract/kg 5% growth inhibition.
body weight daily for
4 days intraperitoneally.
Molecules 2025, 30, 851 34 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
The extract was tested on
3D7: IC50 = 19 ± 0.57 µg/mL;
Leaves/ethyl acetate chloroquine-resistant strains (3D7 and [58]
INDO: IC50 = 20 ± 1.5 µg/mL.
INDO) of Plasmodium falciparum.
Crude extracts were tested in vitro
Leaves and against the chloroquine-sensitive strain
IC50 = 11 µg/mL;
twigs/dichloromethane– D10 of Plasmodium falciparum and by [140]
IC50 < 1000 µg/mL.
methanol (1:1); water the parasite dehydrogenase
lactate assay.
Antiparasitic
At a 0.5% concentration, the
methanol extract: 85% mortality;
The in vitro nematocidal activity of ether insoluble partition:
crude extract and its partitions were 90% mortality; methanolic acidic
Aerial parts/methanol screened against Meloidogyne incognita partition: 88% mortality; ether [99]
larvae, at concentrations of 0.5%, soluble partition: 75% mortality;
0.25%, and 0.125% after 48 h. n-hexane soluble partition:
60% mortality; n-hexane insoluble
partition: 50% mortality.
Wister albino rats (150–200 g) were
divided into 4 groups; groups 2 and 3
received 250 and 500 mg extract/kg Ulcer index inhibition: APL:
orally. Aspirin-induced ulcerogenesis 46.61% and 73.97%; EIM: 55.60%
Antiulcerogenic Leaves/methanol [103]
in a pyloric ligated system (APL); and 63.39%; CYS: 41.43%
ethanol-induced ulcer model (EIM); and 68.90%.
cysteamine-induced duodenal ulcer
model (Cys).
Spreading sunset cultivar: flowers:
IC50 = 4.188 µg/mL; leaves:
IC50 = 8.751 µg/mL. Chelsea gem
cultivar: flowers:
The in vitro anti-COVID-19 activity of IC50 = 3.671 µg/mL; leaves:
Leaves and flowers/ crude extracts from different cultivars IC50 = 3.181 µg/mL. Nivea
Antiviral [103]
95% ethanol were screened by the plaque cultivar: flowers:
reduction assay. IC50 = 15.050 µg/mL; leaves:
IC50 = 6.820 µg/mL. Drap d’or
cultivar: flowers:
IC50 = 5.015 µg/mL; leaves:
IC50 = 8.715 µg/mL.
A dose-dependent effect, with
higher doses of UASG (25 and
50 mg/kg) leading to more
Ursolic acid stearyl glucoside (UASG)
pronounced anxiolytic effects.
was isolated from the leaves of L.
UASG reduced the anxiety and
camara using column chromatography.
increased the locomotor activity.
Anxiolytic Leaves/methanol The compound was administered to [163]
The anxiolytic effects of UASG
the animals in a dose-dependent
were comparable to those of
manner to evaluate its effects at
diazepam (1 mg/kg), a standard
different concentrations
anxiolytic drug, indicating that
UASG may have a similar
therapeutic potential.
Female Swiss albino mice (6 weeks old;
18–22 g). Group III received 400 mg
extract/kg body weight, which was A significant reduction in cancer
Chemoprotective
Leaves/methanol given orally as a suspension in water (76.4%) was observed, and the [141]
effect
and carboxymethyl cellulose, twice a survival rate was 75%.
week (100 nmol/100 µL), applied for
20 weeks topically.
Leaves and
(a): CC50 > 100 µg/mL;
stems/dichloromethane (a);
BALB/c mice peritoneal macrophages. (b): CC50 > 200 µg/mL; [64]
dichloromethane–methanol
(c): CC50 = 125.9 ± 3.1 µg/mL.
(1:1) (b); water (c).
Cytotoxic
The cytotoxicity of dichloromethane
extracts from two different batches was IC50 = 69.5 ± 12.1 µg/mL and IC50
Leaves/dichloromethane [43]
tested in vitro towards normal human = 97.2 ± 2.4 µg/mL.
fetal lung fibroblasts WI-38.
Molecules 2025, 30, 851 35 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
Leaves/ethyl acetate HeLa cells and the MTT assay. IC50 = 42 ± 2.3 µg/mL. [58]
Leaves/ethanol Tested towards Hela cancer cells. IC50 = 43.54 µg/mL. [142]
Cytotoxic IC50 values at 24 h
exposure = 361.44 ± 10.68 µg/mL;
Leaves/methanol Vero cells. [103]
at 48 h
exposure = 319.37 ± 99.80 µg/mL.
H2 O2 photolysis by UV radiation in Treatment with the extract at the
DNA protection Leaves/water the presence of pBR322 plasmid DNA evaluated dose completely [162]
and an aqueous extract (50 g). protected the plasmid DNA.
The hemolytic activity of a
CC50 values (µg/mL): aqueous
crude extract
extract = 8035.9; hexane–ethyl
and the hexane–ethyl acetate (50:50),
acetate (50:50) phase = 4470.4;
Hemolytic Leaves/water chloroform, methanolic, and ethanolic [107]
chloroform phase = 2739.8;
partitions were screened at different
methanolic phase = 12332.0;
concentrations (125, 250, 500, and
ethanolic phase = 9496.4.
1000 µg/mL).
In vivo acetaminophen-induced
hepatotoxicity on a mice model. The
mice of groups III and IV received a
dose of 25 and 75 mg extract/kg daily
for 7 days before receiving a single Among all of the tested groups,
dose of acetaminophen. The mice of pretreatment with a 75 mg
groups V and VI received a dose of extract/kg significantly reduced
Leaves/methanol 25 and 75 mg extract/kg daily for the SGOT = 144.5 ± 3.74 (UI/L), [130]
7 days before receiving a single dose of SGPT = 112.4 ± 9.1 (UI/L), and
acetaminophen. Subsequently, the ALP = 96.8 ± 3.2 (UI/L) activities
serum glutamate oxaloacetate compared to control groups.
transaminase (SGOT), serum
glutamate pyruvate transaminase
(SGPT), and alkaline phosphatase
(ALP) activities were measured.

Hepatoprotective A Ginkgo biloba methanolic extract


effect (GBME) was evaluated against
lantadenes-induced hepatic damage in
guinea pigs. Guinea pigs (200–250 g)
were divided into 5 groups. Group I:
negative control; group II received
25 mg lantadenes/kg body weight;
group III received 25 mg
Leaves/methanol Serum protein levels of group IV
lantadenes/kg body weight + 100 mg
(Lantadenes concentrated were significantly lower than [164]
GBME/kg body weight; group IV
fraction) group II.
received 25 mg lantadenes/kg body
weight + 200 mg GBME/kg body
weight; group V: positive control,
received 100 mg silymarin/kg body
weight. The drugs were administered
orally in gelatin capsules daily for
14 days. Analysis by HPLC of the
lantadenes fraction (72.82% lantadene A).
The methanol extract was more
Methanol and n-hexane extracts were active than the n-hexane extract.
Leaves/methanol, n-hexane tested in vivo against The optimal dose for the repellent [52]
Insecticidal/ Anopheles stephensi (Liston). activity was
larvicidal/ 2 mg/mL.
termiticidal
High insecticidal effect against
Aerial parts/ethanol Phthorimae operculella (Zeller). Phthorimae operculella (Zeller); no [144]
ovocidal effects.
Molecules 2025, 30, 851 36 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
Ethyl acetate extract:
500 ppm, 30 min: 98% mortality
and 500 ppm, 30 min:
Tested against Anopheles stephensi
Leaves/ethyl 93% mortality.
(Liston) and Culex quinquefasciatus [145]
acetate, methanol Methanol extract:
(Say) larvae.
500 ppm, 30 min: 82% mortality
and 500 ppm, 30 min:
86% mortality.
Methanol partitions: leaves:
LC50 = 18 µg/mL; roots:
The in vitro larvicidal activity of the LC50 = 17 µg/mL; twigs and stems:
methanol and petroleum ether LC50 = 0.3 µg/mL.
Whole plant/ethanol partitions from extracts of different Petroleum ether partitions: leaves: [86]
parts of the plant were assayed with LC50 = 54 µg/mL; roots:
the brine shrimp lethality test. LC50 = 47 µg/mL; twigs:
LC50 = 62 µg/mL; stems:
LC50 = 3.6 µg/mL.
The termiticidal activity of several
Most active extract:
Leaves/chloroform extracts was screened against [146]
LD50 = 5 µg/insect.
Microcerotermes beesoni.
63.3% mortality of
Applied to Zea mays L. against
Leaves and seeds/powder Sitophilus zeamais on the [147]
Sitophilus zeamais.
twenty-eighth day.
Different concentrations (10%, 5%, Survival of nymphs at 10%, 5%,
2.5%, 1.25%, 0.1%, 0.05%, 0.025%, 2.5%, and 1.25% concentrations =
0.0125%, and 0.00625%) of a crude 65.33%, 66.67%, 72%, and 85.33%.
Leaves/n-hexane [148]
extract were tested against Dysdercus Reduction in the longevity at
koenigii Fabricius nymphs for 24 h and 10% and 5% concentrations = 5.54
monitored for 7 days. and 5.95 days.
Flowers: A. arabiensis,
LC50 = 15.84 ppm;
Insecticidal/ Crude extracts were tested against
C. quinquefasciatus,
larvicidal/ Leaves and flowers/ethanol Anopheles arabiensis and [149]
LC50 = 21.37 ppm. Leaves:
termiticidal Culex quinquefasciatus larvae.
A. arabiensis, LC50 = 9.54 ppm;
C. quinquefasciatus, LC50 = 5.01 ppm.
The most active extracts: methanol:
A. aegypti, LC50 = 39.54 ppm;
The larvicidal activity of different
A. stephensis, LC50 = 35.65 ppm;
concentrations (25, 50, 75, 100, and
Whole plant/water, acetone, C. quinquefasciatus, LC50 = 35.36 ppm;
150 ppm) of crude extract was screened
chloroform, ethanol, ethanol: A. aegypti [150]
for 24 h against Aedes aegypti,
and methanol LC50 = 60.93 ppm; A. stephensi,
Anopheles stephensis, and
LC50 = 79.03 ppm;
Culex quinquefasciatus.
C. quinquefasciatus,
LC50 = 50.17 ppm.
Different concentrations (25, 50, 75,
and 100 ppm) were tested against
Leaves/diluted LC50 values ranged from 47.47 to
Aedes aegypti, Anopheles subpictus, and [151]
aqueous juice 101.68 ppm.
Culex quinquefasciatus larvae during 6,
12, and 24 h.
The insecticidal activity of different
concentrations (100, 200, 300, 400, and
Larvae: LC50 = 198.52 ppm;
Leaves/acetone 500 ppm) of a crude extract was tested [59]
pupae: LC50 = 309.64 ppm.
against Aedes aegypti L. larvae and
pupae for 24 h.
The insecticidal activity of different
concentrations (62.5, 125, 250, 500, and
A. aegypti L.: LC50 = 35.48 ppm;
1000 ppm) of a crude extract was tested
Leaves/water C. quinquefasciatus: [152]
against Aedes aegypti L. and
LC50 = 35.19 ppm.
Culex quinquefasciatus Say larvae for
24 h.
Different concentrations (250–3000
ppm) of a crude extract were tested
Leaves/95% ethanol LC50 = 477.53 ppm. [153]
against Anopheles arabiensis
Patton larvae.
Molecules 2025, 30, 851 37 of 51

Table 8. Cont.

Biological Part of the


Model Results Reference
Activity Plant/Solvent
The aqueous extract reduced the
Different concentrations (0%, 1.25%, viability of Bidens pilosa seeds
2.5%, 3.75%, and 5%, v/v) of a crude during phase III of germination.
Leaves/water [154]
extract were tested on At any concentration, the aqueous
Bidens pilosa seeds. extract inhibited the root and
epicotyl growth.
A crude extract, at the concentration of
Leaves/methanol–water 5 g/L, was tested on E. crassipes: complete inhibition;
[110]
(70:30) Eichhornia crassipes (Mart.) Solms and M. aeruginosa: 66.1% inhibition.
Microcystis aeruginosa Kütz.
Extract concentration that caused
50% inhibition of seed germination:
The inhibition of seed germination by
B. campestris: leaves = 0.62%,
crude extracts was tested on
Phytotoxic callus = 0.65%; I. aquatica:
Leaves and callus/water Brassica campestris var. chinensis, [165]
leaves = 0.94%, callus = 0.45%;
Ipomoea aquatica Forsk., Sorghum bicolor L.,
S. bicolor: leaves = 0.95%,
and Zea mays L.
callus = 1.19%; Z. mays:
leaves = 4.39%, callus = 3.05%.
An aqueous leaf leachate was tested on The concentration of 5% was the
Leaves/water [166]
Eichhornia crassipes (Mart.) Solms. most toxic after 21 days.
An aqueous leaf leachate was tested on The concentration of 1% was the
Callus/water [167]
Salvinia molesta Mitchell. most toxic after 7 days.
Beads with 5% extract had no
A crude extract encapsulated in
effect on the germination rate,
calcium alginate beads was
Callus/water while beads with 1–4% extract did [168]
tested on Brassica campestris
not reduce the total weight of
var. chinensis.
fresh seedlings.
Daily topical application of 100 mg Mean epithelization time and % of
extract/kg body weight on wounds of wound healing: placebo
Leaves/water male Sprague Dawley rats (200–220 g). group = 19 ± 0.14 days and 88%;
[169]
tested group = 17.20 ± 0.12 days
Wound-healing
Bovine dermatophilosis caused by and 98%.
effects
Dermatophilus congolensis was treated
[108]
Leaves/ethanol with ointments containing L. camara Wound healing was observed
leaf ethanolic extracts once a day for between the third and fourth day
10 days. of application without recurrence.
aBiological activities are ordered in alphabetic order. * MIC = minimum inhibitory concentration; MBC = minimum
bactericidal concentration; IC50 = sample concentration causing 50% inhibition; LC50 = sample concentration that
causes 50% mortality; CC50 = sample concentration causing 50% cytotoxicity.

Table 9. Bioactivities determined for the compounds isolated from Lantana camara and semisynthetic
derivatives.

Compound a, * (Nº) Biological Activity Reference


In vitro antibacterial activity against Mycobacterium tuberculosis strain H37Rv (ATCC 27294):
Acetyl lantanoside * (144) [16,134]
98% inhibition, MIC < 11.15 µM.
In vitro cytotoxic activity towards human leukemia HL-60 cells: IC50 = 75.09 ± 0.09 µM;
22β-Acetyloxy-oleanonic acid * (37) human cervical carcinoma Hela cells: IC50 = 72.75 ± 0.29 µM; colon 502,713 cells: IC50 = 67.1 [94]
± 0.04 µM; lung carcinoma A549 cells: IC50 = 71.77 ± 0.15 µM.
In vitro cytotoxic activity towards HL-60 cells: IC50 = 88.38 ± 0.15 µM; Hela cells:
22β-Benzoyloxy-oleanonic
IC50 = 80.55 ± 0.15 µM; colon 502,713 cells: IC50 = 89.07 ± 0.04 µM; lung A549 cells: [94]
acid * (85)
IC50 > 100 µM.
In vitro cytotoxic activity towards HL-60 cells: IC50 = 39.94 ± 0.23 µM; Hela cells:
IC50 = 42.16 ± 0.15 µM; colon 502,713 cells: IC50 = 46.6 ± 0.28 µM; lung A549 cells:
22β-Butanoyloxy-oleanonic IC50 = 50.11 ± 0.09 µM. In vivo antitumor activity: squamous cell carcinogenesis induced by
[94]
acid * (50) 7,12-dimethylbenz[a]anthracene/12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg/kg body weight administered orally for 20 weeks: 80% mice
survival and 17.2% overall papilloma incidence.
Molecules 2025, 30, 851 38 of 51

Table 9. Cont.

Compound a, * (Nº) Biological Activity Reference


In vitro nematocidal activity towards Meloidogyne incognita larvae: 95% mortality at
0.5% concentration after 48 h. In vitro antiparasitic activity towards Leishmania mexicana
promastigotes: IC50 = 2.52 ± 0.08 µM. In vitro protein tyrosine phosphatase 1B inhibition [80,82,84,
Camaric acid (62)
assay: IC50 = 5.1 µM [84]. In vitro cytotoxic activity towards HL-60 human promyelocytic 99,117,118]
leukemia cells (JCRB0085): IC50 = 1.71 ± 0.10 µM. In vitro anti-inflammatory activity
(inhibition of LPS-induced NO production in BV-2 cells): IC50 > 3 µM.
In vitro nematocidal activity against Meloidogyne incognita larvae: 100% mortality at
Camarin (23) [90]
1 mg/mL concentration after 48 h.
In vitro antimicrobial and antifungal activity index values: E. coli = 2, S. aureus = 0.95,
P. aeruginosa = 0.15, S. typhi = 0.7, C. albicans = 0.2, T. mentagrophytes = 2.3. In vivo
antimutagenic evaluation: micronucleus test (2.75 mg mitomycin D/kg body weight and
Camarinic acid (114) 6.75 mg/kg body weight given orally to Swiss strain mice, once a day-48 h): 76.7% reduction [16,78]
in the number of micronucleated polychromatic erythrocytes. In vitro nematocidal activity
against Meloidogyne incognita larvae: 100% mortality at 1% concentration after 24 h. In vitro
antiparasitic activity against Leishmania major promastigotes: IC50 = 89 ± 0.3 µM.
In vitro nematocidal activity against Meloidogyne incognita larvae: 100% mortality at
Camarinin (56) [90]
1 mg/mL concentration after 48 h.
In vitro antibacterial activity (disk diffusion method), zone inhibition diameter:
Di-(2-ethylhexyl) Escherichia coli = 20 mm, Staphylococcus aureus = 22 mm, Salmonella typhimurium = 21 mm,
[73,84]
phthalate (166) Pseudomonas aerugionosa = 23 mm. In vitro protein tyrosine phosphatase inhibition assay:
IC50 = 8.1 µM.
Ethyl-β-D-galactopyranoside (163) Inactive in an in vitro antiparasitic activity assay towards Brugia malayi. [74]
In vivo antiulcer activity: aspirin-induced and ethanol-induced ulcer models; Albino Wistar
3-O-β-D-Glucosyl oleanolic rats (150–200 g) were divided into 4 groups. Groups III and IV received 25 and 50 mg
[126]
acid (84) compound/kg body weight, respectively, orally once a day for 5 days. Ulcer index:
3.48± 0.83 and 1.99 ± 0.34, respectively; protection: 21.24 and 38.37%, respectively.
Hispidulin (128) In vitro protein tyrosine phosphatase inhibition assay: IC50 > 33 µM. [72,79,84]
In vitro antifungal activity against Fusarium subglutinans (PPRI 6740), F. solani (PPRI 19147),
F. graminearum (PPRI 10728), and F. semitectum (PPRI 6739): MIC > 1000 µM; against
9-Hydroxy-lantadene A (64) [120]
F. proliferatum (PPRI 18679): MIC = 70.32 µM. In vitro cytotoxic activity towards Raw 264.7
cells: IC50 > 100 µM.
Binding affinity to the antiapoptotic protein Bcl-xL: Ki = 5.3 µM. In vitro cytotoxic activity
24-Hydroxy-lantadene B
towards papilloma KB cells: IC50 = 35.5 µM; colon carcinoma HCT-116 cells: IC50 = 11.4 µM;
≡ 24-Hydroxy-22β-senecioyloxy- [84,116]
breast adenocarcinoma MCF7 cells: IC50 = 42.5 µM; lymphocytic leukemia L1210 cells:
oleanonic acid (65)
IC50 = 12.3 µM. In vitro protein tyrosine phosphatase inhibition assay: IC50 = 7.3 µM.
24-Hydroxy-lantadene D (53) In vitro protein tyrosine phosphatase inhibition assay: IC50 >18 µM. [84]
In vitro cytotoxic activity: tested on multiple cancer cells. In vitro anti-inflammatory activity
(TNF-α-induced NF-KB activation inhibitory activity): IC50 > 10 µM; COX-2 inhibition: [80,84,95,
22β-Hydroxy- oleanolic acid (32)
IC50 > 100 µM. In vitro cytotoxic activity towards A549 cells: IC50 > 10 µM. In vitro protein 96]
tyrosine phosphatase inhibition assay: IC50 = 7.9 µM.
In vitro antitumor activity: Epstein–Barr virus early antigen activation assay induced by
12-O-tetradecanoylphorbol-13-O-acetate (TPA) in Raji cells: 35.3% inhibition at 100 mol
tested compound/1 mol TPA. In vivo hepatotoxicity evaluation (adult female guinea pigs
received 125 mg compound/kg body weight orally in gelatin capsules): bilirubin:
22β-Hydroxy-oleanonic acid (26) 0.67 ± 0.001 mg/100 mL, SGOT: 46.1 ± 0.4 U/L, SGPT: 39 ± 0.3 U/L; nontoxic. In vitro [84,92–96]
cytotoxic activity towards HL-60, Hela, colon 502,713, and lung A549 cells: IC50 > 100 µM;
A549 cells: IC50 > 10 µM [94]. In vitro anti-inflammatory activity (inhibitory activity of
TNF-α-induced NF-KB activation): IC50 = 6.42 ± 1.24 µM; COX-2 inhibition: IC50 > 100 µM.
In vitro protein tyrosine phosphatase inhibition assay: IC50 = 6.9 µM.
11α-Hydroxy-3-oxo-urs-12-en-28- In vitro nematocidal activity against Meloidogyne incognita larvae: 70% mortality at
[82]
oic acid (101) 0.25% concentration after 72 h.
Binding affinity to the antiapoptotic protein Bcl-xL: Ki = 7.6 µM. In vitro cytotoxic activity
towards KB cells: IC50 = 15 µM; HCT-116 colon cancer cells: IC50 = 5.8 µM; MCF7 cells:
IC50 = 11.3; L1210 lymphocytic leukemia cells: IC50 = 6.8 µM; HL-60 human promyelocytic
Icterogenin (67) leukemia cells (JCRB0085): IC50 = 34.2 ± 0.7 µM. In vitro protein tyrosine phosphatase [109]
inhibition assay: IC50 = 11 µM. In vitro anti-inflammatory activity (inhibition of LPS-induced
NO production in BV-2 cells): IC50 > 3 µM. DPPH radical scavenging activity:
IC50 = 169.7 µg/mL.

In vitro screening against a variety of Gram-positive and Gram-negative bacteria (disk


Lancamarinic acid (41) [105]
diffusion method).
Molecules 2025, 30, 851 39 of 51

Table 9. Cont.

Compound a, * (Nº) Biological Activity Reference


In vitro nematocidal activity against Meloidogyne incognita larvae: 80% mortality at
Lancamarolide (42) [81]
0.25% concentration after 48 h.
In vitro cytotoxic activity towards HL-60 human promyelocytic leukemia cells (JCRB0085):
Lantacamaric acid A (29) [99]
IC50 = 30.8 ± 2.7 µM.
In vitro cytotoxic activity towards HL-60 human promyelocytic leukemia cells (JCRB0085):
Lantacamaric acid B (70) [99]
IC50 = 6.60 ± 0.46 µM.
In vitro nematocidal activity against Meloidogyne incognita larvae: 100% mortality at
Lantacin (119) [81,90]
1 mg/mL concentration after 48 h.
In vitro larvicidal activity: very toxic in the brine shrimp lethality test; insecticidal activity at
5.0 mg/mL towards Spodoptera littoralis Biosduval: 40% lethality after 48 h; fecundity
inhibition assay in Clavigralla tomentosicollis Stal.: 50% fecundity suppression; inactive
towards Aphis craccivora Koch. In vivo antimotility effect evaluation (Laca strain male mice
(20–25 g) received a single injection of 85 and 170 mg compound/kg body weight):
% intestinal transit = 39.47 ± 10.05 and 27.34 ± 4.58, respectively. Phytotoxic activity towards
Eichhornia crassipes (Mart.) Solms and Microcystis aeruginosa Kutz: ErC50 = 24.78 and
21.34 mg/L, respectively. In vivo hepatotoxicity evaluation (adult female guinea pigs
received 125 mg compound/kg body weight orally in gelatin capsules):
bilirubin = 8.74 ± 2.5 mg/100 mL, SGOT = 696.3 ± 3.1 U/L, SGPT = 305.2 ± 3.9 U/L; toxic.
In vitro cytotoxicity towards HL-60 cells: IC50 = 35.81 ± 0.40 and 35 ± 1 µM; HeLa cells:
IC50 = 42.15 ± 0.09 and 42 ± 8 µM; colon 502,713 cells: IC50 = 38.53 ± 0.09 and 38 ± 5 µM
µM; lung A549 cells: IC50 = 39.43 ± 0.21, 39 ± 1 µM µM, and 2.84 ± 0.72 µM; KB cells:
IC50 = 15.8 µM; HCT-116 cells: IC50 = 41.8 µM; MCF7 cells: IC50 = 44.7 and >100 µM; [78,82,84,
L1210 cells: IC50 = 16.3 µM; HL-60 human promyelocytic leukemia cells (JCRB0085): 86,91–
Lantadene A (Rehmannic acid) (60) IC50 = 25.4 ± 3.1 µM; LNcap prostatic cancer cells: IC50 > 100 µM; RWPE-1 prostatic cancer 96,99,100,
cells: IC50 > 100 µM. Lantadene A-gold nanoparticles reduced MCF-7 (breast cancer cells) 105,109–
viability, upregulated the p53 expression, and downregulated the BCL-2 expression. In vivo 116]
antitumor activity: squamous cell carcinogenesis induced by
7,12-dimethylbenz[a]anthracene-12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg compound/kg body weight administered orally for 20 weeks:
80–100% mice survival and 17.9–18.1% overall papilloma incidence. Binding affinity to the
antiapoptotic protein Bcl-xL: Ki > 100 µM. Antioxidant activity in a dose-dependent manner.
Toxicity evaluation: toxic (2 g) orally to sheep; nontoxic to lambs (167 mg compound/kg
body weight administered orally in gelatin capsules) and guinea pigs (667 mg compound/kg
body weight administered orally in gelatin capsules). In vitro antiparasitic activity against
Leishmania major promastigotes: IC50 = 20.4 ± 0.1 µM. In vitro anti-inflammatory activity
(inhibition of TNF-α-induced NF-KB activation): IC50 = 1.06 ± 0.46 µM; COX-2 inhibition:
IC50 > 100 µM. In vitro nematocidal activity towards Meloidogyne incognita larvae:
70% mortality at 0.5% concentration after 48 h. In vitro protein tyrosine phosphatase
inhibition assay: IC50 = 5.2 µM. DPPH radical scavenging activity: IC50 = 93.94 µM.
In vitro cytotoxic activity towards HL-60 cells: IC50 = 47.79 ± 0.24 µM; Hela cells:
Lantadene A acyl chloride * (58) IC50 = 46.21 ± 0.17 µM; colon 502,713 cells: IC50 = 49.19 ± 0.17 µM; lung A549 cells: [93]
IC50 = 50.07 ± 0.14 µM.
In vitro cytotoxic activity towards HL-60 cells: IC50 = 34.04 ± 0.26 and 34 ± 1.4 µM; Hela
cells: IC50 = 37.93 ± 0.09 and 37 ± 5 µM; colon 502,713 cells: IC50 = 37.22 ± 0.15 and
37 ± 8 µM; lung A549 cells: IC50 = 33.87 ± 0.09 and 33 ± 5 µM. In vivo antitumor activity:
Lantadene A methyl ester * (81) squamous cell carcinogenesis induced by [112]
7,12-dimethylbenz[a]anthracene-12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg compound/kg body weight administered orally for 20 weeks:
87.5–100% mice survival and 13.6–19.6% overall papilloma incidence.
In vitro cytotoxic activity towards HL-60 cells: IC50 = 70.43 ± 0.22 µM; Hela cells:
IC50 = 74.0 ± 0.09 µM; colon 502,713 cells: IC50 = 78.68 ± 0.15 µM; lung A549 cells:
IC50 = 82.80 ± 0.18 µM. In vivo antitumor activity: squamous cell carcinogenesis induced by
Lantadene A nitrile * (57) [93]
7,12-dimethylbenz[a]anthracene-12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg compound/kg body weight given orally for 20 weeks:
75% mice survival and 24.9% overall papilloma incidence.
Phytotoxic activity against Eichhornia crassipes (Mart.) Solms and Microcystis aeruginosa Kütz:
ErC50 = 19.53 and 17.37 mM, respectively. Binding affinity to the antiapoptotic protein
Bcl-xL: Ki > 100 µM. In vitro cytotoxic activity against KB cells: IC50 = 25.3 µM; HCT-116
cells: IC50 = 11.4 µM; MCF-7 cells: IC50 = 44 µM and >100 µM; L1210 cells: IC50 = 16.1 µM;
[82,84,95,
A549 (lung carcinoma) cells: IC50 = 1.19 ± 0.28 µM. In vitro cytotoxic activity (MTT test)
96,109,110,
Lantadene B (61) towards MCF-7 breast cancer cells: IC50 = 1.13 µM. In vitro anti-inflammatory activity
116,117,
(inhibition of TNF-α-induced NF-KB activation): IC50 = 1.56 ± 0.04 µM; COX-2 inhibition:
124]
IC50 > 100 µM. In vitro nematocidal activity against Meloidogyne incognita larvae:
60% mortality at 0.25% concentration after 48 h; against Leishmania mexicana promastigotes,
IC50 = 23.45 ± 2.15 µM. In vitro protein tyrosine phosphatase inhibition assay: IC50 = 5.5 µM.
DPPH radical scavenging activity: IC50 = 76.45 µM.
Molecules 2025, 30, 851 40 of 51

Table 9. Cont.

Compound a, * (Nº) Biological Activity Reference


Binding affinity to the antiapoptotic protein Bcl-xL: Ki > 100 µM. In vitro cytotoxic activity
towards KB cells: IC50 = 15.8 µM; HCT-116 cells: IC50 = 41.8 µM; MCF7 cells: IC50 = 44.7 and
Lantadene C (72) >100 µM; L1210 cells: IC50 = 16.3 µM; HL-60 cells: IC50 > 100 µM; Hela cells: IC50 > 100 µM; [84,116]
colon 502,713 cells: IC50 > 100 µM; lung A549 cells: IC50 > 100 µM. DPPH radical
scavenging activity: IC50 > 100 µM.
In vivo antitumor activity: squamous cell carcinogenesis induced by
7,12-dimethylbenz[a]anthracene-12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
Lantadene D (51) mice (LACCA/female); 50 mg compound/kg body weight given orally for 20 weeks: [84,97]
approximately 85% mice survival and 30% overall papilloma incidence. In vitro protein
tyrosine phosphatase inhibition assay: IC50 = 7.9 µM [84].
In vitro nematocidal activity against Meloidogyne incognita larvae: 98.66% mortality at
0.5% concentration after 48 h. In vitro antiparasitic activity against Leishmania mexicana
promastigotes: IC50 = 9.50 ± 0.28 µM; L. major promastigotes: IC50 = 21.3 ± 0.02 µM; brine
[78,80,81,
shrimp toxicity assay: LC50 = 49.20 µM. In vitro antibacterial and antifungal activity:
84,98,99,
Lantanilic acid (68) diameter of inhibition zone at a concentration of 500 µg/mL against S. aureus = 1.7 mm and
118,122,
against C. albicans = 9.3 mm. In vitro protein tyrosine phosphatase inhibition assay:
123]
IC50 = 7.5 µM [84]. In vitro cytotoxic activity towards HL-60 human promyelocytic leukemia
cells (JCRB0085): IC50 = 4.00 ± 0.67 µM. In vitro anti-inflammatory activity (inhibition of
LPS-induced NO production in BV-2 cells): IC50 > 3 µM.
In vitro antiparasitic activity against Leishmania major promastigotes: IC50 = 164 ± 0.8 µM.
In vitro nematocidal activity against Meloidogyne incognita larvae: 60% mortality at a
Lantaninilic acid (30) [78,82]
concentration of 0.125% after 48 h. In vitro cytotoxic activity towards HL-60 human
promyelocytic leukemia cells (JCRB0085): IC50 = 68.4 ± 15.4 µM.
In vitro nematocidal activity against Meloidogyne incognita larvae: 100% mortality at a
Lantanolic acid (27) concentration of 1 mg/mL after 24 h. In vitro protein tyrosine phosphatase inhibition assay: [84,90]
IC50 = 13 µM.
In vitro nematocidal activity against
Meloidogyne incognita larvae: 95% mortality at a concentration of 1% after 48 h. In vitro
Lantanoside (140) [16,134]
antibacterial activity against Mycobacterium tuberculosis strain H37Rv (ATCC 27294): 37%
inhibition, MIC > 12.05 µM.
In vitro antimicrobial activity (bioautography assays): minimum growth inhibition values
for B. subtilis (ATCC 6633), M. luteus (ATCC 9341), S. aureus (ATCC 6538P), and P. mirabilis
(ATCC 14153) = 0.3 µg; B. cereus (ATCC 11778) = 0.1 µg; S. faecalis (ATCC 8043) and
Lantic acid (102) [90]
P. aeruginosa (ATCC 25619) = 1.1 nmol; E. coli (ATCC 25922) = 0.17 nmol. In vitro nematocidal
activity against Meloidogyne incognita larvae: 100% mortality at a concentration of 1 mg/mL
after 24 h.
In vitro nematocidal activity against Meloidogyne incognita larvae: 100% mortality at a
Lantoic acid (105) concentration of 1 mg/mL after 24 h. In vitro antiparasitic activity against Leishmania major [81,90]
promastigotes: IC50 = 97 ± 0.02 µM.
In vitro anti-inflammatory activity (inhibition of LPS-induced NO production in BV-2 cells):
Lantrieuphpene A (13) [80]
IC50 > 30 µM.
In vitro anti-inflammatory activity: inhibition of LPS-induced NO production in BV-2 cells,
IC50 = 24 ± 0.30 µM; ROS and NO levels in LPS-stimulated zebrafish embryos significantly
Lantrieuphpene B (8) [80]
decreased in a concentration-dependent manner. Western blotting: iNOS protein expression
decreased in a dose-dependent manner on pretreated cells.
In vitro anti-inflammatory activity: inhibition of LPS-induced NO production in BV-2 cells,
IC50 = 27.98 ± 0.98 µM; ROS and NO levels in LPS-stimulated zebrafish embryos
Lantrieuphpene C (9) [80]
significantly decreased in a concentration-dependent manner. Western blotting: iNOS
protein expression decreased in a dose-dependent manner on pretreated cells.
In vitro anti-inflammatory activity: inhibition of LPS-induced NO production in BV-2 cells,
Lantrieuphpene D (12) [80]
IC50 > 10 µM.
In vitro nematocidal activity against Meloidogyne incognita larvae: 90% mortality at a
concentration of 1% after 48 h. In vitro antibacterial activity against the [16,134,
Linaroside (139)
Mycobacterium tuberculosis strain H37Rv (ATCC 27294): 30% inhibition, MIC = 13.12 µM. 162]
In vitro antioxidant activity (DPPH test): IC50 = 149.09 mM.
In vitro cytotoxic activity towards HL-60 cells: IC50 = 72.75 ± 0.29 µM; Hela cells:
Methyl 22β-acetyloxy-
IC50 = 70.6 ± 0.10 µM; colon 502,713 cells: IC50 = 67.48 ± 0.15 µM; lung A549 cells: [94]
oleanonate * (45)
IC50 = 71.77 ± 0.10 µM.
Molecules 2025, 30, 851 41 of 51

Table 9. Cont.

Compound a, * (Nº) Biological Activity Reference


In vitro cytotoxic activity towards HL-60 cells: IC50 = 26 ± 6 µM; HeLa cells:
IC50 = 31 ± 5 µM; colon 502,713 cells: IC50 = 32 ± 1 µM; lung A549 cells: IC50 = 28 ± 4 µM.
Methyl 22β-angelyloxy-2-hydroxy- In vivo antitumor activity: squamous cell carcinogenesis induced by
[124]
3-oxo-olean-1,12-diene-28-oate (78) 7,12-dimethylbenz[a]anthracene-12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg compound/kg body weight administered orally for 20 weeks:
approximately 100% mice survival and 17.2% overall papilloma incidence.
Methyl 22β-benzoyloxy- In vitro cytotoxic activity towards HL-60 cells: IC50 = 81.52 ± 0.08 µM; Hela cells:
[94]
oleanonate * (86) IC50 = 86.10 ± 0.08 µM; colon 502,713 and lung A-549 cells: IC50 > 100 µM.
In vitro cytotoxic activity towards HL-60 cells: IC50 = 34.79 ± 0.14 µM; Hela cells:
IC50 = 36.23 ± 0.38 µM; colon 502,713 cells: IC50 = 38.03 ± 0.09 µM; lung A549 cells:
Methyl 22β-butanoyloxy- IC50 = 40.37 ± 0.09 µM. In vivo antitumor activity: squamous cell carcinogenesis induced by
[94]
oleanonate * (73) 7,12-dimethylbenz[a]anthracene-12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg compound/kg body weight administered orally for 20 weeks:
80% mice survival and 12.4% overall papilloma incidence.
Methyl 22β-hydroxy- In vitro cytotoxicity towards HL-60 cells: IC50 > 100 µM; Hela cells: IC50 > 100 µM;
[91,94]
oleanonate * (35) colon 502,713 cells: IC50 > 100 µM; lung A549 cells: IC50 > 100 µM.
In vitro cytotoxicity towards HL-60 cells: IC50 = 71.19 ± 0.09 µM; Hela cells:
Methyl 22β-isobutyryloxy-
IC50 = 74.08 ± 0.38 µM; colon 502,713 cells: IC50 = 68.67 ± 0.09 µM; lung A549 cells: [94]
oleanonate * (74)
IC50 = 76.06 ± 0.14 µM.
In vitro cytotoxicity towards HL-60 cells: IC50 = 44.75 ± 0.39 µM; Hela cells:
IC50 = 48.81 ± 0.15 µM; colon 502,713 cells: IC50 = 41.42 ± 0.15 µM; lung A549 cells:
Methyl 22β-propanoyloxy- IC50 = 52.52 ± 0.39 µM. In vivo antitumor activity: squamous cell carcinogenesis induced by
[94]
oleanonate * (52) 7,12-dimethylbenz[a] anthracene-12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg compound/kg body weight administered orally for 20 weeks:
80% mice survival and 17.9% overall papilloma incidence.
In vitro larvicidal activity: 30% lethality in the brine shrimp lethality test against
Spodoptera littoralis Biosduval after 48 h at a concentration of 10.95 mM. Inactive in the
fecundity inhibition assay against Clavigralla tomentosicollis Stal. and Aphis craccivora Koch.
In vitro nematocidal activity against Meloidogyne incognita larvae: 70.33% mortality at a
concentration of 0.5% after 48 h. In vitro antifilarial activity against Brugia malayi:
LC100 = 136.85 µM. In vivo antifilarial activity against Brugia malayi in rodel model
[75,78,80,
Mastomys coucha (100 or 200 mg compound/kg body weight administered intraperitoneally
Oleanolic acid (31) 84,86,87,
for 5 days): macrofilaricidal efficacy = 9.09% and 18.18%, respectively; percent female
118]
sterility = 49.22 ± 10.57 and 56.50 ± 9.50, respectively. In vitro cytotoxic activity towards
HCT-15 cells: IC50 = 52 µM; SW-620 cells: IC50 = 25 µM; A549 cells: IC50 = 52 µM; IGROV-1
cells: IC50 = 8 µM; IMR-32 cells: IC50 = 61 µM. In vitro antiparasitic activity against
Leishmania major promastigotes: IC50 = 53 ± 0.02 µM. In vitro protein tyrosine phosphatase
inhibition assay: IC50 = 2 µM. In vitro anti-inflammatory activity (inhibition of LPS-induced
NO production in BV-2 cells): IC50 > 60 µM.
In vitro larvicidal activity: 20% lethality in the brine shrimp lethality test against
Spodoptera littoralis Biosduval: after 48 h at a concentration of 10.996 mM. Inactive in the
fecundity inhibition assay towards Clavigralla tomentosicollis Stal. and Aphis craccivora Koch.
In vitro antifilarial activity against Brugia malayi: LC100 = 68.73 µg/mL. In vivo antifilarial
activity against Brugia malayi in rodel model Mastomys coucha (100 or 200 mg compound/kg
[26,75,82,
body weight administered intraperitoneally for 5 days), macrofilaricidal efficacy: inactive;
Oleanonic acid (22) 84,86,88,
% female sterility: 56.56 ± 9.49 and 29.71 ± 6.52, respectively. In vitro cytotoxic activity
99]
towards EAC cells: IC50 = 7.1 ± 1.3 µM; A375 cells: IC50 = 10.9 ± 1.5 µM; Hep2 cells:
IC50 = 59.3 ± 1.1 µM; U937 cells: IC50 = 16.5 ± 1.3 µM; HL-60 human promyelocytic
leukemia cells (JCRB0085): IC50 = 9.79 ± 2.13 µM; PMBC cells: IC50 > 100 µM. In vitro
nematocidal activity against Meloidogyne incognita larvae: 80% mortality at a concentration of
0.5% after 48 h. In vitro protein tyrosine phosphatase inhibition essay: IC50 = 6.9 µM [84].
In vitro antifungal activity against Fusarium subglutinans (PPRI 6740) and F. semitectum
(PPRI 6739): MIC = 1.338 mM; F. proliferatum (PPRI 18679): MIC = 2.762 mM; F. solani
11-Oxo-β-boswellic acid (103) [120]
(PPRI 19147) and F. graminearum (PPRI 10728): MIC = 5.311 mM. In vitro cytotoxicity
towards Raw 264.7 cells: IC50 > 100 µM.
In vitro nematocidal activity against Meloidogyne incognita larvae: 100% mortality at a
Pomolic acid (107) concentration of 1 mg/mL after 24 h. In vitro protein tyrosine phosphatase inhibition assay: [84,90]
IC50 = 10.6 µM.
In vitro protein tyrosine phosphatase inhibition assay: IC50 = 10.5 µM. In vitro
Pomonic acid (104) anti-inflammatory activity (inhibition of LPS-induced NO production in BV-2 cells): [80,84]
IC50 > 30 µM.
Molecules 2025, 30, 851 42 of 51

Table 9. Cont.

Compound a, * (Nº) Biological Activity Reference


In vitro cytotoxic activity towards HL-60 cells: IC50 = 50.12 ± 0.32 µM; Hela cells:
IC50 = 54.29 ± 0.09 µM; colon 502,713 cells: IC50 = 48.22 ± 0.09 µM; lung A549 cells:
22β-Propanoyl- IC50 = 56.19 ± 0.26 µM. In vivo antitumor activity: squamous cell carcinogenesis induced by
[94]
oxy-oleanonic acid * (46) 7,12-dimethylbenz[a] anthracene/12-O-tetradecanoylphorbol-13-O-acetate in Swiss albino
mice (LACCA/female); 50 mg compound/kg body weight administered orally for 20 weeks:
80% mice survival and 19.6% overall papilloma incidence.
Evaluation of toxicity to sheep: 80 mg compound/kg body weight administered orally in
gelatin capsules: nontoxic; 80 mg compound/kg body weight, dissolved in DMSO,
intraruminal administration: toxic. Evaluation of toxicity to Wistar female rats: 15 mg
Reduced lantadene A
compound/kg body weight administered orally in olive oil: toxic. In vitro antitumor
(22β-angelyloxy-3β-hydroxy-olean- [84,91,95]
activity: Epstein–Barr virus early antigen activation assay induced by
12-en-28-oic acid) (75)
12-O-tetradecanoylphorbol-13-O-acetate (TPA) in Raji cells: 30.6% inhibition at a
concentration of 100 mol compound/1 mol TPA. In vitro cytotoxicity was tested towards
multiple cancer cells. In vitro protein tyrosine phosphatase inhibition assay: IC50 = 7.2 µM.
Reduced lantadene B
In vitro cytotoxicity was tested towards multiple cancer cells. In vitro protein tyrosine
(3β-hydroxy-22β-senecioyloxy- [95]
phosphatase inhibition assay: IC50 = 5.1 µM.
olean-12-en-28-oic acid) (76)
Reduced lantadene C
3β-hydroxy-22β-
[2-methylbutanoyloxy]-olean-12-en- In vitro protein tyrosine phosphatase 1B inhibition assay: IC50 = 7.3 µM. [84]
28-oic acid (77)

In vitro antiparasitic activity against Brugia malayi: LC100 > 1.2 mM. Antibacterial activity
(disk diffusion method): diameter of inhibition zone = 14 mm for Escherichia coli, 19 mm for
β-Sitosterol (3) Staphylococcus aureus, 17 mm for Salmonella typhimurium, 24 mm for Pseudomonas aeruginosa. [73,74,170]
The cytotoxic potential was tested in vitro by an MTT assay against T47D (breast cancer cells)
and HeLa (cervical cancer cells): IC50 = 24.06 and 24.86 µM, respectively.
β-Sitosterol 3-O-β-D-
In vitro antiparasitic activity against Brugia malayi: LC100 > 0.86 mM. [71]
glucopyrano-side (4)
Stearic acid (158) In vitro antiparasitic activity against Brugia malayi: LC100 > 1.7 mM. [74,77]
In vitro antibacterial activity (disk diffusion method): diameter of inhibition zone = 20 mm
Trilinolein (168) for Escherichia coli, 19 mm for Staphylococcus aureus, 18 mm for Salmonella typhimurium, 21 mm [73]
for Pseudomonas aeruginosa.
In vivo antidiabetic activity: Wistar albino rats (150–200 g) received 0.3 mg/kg body weight
Urs-12-en-3β-ol-28-oic acid
orally for 21 days. Blood glucose levels: 8th day = 183.56 ± 3.61 mg/dL, 14th
3-O-β-D-glucopyrano- [125]
day = 143.43 ± 2.79 mg/dL, 21st day = 118.67 ± 2.40 mg/dL. In vivo anxiolytic activity:
syl-4′ -octadecano- ate (127)
dose-dependent effect.
In vitro nematocidal activity against Meloidogyne incognita larvae: 100% mortality at a
Ursolic acid (106) concentration of 1 mg/mL after 48 h. In vitro antiparasitic activity against Leishmania major [78,90]
promastigotes: IC50 = 12.4 ± 0.03 µM.
a Compounds are ordered in alphabetic order. Cytotoxicity values (IC ) are expressed in µM for homogeneity.
50
* Semisynthetic derivative. ErC50 = concentration of test substance which caused 50% reduction in growth rate
relative to the control for a 72 h exposure.

A few compounds isolated from L. camara were also submitted to molecular docking
studies (Table 10) towards the active site of RNA-dependent RNA polymerase (RdRp)
of SARS-CoV-2. The highest binding energy (around 6 kcal/mol) was determined for
camarolic acid (69) and lantoic acid (105). These values, when compared to remdesivir
(−5.75 kcal/mol), indicated that compounds 69 and 105 can serve as promising anti-
COVID-19 candidates. Moreover, lantrieuphpene B (8) and C (9) exhibited a high binding
affinity (a binding energy of around 9 kcal/mol) to the TYR-341, TYR-367, and ASP-376
residues of inducible Nitric Oxide Synthase (iNOS). In addition, a recent in silico study has
evaluated 20 selected constituents of L. camara as potent inhibitors of the human enzymes
acetylcholinesterase (hAchE), carbonic anhydrase II (hCA-II), and carboxylesterase 1 (hCES-
1), which are pharmacological targets for the treatment of neurodegenerative diseases,
glaucoma, obesity, and type 2 diabetes [171]. All of the twenty ligands docked effectively
with the CA-II enzyme. Only ursonic acid (100) was ineffective in both docking and binding
with AchE and CES-1, while lantic acid (102) exhibited the least atomic binding energy with
Molecules 2025, 30, 851 43 of 51

all three enzymes. The glucosyl flavone camaroside [17] exhibited the maximum binding
energy (−9.34 kcal/mol) with hAchE, while the phenylethanoid glycoside isonuomioside
A [17] demonstrated the highest binding energy (−9.72 kcal/mol) with hCA-II, and the
flavone pectolinarin (146) showed the highest binding energy (−9.21 kcal/mol) with
hCES-1 [172].

Table 10. In silico studies of compounds isolated from Lantana camara.

Compound (Nº) Docking Value Reference


Molecular docking into the active site of SARS-CoV-2 RNA-dependent RNA polymerase (RdRp):
Camaranoic acid (99) [81]
binding free energy = 1.272 kcal/mol.
Camaric acid (62) Molecular docking into the active site of SARS-CoV-2 RdRp: binding free energy = −3.198 kcal/mol. [81]
Camarolic acid (69) Molecular docking into the active site of SARS-CoV-2 RdRp: binding free energy = −6.73 kcal/mol. [81]
Icterogenin (67) Molecular docking into the active site of SARS-CoV-2 RdRp: binding energy = −2.311 kcal/mol. [81]
Lantabetulic acid (17) Molecular docking into the active site of SARS-CoV-2 RdRp: binding free energy = −2.958 kcal/mol. [81]
Lantacin (119) Molecular docking into the active site of SARS-CoV-2 RdRp: binding free energy = −2.919 kcal/mol. [81]
Lantaiursolic acid (120) Molecular docking into the active site of SARS-CoV-2 RdRp: binding free energy = −3.867 kcal/mol. [81]
Lantanilic acid (68) Molecular docking into the active site of SARS-CoV-2 RdRp: binding free energy = −3.633 kcal/mol. [81]
Lantoic acid (105) Molecular docking into the active site of SARS-CoV-2 RdRp: binding free energy = −6.07 kcal/mol. [81]
Binding free energy = −9.8 Kcal/mol with TYR-341, TYR-367, and ASP-376 residues of iNOS (inducible
Lantrieuphpene B (8) [80]
Nitric Oxide Synthase).
Molecular binding free energy = −9.3 Kcal/mol with TYR-341, TYR-367, and ASP-376 residues
Lantrieuphpene C (9) [80]
of iNOS.
Molecular binding free energies towards Bcl-2 and HPV16 E7 protein receptors: −8.11 and
β-Sitosterol (3) [170]
−7.276 kcal/mol, respectively

Finally, in unusual applications, leaf, fruit, flower, root, and seed extracts of
Lantana camara have been used to prepare several metal (Ag, Au, Fe, Cu, Zn, Pd, and
Pt) [173] and metal oxide (ZnO, SrO, CuO, NiO, and Y2 O3 ) nanoparticles with potential
photocatalytic, electrochemical, anticancer, antiarthritic, and antibacterial properties, and
other medical applications. The biomass and leaves of L. camara have also been used as a
sustainable alternative for the removal of antibiotics and metals, such as Pb (II), Zn (II), and
Mn (II) from contaminated rivers and waste waters [171,174–176].

4. Conclusions
This review, reporting on the recently published information on the phytochemistry
and bioactivities of L. camara, clearly demonstrates that this species continues to be one of
the most investigated plants due to the various traditional uses, the rich phytochemical
contents of the extracts, and the wide variety of biological activities exhibited by total
extracts, several isolated compounds, and the many semisynthetic derivatives.
Perspectives. In our opinion, among the various biological effects exhibited by spe-
cialized metabolites from L. camara (Tables 8 and 9), the nematocidal and antiparasitic
properties of several compounds and the antimalarial effects of leaf extracts against the
chloroquine-sensitive strains 3D7 and D10, and the chloroquine-resistant strain W2, of
Plasmodium falciparum deserve further investigations with in vivo and in the field tests.
Given the various structural features of active compounds, there may also be an opportu-
nity to conduct QSAR studies and to clarify the mechanism(s) of action, and to identify
the molecular target(s) and the biological processes involved in the nematocidal and an-
tiparasitic properties. Moreover, the interesting antidiabetic and anti-COVID-19 properties
in vitro of leaf extracts and a few isolated compounds must be confirmed by additional in
silico and in vivo studies. Computational strategies involving artificial intelligence and
machine learning algorithms are expected to help in the full exploration of the biological
Molecules 2025, 30, 851 44 of 51

space of natural molecules from L. camara, and to identify the unexplored human receptors
and enzymes to which they can bind. Semisynthesis is an important technique to harness
nature’s diversity for novel drugs. In this regard, semisynthetic efforts to prepare analogs
of natural products isolated from L. camara to enhance their biological properties are limited
and, therefore, they must be intensified.
Finally, preclinical and clinical research studies, which are missing so far, are
necessary to evaluate the efficacy and safety of the products with the most promising
medicinal properties.

Author Contributions: Conceptualization, J.R. and C.A.; methodology, N.E.-O.; software, L.N.C.;
validation, J.R., N.E.-O. and L.N.C.; investigation, N.E-O.; writing—original draft preparation,
C.A. and J.R.; writing—review and final editing, J.R., C.A., L.N.C. and G.V.; supervision, J.R. and
G.V.; project administration, J.R. All authors have read and agreed to the published version of
the manuscript.

Funding: This research was funded by the Universidad Técnica Particular de Loja (UTPL). Nº Grant:
PROY_PROY_ARTIC_QU_2022_3652.

Data Availability Statement: All data are available on database reported.

Acknowledgments: We are grateful to the Universidad Técnica Particular de Loja (UTPL) for
supporting open-access publication.

Conflicts of Interest: The authors declare no conflicts of interest.

References
1. Byng, J.W.; Chase, M.W.; Christenhusz, M.J.; Fay, M.F.; Judd, W.S.; Mabberley, D.J. An update of the angiosperm phylogeny group
classification for the orders and families of flowering plants: APG IV. Bot. J. Linn. Soc. 2016, 181, 1–20.
2. WFO Plant List. Snapshots of the Taxonomy. Search for Lantana L. WFO Plant List | World Flora Online. Available online:
https://wfoplantlist.org/taxon/wfo-4000020568-2023-12?page=1 (accessed on 9 March 2024).
3. Cardoso, P.H.; O’Leary, N.; Olmstead, R.G.; Moroni, P.; Thode, V.A. An update of the Verbenaceae genera and species numbers.
Plant Ecol. Evol. 2021, 154, 80–86. [CrossRef]
4. Lu-Irving, P.; Bedoya, A.M.; Salimena, F.R.G.; dos Santos Silva, T.R.; Viccini, L.F.; Bitencourt, C. Phylogeny of Lantana, Lippia, and
related genera (Lantaneae: Verbenaceae). Am. J. Bot. 2021, 108, 1354–1373. [CrossRef]
5. Babar, V.B.; Khapale, P.R.; Nagarale, S.N. Preliminary investigation and in-vitro anticancer activity of Lantana camara L. (Verbe-
naceae). J. Pharmacogn. Phytochem. 2019, 8, 2524–2527.
6. El Baroty, G.S.; Goda, H.M.; Khalifa, E.A.; Abd El Baky, H.H. Antimicrobial and antioxidant activities of leaves and flowers
essential oils of Egyptian Lantana camara L. Der Pharma Chem. 2014, 6, 246–255.
7. Saraf, A.; Quereshi, S.; Sharma, K.; Khan, N.A. Antimicrobial activity of Lantana camara L. J. Exp. Sci. 2011, 2, 50–54.
8. Al-Hakeim, H.K.; Al-Zabibah, R.S.; Alzihari, H.F.; Almensoori, A.K.; Al-Zubaidi, H.A.; Hassan, L.E.A.H. Anticancer and
antiangiogenic activities of alkaloids isolated from Lantana camara by adsorption on the magnetic nanoparticles. Karbala Int. J.
Mod. Sci. 2021, 7, 11. [CrossRef]
9. Sharma, G.P.; Raghubanshi, A.S.; Singh, J.S. Lantana invasion: An overview. Weed Biol. Manag. 2005, 5, 157–165. [CrossRef]
10. Day, M.D.; Zalucki, M.P. Lantana camara Linn. (Verbenaceae). In Biological Control of Tropical Weeds Using Arthropods; Cambridge
University Press: Cambridge, UK, 2009; pp. 211–246.
11. Thomas, S.E. Lantana camara (lantana). In CABI Compendium; CABI Digital Library: Wallingford, UK, 2022. [CrossRef]
12. Goyal, N.; Sharma, G.P. Lantana camara L. (sensu lato): An enigmatic complex. NeoBiota 2015, 25, 15–26.
13. Sanders, R.W. Taxonomy of Lantana Sect. Lantana (Verbenaceae): II. Taxonomic revision. J. Bot. Res. Inst. Texas 2012, 6, 403–441.
14. Urban, A.J.; Simelane, D.O.; Retief, E.; Heystek, F.; Williams, H.E.; Madire, L.G. The invasive “Lantana camara L.” hybrid complex
(Verbenaceae): A review of research into its identity and biological control in South Africa. Afr. Entomol. 2011, 19, 315–348.
[CrossRef]
15. Begum, S.; Zehra, S.Q.; Syed, I.; Hassan, S.I.; Siddiqui, B.S. Noroleanane triterpenoids from the aerial parts of Lantana camara.
Helv. Chim. Acta 2008, 91, 460–467. [CrossRef]
16. Begum, S.; Wahab, A.; Siddiqui, B.S.; Qamar, F. Nematocidal constituents of the aerial parts of Lantana camara. J. Nat. Prod. 2000,
63, 765–767. [CrossRef] [PubMed]
17. Hussain, H.; Hussain, J.; Al-Harrasi, A.; Shinwari, Z.K. Chemistry of some species genus Lantana. Pak. J. Bot. 2011, 43, 51–62.
Molecules 2025, 30, 851 45 of 51

18. Ved, A.; Arsi, T.; Prakash, O.; Gupta, A. A Review on phytochemistry and pharmacological activity of Lantana camara Linn. Int. J.
Pharm. Sci. Res. 2018, 9, 37–43. [CrossRef]
19. Al-Fadhli, A.A.; Nasser, J.A. Constituents from the root of Lantana camara. Asian J. Chem. 2014, 26, 8019–8021. [CrossRef]
20. Madke, G.; Bhutkar, K.; Chaudhari, S.R. Overview on Lantana camara showing various pharmacological benefits. Int. Adv. Res. J.
Sci. Eng. Technol. 2024, 11, 86–92. [CrossRef]
21. Kumar, M.A.; Santoshi, S.; Patil, S.M.; Bhatt, S.P. A review on phytochemical and pharmacological profiling of Lantana camara
Linn. W. J. Pharm. Pharm. Sci. 2023, 12, 15–25.
22. Naphade, V.; Bhamre, J.S.; Avaghade, M.M.; Kale, M.S.; Bendale, A.R.; Borse, S.L.; Borse, L.B. Phytochemical and pharmacological
potential of Lantana Camara Linn. Int. J. Life Sci. Pharma Res. 2023, 13, 70–80. [CrossRef]
23. Shah, M.; Alharby, H.F.; Hakeem, K.R.; Munawar, M. A Comprehensive review on Lantana camara: An important medicinal and
ornamental plant. Life Sci. J. 2024, 21, 10–17.
24. Ghisalberti, E.L. Lantana camara L. (Verbenaceae). Fitoterapia 2000, 71, 467–486. [CrossRef]
25. Beeby, P.J. Chemical modification of triterpenes from Lantana camara. 22β-Ester analogues of lantadene A. Aust. J. Chem. 1978, 31,
1313–1322. [CrossRef]
26. Begum, S.; Raza, S.M.; Siddiqui, B.S.; Siddiqui, S. Triterpenoids from the aerial parts of Lantana camara. J. Nat. Prod. 1995, 58,
1570–1574. [CrossRef]
27. Barua, A.K.; Chakrabarti, P.; Sanyal, P.K.; Das, B.C. Triterpenoids: XXXII. The structure of lantic acid—A new triterpene from Lantana
camara. J. Indian Chem. Soc. 1969, 46, 100–101. [CrossRef]
28. Sharma, O.P.; Sharma, S.; Pattabhi, V.; Mahato, S.B.; Sharma, P.D. A review of the hepatotoxic plant Lantana camara. Crit. Rev.
Toxicol. 2007, 37, 313–352. [CrossRef]
29. Ntalo, M.; Ravhuhali, K.E.; Moyo, B.; Hawu, O.; Msiza, N.H. Lantana camara: Poisonous species and a potential browse species for
goats in southern Africa—A review. Sustainability 2022, 14, 751. [CrossRef]
30. Kalita, S.; Kumar, G.; Karthik, L.; Rao, K.V.B. A review on medicinal properties of Lantana camara Linn. Res. J. Pharm. Technol.
2012, 5, 711–715.
31. Reddy, N.M. Lantana camara Linn. Chemical constituents and medicinal properties: A review. Acad. J. Pharm. 2013, 2, 445–448.
32. Kumar, R.; Katiyar, R.; Kumar, S.; Kumar, T.; Singh, V. Lantana camara: An alien weed, its impact on animal health and strategies
to control. J. Exp. Biol. Agric. Sci. 2016, 4, 321–337. [CrossRef]
33. Al-Snafi, A.E. Chemical constituents and pharmacological activities of Lantana camara—A review. Asian J. Pharm. Clin. Res. 2019,
12, 10–20. [CrossRef]
34. Battase, L.; Attarde, D. Phytochemical and medicinal study of Lantana camara Linn (Verbenaceae)—A review. Asian J. Pharm. Clin.
Res. 2021, 14, 20–27. [CrossRef]
35. Kathuria, I.; Joshi, M.; Patel, B.M.; Dhobi, M. Phytoconstituents of Lantana camara L.: Rekindling hope in the cancer treatment.
Curr. Bioact. Compd. 2022, 18, e311221199740.
36. Kimpouni, V.; Mamboueni, J.C.; Tsoungould, F.G.M.; Mikoko, E.N. Ethnobotanical and phytotherapeutic study from Kouni
community of the sub-prefecture of Kayes (Bouenza—Congo). Heliyon 2019, 5, e02007. [CrossRef] [PubMed]
37. Manya, M.H.; Keymeulen, F.; Ngezahayo, J.; Bakari, A.S.; Kalonda, M.E.; Kahumba, B.J. Antimalarial herbal remedies of Bukavu
and Uvira areas in DR Congo: An ethnobotanical survey. J. Ethnopharmacol. 2020, 249, 112422. [CrossRef] [PubMed]
38. Pathy, K.K.; Flavien, N.B.; Honoré, B.K.; Vanhove, W.; Patrick, V.D. Ethnobotanical characterization of medicinal plants used in
Kisantu and Mbanza-Ngungu territories, Kongo-Central Province in DR Congo. J. Ethnobiol. Ethnomed. 2021, 17, 5.
39. Geremew, Y.A.; Desta, Y.D.; Kumar, P.A.; Tonjo, Z.T. Traditional medicinal plants used by people in Libo-Kemkem District, South
Gondar, Ethiopia. Asian J. Agric. Sci. 2012, 4, 171–176.
40. Lulekal, E.; Kelbessa, E.; Bekele, T.; Yineger, H. An ethnobotanical study of medicinal plants in Mana Angetu District, southeastern
Ethiopia. J. Ethnobiol. Ethnomed. 2008, 4, 10. [CrossRef]
41. Mesfin, F.; Demissew, S.; Teklehaymanot, T. An ethnobotanical study of medicinal plants in Wonago Woreda, SNNPR, Ethiopia.
J. Ethnobiol. Ethnomed. 2009, 5, 28. [CrossRef]
42. Giday, M.; Asfaw, Z.; Woldu, Z. Ethnomedicinal study of plants used by Sheko ethnic group of Ethiopia. J. Ethnopharmacol. 2010,
132, 75–85. [CrossRef]
43. Jonville, M.C.; Kodja, H.; Humeau, L.; Fournel, J.; De Mol, P.; Cao, M. Screening of medicinal plants from Reunion Island for
antimalarial and cytotoxic activity. J. Ethnopharmacol. 2008, 120, 382–386. [CrossRef] [PubMed]
44. Magassouba, F.B.; Diallo, A.; Kouyaté, M.; Mara, F.; Mara, O.; Bangoura, O. Ethnobotanical survey and antibacterial activity of
some plants used in Guinean traditional medicine. J. Ethnopharmacol. 2007, 114, 44–53. [CrossRef] [PubMed]
45. Njoroge, G.N.; Bussmann, R.W. Traditional management of ear, nose and throat (ENT) diseases in Central Kenya. J. Ethnobiol.
Ethnomed. 2006, 2, 54. [CrossRef] [PubMed]
46. Njoroge, G.N.; Bussmann, R.W. Diversity and utilization of antimalarial ethnophytotherapeutic remedies among the Kikuyus
(Central Kenya). J. Ethnobiol. Ethnomed. 2006, 2, 8. [CrossRef] [PubMed]
Molecules 2025, 30, 851 46 of 51

47. Kareru, P.G.; Kenji, G.M.; Gachanja, A.N.; Keriko, J.M.; Mungai, G. Traditional medicines among the Embu and Mbeere people of
Kenya. Afr. J. Tradit. Complement. Altern. Med. 2007, 4, 75–86. [CrossRef] [PubMed]
48. Geissler, P.W.; Harris, S.A.; Prince, R.J.; Olsen, A.; Odhiambo, R.A.; Oketch-Rabah, H. Medicinal plants used by Luo mothers and
children in Bondo district, Kenya. J. Ethnopharmacol. 2002, 83, 39–54. [CrossRef] [PubMed]
49. Nguta, J.M.; Mbaria, J.M.; Gakuya, D.W.; Gathumbi, P.K.; Kiama, S.G. Antimalarial herbal remedies of Msambweni, Kenya.
J. Ethnopharmacol. 2010, 128, 424–432. [CrossRef]
50. Seyoum, A.; Pålsson, K.; Kung’a, S.; Kabiru, E.W.; Lwande, W.; Killeen, G.F. Traditional use of mosquito-repellent plants in
western Kenya and their evaluation in semi-field experimental huts against Anopheles gambiae: Ethnobotanical studies and
application by thermal expulsion and direct burning. Trans. R. Soc. Trop. Med. Hyg. 2002, 96, 225–231. [CrossRef] [PubMed]
51. Rivière, C.; Nicolas, J.P.; Caradec, M.L.; Désiré, O.; Schmitt, A. Les plants médicinales de la région nord de Madagascar une
approche ethnopharmacologique. Ethnopharmacologia 2005, 36, 1–295.
52. Egunyomi, A.; Gbadamosi, I.T.; Osiname, K.O. Comparative effectiveness of ethnobotanical mosquito repellents used in Ibadan,
Nigeria. J. Appl. Biosci. 2010, 36, 2383–2388.
53. Tabuti, J.R.S. Herbal medicines used in the treatment of malaria in Budiope county, Uganda. J. Ethnopharmacol. 2008, 116, 33–42.
[CrossRef]
54. Baana, K.; Angwech, H.; Malinga, G.M. Ethnobotanical survey of plants used as repellents against housefly, Musca domestica L.
(Diptera: Muscidae) in Budondo Subcounty, Jinja District, Uganda. J. Ethnobiol. Ethnomed. 2018, 14, 35. [CrossRef]
55. Kamatenesi, M.M.; Acipa, A.; Oryem-Origa, H. Medicinal plants of Otwal and Ngai Sub Counties in Oyam District, Northern
Uganda. J. Ethnobiol. Ethnomed. 2011, 7, 7. [CrossRef] [PubMed]
56. Gou, Y.; Li, Z.; Fan, R.; Guo, C.; Wang, L.; Sun, H.; Li, J.; Zhou, C.; Wang, C.; Wang, Y. Ethnobotanical survey and evaluation of
traditional mosquito repellent plants of Dai people in Xishuangbanna, Yunnan Province, China. J. Ethnopharmacol. 2020, 262,
113124. [CrossRef]
57. Namsa, N.D.; Mandal, M.; Tangjang, S. Anti-malarial herbal remedies of northeast India, Assam: An ethnobotanical survey.
J. Ethnopharmacol. 2011, 133, 565–572. [CrossRef] [PubMed]
58. Kamaraj, C.; Kaushik, N.K.; Rahuman, A.A.; Mohanakrishnan, D.; Bagavan, A.; Elango, G. Antimalarial activities of medicinal
plants traditionally used in the villages of Dharmapuri regions of South India. J. Ethnopharmacol. 2012, 141, 796–802. [CrossRef]
[PubMed]
59. Rao, M.R.K. Lethal efficacy of phytochemicals as sustainable sources of insecticidal formulations derived from the leaf extracts of
Indian medicinal plants to control Dengue and Zika vector, Aedes aegypti (Dipetra: Culicide). Int. Res. J. Environ. Sci. 2020, 9,
44–54.
60. Kumar, V.P.; Chauhan, N.S.; Padh, H.; Rajani, M. Search for antibacterial and antifungal agents from selected Indian medicinal
plants. J. Ethnopharmacol. 2006, 107, 182–188. [CrossRef] [PubMed]
61. Obico, J.J.; Ragragio, E.M. A survey of plants used as repellents against hematophagous insects by the Ayta people of Porac,
Pampanga province, Philippines. Philipp. Sci. Lett. 2014, 7, 179–186.
62. Lee, C.; Kim, S.Y.; Eum, S.; Paik, J.H.; Bach, T.T.; Darshetkar, A.M. Ethnobotanical study on medicinal plants used by local Van
Kieu ethnic people of Bac Huong Hoa nature reserve, Vietnam. J. Ethnopharmacol. 2019, 231, 283–294. [CrossRef]
63. Hussein, S.; Dhabe, A. Ethnobotanical study of folk medicinal plants used by villagers in Hajjah district—Republic of Yemen.
J. Med. Plants Stud. 2018, 6, 24–30.
64. Delgado-Altamirano, R.; Monzote, L.; Piñón-Tápanes, A.; Vibrans, H.; Fausto Rivero-Cruz, J.; Ibarra-Alvarado, C.; Rojas-Molina,
A. In vitro antileishmanial activity of Mexican medicinal plants. Heliyon 2017, 3, e00394. [CrossRef] [PubMed]
65. González-Esquinca, A.R.; Luna-Cazáres, L.M.; Gispert-Cruells, M.; Schlie-Guzmán, M.A.; Gutiérrez-Jiménez, J. Plantas Medicinales
Zoques: Padecimientos Gastrointestinales y Respiratorios; Colección Jaguar; UNICACH: Tuxtla Gutiérrez, Mexico, 2013; Volume 1,
pp. 39–40.
66. Hernández, T.; Canales, M.; Avila, J.G.; Duran, A.; Caballero, J.; Romo de Vivar, A. Ethnobotany and antibacterial activity of some
plants used in traditional medicine of Zapotitlán de las Salinas, Puebla (México). J. Ethnopharmacol. 2003, 88, 181–188. [CrossRef]
[PubMed]
67. Braga, F.G.; Bouzada, M.L.M.; Fabri, R.L.; Matos, M.; Moreira, F.O.; Scio, E. Antileishmanial and antifungal activity of plants used
in traditional medicine in Brazil. J. Ethnopharmacol. 2007, 111, 396–402. [CrossRef] [PubMed]
68. Van’t Klooster, C.; van Andel, T.; Reis, R. Patterns in medicinal plant knowledge and use in a Maroon village in Suriname. J.
Ethnopharmacol. 2016, 189, 319–330. [CrossRef] [PubMed]
69. Vásquez, J.; Alarcón, J.C.; Jiménez, S.L.; Jaramillo, G.I.; Gómez-Betancur, I.C.; Rey-Suárez, J.P. Main plants used in traditional
medicine for the treatment of snake bites in the regions of the department of Antioquia, Colombia. J. Ethnopharmacol. 2015, 170,
158–166. [CrossRef]
70. Boulogne, I.; Germosén-Robineau, L.; Ozier-Lafontaine, H.; Fleury, M.; Loranger-Merciris, G. TRAMIL ethnopharmalogical
survey in Les Saintes (Guadeloupe, French West Indies): A comparative study. J. Ethnopharmacol. 2011, 133, 1039–1050. [CrossRef]
Molecules 2025, 30, 851 47 of 51

71. Huang, K.F.; Huang, K.W. Constituents from the stems of Lantana camara (III). J. Chin. Med. 2004, 15, 109–114.
72. Juang, F.C.; Chen, Y.F.; Lin, F.M.; Huang, K.F. Constituents from the leaves of Lantana camara (IV). J. Chin. Med. 2005, 16, 149–155.
73. Gebre, Z.; Tariku, Y.; Jayakumar, V. Bio-assay guided isolation of bioactive molecules from chloroform extract of fruits of
Lantana camara (L.). J. Pharmacogn. Phytochem. 2019, 8, 1335–1342. [CrossRef]
74. Misra, L.; Laatsch, H. Triterpenoids, essential oil and photo-oxidative 28 → 13-lactonization of oleanolic acid from Lantana camara.
Phytochemistry 2000, 54, 969–974. [CrossRef] [PubMed]
75. Misra, N.; Sharma, M.; Raj, K.; Dangi, A.; Srivastava, S.; Misra-Bhattacharya, S. Chemical constituents and antifilarial activity of
Lantana camara against human lymphatic filariid Brugia malayi and rodent filariid Acanthocheilonema viteae maintained in rodent
hosts. Parasitol. Res. 2007, 100, 439–448. [CrossRef]
76. Ðậu, N.V.; Huyền, L.T. Các tritecpen oleanan từ cây bông ổi Lantana camara L. Tạp Chí Hóa Học 2009, 47, 144–148.
77. Begum, S.; Wahab, A.; Siddiqui, B.S. Pentacyclic triterpenoids from the aerial parts of Lantana camara. Chem. Pharm. Bull. 2003, 51,
134–137. [CrossRef]
78. Begum, S.; Ayub, A.; Zehra, S.Q.; Siddiqui, B.S.; Choudhary, M.I.; Samreen. Leishmanicidal triterpenes from Lantana camara.
Chem. Biodivers. 2014, 11, 709–718. [CrossRef] [PubMed]
79. Patil, G.; Khare, A.B.; Huang, K.-F.; Lin, F.-M. Bioactive chemical constituents from the leaves of Lantana camara L. Indian J. Chem.
2015, 54, 691–697.
80. Wu, P.; Song, Z.; Wang, X.; Li, Y.; Li, Y.; Cui, J.; Tuerhong, M.; Jin, D.-Q.; Abudukeremu, M.; Lee, D.; et al. Bioactive triterpenoids
from Lantana camara showing anti-inflammatory activities in vitro and in vivo. Bioorg. Chem. 2020, 101, 104004. [CrossRef]
[PubMed]
81. Darwish, R.S.; El-Banna, A.A.; Ghareeb, D.A.; El-Hosseny, M.F.; Seadawy, M.G.; Dawood, H.M. Chemical profiling and unraveling
of anti-COVID-19 biomarkers of red sage (Lantana camara L.) cultivars using UPLC-MS/MS coupled to chemometric analysis,
in vitro study and molecular docking. J. Ethnopharmacol. 2022, 291, 115038. [CrossRef]
82. Begum, S.; Ayub, A.; Siddiqui, B.S.; Fayyaz, S.; Kazi, F. Nematocidal triterpenoids from Lantana camara. Chem. Biodivers. 2015, 12,
1435–1442. [CrossRef] [PubMed]
83. Begum, S.; Zehra, S.Q.; Siddiqui, B.S. Two new pentacyclic triterpenoids from Lantana camara LINN. Chem. Pharm. Bull. 2008, 56,
1317–1320. [CrossRef]
84. Abdjul, D.B.; Yamazaki, H.; Maarisit, W.; Rotinsulu, H.; Wewengkang, D.S.; Sumilat, D.A.; Kapojos, M.M.; Losung, F.;
Ukai, K.; Namikoshi, M. Oleanane triterpenes with protein tyrosine phosphatase 1B inhibitory activity from aerial parts of
Lantana camara collected in Indonesia and Japan. Phytochemistry 2017, 144, 106–112. [CrossRef] [PubMed]
85. Begum, S.; Siddiqui, B.S.; Wahab, A. Two new pentacyclic triterpenoids from the aerial parts of Lantana camara Linn. Heterocycles
2000, 53, 681–687. [CrossRef]
86. Fatope, M.O.; Salihu, L.; Asante, S.K.; Takeda, Y. Larvicidal activity of extracts and triterpenoids from Lantana camara. Pharm. Biol.
2002, 40, 564–567. [CrossRef]
87. Gupta, S.; Kalani, K.; Saxena, M.; Srivastava, S.K.; Agrawal, S.K.; Suri, N. Cytotoxic evaluation of semisynthetic ester and amide
derivatives of oleanolic acid. Nat. Prod. Commun. 2010, 5, 1567–1570. [CrossRef] [PubMed]
88. Ghosh, S.; Das Sarma, M.; Patra, A.; Hazra, B. Anti-inflammatory and anticancer compounds isolated from
Ventilago madraspatana Gaertn., Rubia cordifolia Linn. and Lantana camara Linn. J. Pharm. Pharmacol. 2010, 62, 1158–1166.
[CrossRef] [PubMed]
89. Begum, S.; Zehra, S.Q.; Wahab, A.; Siddiqui, B.S. Triterpenoidal secondary metabolites from Lantana camara LINN. Helv. Chim.
Acta 2006, 89, 1932–1941. [CrossRef]
90. Begum, S.; Zehra, S.Q.; Siddiqui, B.S.; Fayyaz, S.; Ramzan, M. Pentacyclic triterpenoids from the aerial parts of Lantana camara
and their nematocidal activity. Chem. Biodivers. 2008, 5, 1856–1866. [CrossRef] [PubMed]
91. Sharma, O.P.; Singh, A.; Sharma, S. Levels of lantadenes, bioactive pentacyclic triterpenoids, in young and mature leaves of
Lantana camara var. aculeata. Fitoterapia 2000, 71, 487–491. [CrossRef] [PubMed]
92. Sharma, M.; Sharma, P.D. Optimization of lantadenes isolation and preparation of 22β-hydroxyoleanonic. Chem. Nat. Compd.
2006, 42, 357–358. [CrossRef]
93. Sharma, M.; Sharma, P.D.; Bansal, M.P.; Singh, J. Synthesis, cytotoxicity, and antitumor activity of lantadene—A congeners. Chem.
Biodivers. 2007, 4, 932–939. [CrossRef] [PubMed]
94. Sharma, M.; Sharma, P.D.; Bansal, M.P. Lantadenes and their esters as potential antitumor agents. J. Nat. Prod. 2008, 71, 1222–1227.
[CrossRef] [PubMed]
95. Tailor, N.K.; Boon, H.L.; Sharma, M. Synthesis and in vitro anticancer studies of novel C-2 arylidene congeners of lantadenes. Eur.
J. Med. Chem. 2013, 64, 285–291. [CrossRef] [PubMed]
96. Suthar, S.K.; Lee, H.B.; Sharma, M. The synthesis of non-steroidal anti-inflammatory drug (NSAID)-lantadene prodrugs as novel
lung adenocarcinoma inhibitors via the inhibition of cyclooxygenase-2 (COX-2), cyclin D1 and TNF-α-induced NF-κB activation.
RSC Adv. 2014, 4, 19283–19293. [CrossRef]
Molecules 2025, 30, 851 48 of 51

97. Sharma, M.; Sharma, P.D.; Bansal, M.P.; Singh, J. Synthesis and antitumor activity of novel pentacyclic triterpenoid lantadene D.
Lett. Drug Des. Discov. 2007, 4, 201–206. [CrossRef]
98. Delgado-Altamirano, R.; López-Palma, R.I.; Monzote, L.; Delgado-Domínguez, J.; Becker, I.; Rivero-Cruz, J.F.; Esturau-Escofet, N.;
Vázquez-Landaverde, P.A.; Rojas-Molina, A. Chemical constituents with leishmanicidal activity from a pink-yellow cultivar of
Lantana camara var. aculeata (L.) collected in Central Mexico. Int. J. Mol. Sci. 2019, 20, 872. [CrossRef]
99. Ono, M.; Hashimoto, A.; Miyajima, M.; Sakata, A.; Furusawa, C.; Shimode, M. Two new triterpenoids from the leaves and stems
of Lantana camara. Nat. Prod. Res. 2020, 35, 3757–3765. [CrossRef] [PubMed]
100. Sagar, L.; Sehgal, R.; Ojha, S. Evaluation of antimotility effect of Lantana camara L. var. aculeata constituents on neostigmine induced
gastrointestinal transit in mice. BMC Complement. Altern. Med. 2005, 5, 18. [CrossRef]
101. Banik, R.M.; Pandey, D.K. Optimizing conditions for oleanolic acid extraction from Lantana camara roots using response surface
methodology. Ind. Crops Prod. 2008, 27, 241–248. [CrossRef]
102. Verma, S.C.; Jain, C.L.; Nigam, S.; Padhi, M.M. Rapid extraction, isolation, and quantification of oleanolic acid from
Lantana camara L. roots using microwave and HPLC-PDA techniques. Acta Chromatogr. 2013, 25, 181–199. [CrossRef]
103. Pour, M.B.; Latha, L.Y.; Sasidharan, S. Cytotoxicity and oral acute toxicity studies of Lantana camara leaf extract. Molecules 2011, 16,
3663–3674. [CrossRef]
104. Ma, L.-h.; Wu, H.-b.; Wu, H.-b. A new oleanane triterpene with allelopathic activity from Lantana camara. Chem. Nat. Compd. 2023,
59, 709–711. [CrossRef]
105. Ayub, A.; Begum, S.; Ali, S.N.; Ali, S.T.; Siddiqui, B.S. Triterpenoids from the aerial parts of Lantana camara. J. Asian Nat. Prod. Res.
2017, 21, 141–149. [CrossRef] [PubMed]
106. Begum, S.; Wahab, A.; Siddiqui, B.S. Three new pentacyclic triterpenoids from Lantana camara. Helv. Chim. Acta. 2002, 85,
2335–2341. [CrossRef]
107. Kalita, S.; Kumar, G.; Karthik, L.; Rao, K.V.B. Phytochemical composition and in vitro hemolytic activity of Lantana Camara L.
(Verbenaceae) leaves. Pharmacologyonline 2011, 1, 59–61.
108. Ali-Emmanuel, N.; Moudachirou, M.; Akakpo, J.A.; Quetin-Leclercq, J. Treatment of bovine dermatophilosis with Senna alata,
Lantana camara and Mitracarpus scaber leaf extracts. J. Ethnopharmacol. 2003, 86, 167–171. [CrossRef] [PubMed]
109. Shamsee, Z.R.; Al-Saffar, A.Z.; Al-Shanon, A.F.; Al-Obaidi, J.R. Cytotoxic and cell cycle arrest induction of pentacyclic triter-
penoides separated from Lantana camara leaves against MCF-7 cells in vitro. Mol. Biol. Rep. 2019, 46, 381–390. [CrossRef]
[PubMed]
110. Kong, C.H.; Wang, P.; Zhang, C.X.; Zhang, M.X.; Hu, F. Herbicidal potential of allelochemicals from Lantana camara against
Eichhornia crassipes and the alga Microcystis aeruginosa. Weed Res. 2006, 46, 290–295. [CrossRef]
111. Giri, A.G.; Oza, R. Isolation, purification and characterization of triterpenes from Lantana camara. Chem. Sci. Rev. Lett. 2015, 4,
520–525.
112. Sharma, M.; Rakhi, A.; Dalal, N.; Sharma, N. Design, synthesis and evaluation of lantadene A congener with hydroxyl functionality
in ring A as an antitumour agent. Nat. Prod. Res. 2011, 25, 387–396. [CrossRef]
113. Grace-Lynn, C.; Darah, I.; Chen, Y.; Latha, L.Y.; Jothy, S.L.; Sasidharan, S. In vitro antioxidant activity potential of lantadene A, a
pentacyclic triterpenoid of Lantana plants. Molecules 2012, 17, 11185–11198. [CrossRef] [PubMed]
114. Jaafar, N.D.; Al-Saffar, A.Z.; Yousif, E.A. Genotoxic and cytotoxic activities of lantadene A-loaded gold nanoparticles (LA-AuNPS)
in MCF-7 cells: An in vitro assessment. Int. J. Toxicol. 2020, 39, 422–432. [CrossRef] [PubMed]
115. Dawood, D.A.S.; Chua, L.S.; Tan, T.S.; Alshemary, A.F. Apoptotic mechanism of lantadene A from Lantana camara leaves against
prostatic cancer cells. Egypt. J. Chem. 2021, 64, 7503–7510. [CrossRef]
116. Litaudon, M.; Jolly, C.; Le Callonec, C.; Dao, D.C.; Retailleau, P.; Nosjean, O. Cytotoxic pentacyclic triterpenoids from
Combretum sundaicum and Lantana camara as inhibitors of Bcl-xL/BakBH3 domain peptide interaction. J. Nat. Prod. 2009,
72, 1314–1320. [CrossRef]
117. Delgado-Altamirano, R.; Rojas, A.; Esturau-Escofet, N. 1 H and 13 C NMR reassignment of some chemical shifts of lantanilic acid
and camaric acid. Magn. Reson. Chem. 2019, 57, 320–325. [CrossRef] [PubMed]
118. Qamar, F.; Begum, S.; Raza, S.M.; Wahab, A.; Siddiqui, B.S. Nematocidal natural products from the aerial parts of Lantana camara
Linn. Nat. Prod. Res. 2005, 19, 609–613. [CrossRef]
119. Kartika, M.Z.; Suryati, S.; Efdi, M. A triterpenoid compound from the leaves of Tahi ayam (Lantana camara Linn). Indones. J.
Fundam. Appl. Chem. 2018, 3, 18–22.
120. Seepe, H.A.; Raphoko, L.; Amoo, S.O.; Nxumalo, W. Lantadene A and boswellic acid isolated from the leaves of Lantana camara L.
have the potential to control phytopathogenic Fusarium species. Heliyon 2022, 8, e12216. [CrossRef] [PubMed]
121. Begum, S.; Zehra, S.Q.; Ayub, A.; Siddiqui, B.S. A new 28-noroleanane triterpenoid from the aerial parts of Lantana camara Linn.
Nat. Prod. Res. 2010, 24, 1227–1234. [CrossRef] [PubMed]
122. Ediruslan, E.; Manjang, Y.; Suryati, S.; Aziz, H. Structure elucidation of brine shrimp toxic compound from Lantana camara L.
leaves. J. Chem. Pharm. Res. 2015, 7, 250–255.
Molecules 2025, 30, 851 49 of 51

123. Suryati; Efdi, M.; Oktavia, S.H.; Aziz, H. Antibacterial and antifungal activity of lantanilic acid isolated from Lantana camara Linn.
Der Pharma Chem. 2017, 9, 105–108.
124. Suryati, S.; Mardiah, E.; Efdi, M.; Kartika, M.Z.; Sari, Y.M. Bioactivity of compounds isolated from the leaves of the Lantana camara
Linn plant. J. Chin. Pharm. Sci. 2019, 28, 360–368.
125. Kazmi, I.; Rahman, M.; Afzal, M.; Gupta, G.; Saleem, S.; Afzal, O.; Shaharyar, M.A.; Nautiyal, U.; Ahmed, S.; Anwar, F. Anti-
diabetic potential of ursolic acid stearoyl glucoside: A new triterpenic glycosidic ester from Lantana camara. Fitoterapia 2012, 83,
142–146. [CrossRef] [PubMed]
126. Kazmi, I.; Saleem, S.; Ahmad, T.; Afzal, M.; Al-Abbasi, F.A.; Kumar, V.; Anwar, F. Protective effect of oleane-12-en-3β-ol-28-oic
acid 3β-D-glucopyranoside in ethanol induced gastric ulcer by enhancing the prostaglandin E2 level. J. Ethnopharmacol. 2018, 211,
394–399. [CrossRef] [PubMed]
127. Yadav, S.B.; Tripathi, V. A new triterpenoid from Lantana camara. Fitoterapia 2003, 74, 320–321. [CrossRef]
128. Sathish, R.; Vyawahare, B.; Natarajan, K. Antiulcerogenic activity of Lantana camara leaves on gastric and duodenal ulcers in
experimental rats. J. Ethnopharmacol. 2011, 134, 195–197. [CrossRef] [PubMed]
129. Begum, S.; Wahab, A.; Siddiqui, B.S. Ursethoxy acid, a new triterpene from Lantana camara. Nat. Prod. Lett. 2002, 16, 235–238.
[CrossRef] [PubMed]
130. Yadav, S.B.; Tripathi, V. Chemical components of Lantana camara Linn. Indian J. Heterocyclic Chem. 2000, 10, 71–72.
131. Xavier, M.R.; da Fonseca, A.M.; Gonçalves Cruz, B.; dos Santos Mendes, A.M.; Oliveira, L.S.; Bandeira, P.N.; Colares, R.P.;
Braz-Filho, R.; Marinho, E.S.; Freitas, T.S.; et al. Modulating antibacterial activity against multidrug-resistant Escherichia coli and
Staphylococcus aureus of the flavonoid pectolinarin isolated from Lantana camara leaves. J. Anal. Pharm. Res. 2021, 10, 217–220.
[CrossRef]
132. El-Kassem, L.T.A.; Mohammed, R.S.; El Souda, S.S.; El-Anssary, A.A.; Hawas, U.W.; Mohmoud, K. Digalacturonide flavones
from Egyptian Lantana camara flowers with in vitro antioxidant and in vivo hepatoprotective activities. Z. Naturforsch. C 2012, 67,
381–390. [CrossRef] [PubMed]
133. Qureshi, H.; Anwar, T.; Ali, Q.; Haider, M.Z.; Habib, N.; Fatima, S. Isolation of natural herbicidal compound from Lantana camara.
Int. J. Environ. Anal. Chem. 2019, 101, 631–638. [CrossRef]
134. Begum, S.; Wahab, A.; Siddiqui, B.S. Antimycobacterial activity of flavonoids from Lantana camara Linn. Nat. Prod. Res. 2008,
22, 467–470. [CrossRef]
135. Benjamin, R.; Akilan, A.; Senguttuvan, S. Isolation, characterization, and anti-cancer activity of kaempferol-6-methoxy-7-O-
glucoside from Lantana camara flowers. Orient. J. Chem. 2022, 38, 72–76. [CrossRef]
136. Millycent, S.A.; Mwonjoria, M.; Juma, K.K.; Ngugi, M.P.; Njagi, E. Evaluation of analgesic, anti-inflammatory and toxic effects of
Lantana camara. Int. J. Phytopharm. 2017, 8, 89–97.
137. Naz, R.; Bano, A. Phytochemical screening, antioxidants and antimicrobial potential of Lantana camara in different solvents. Asian
Pac. J. Trop. Dis. 2013, 3, 480–486. [CrossRef]
138. Indrayani, F.; Wirastuty, R.Y. In-vitro anti-tuberculosis activity and phytochemical screening of Lantana (Lantana camara L.) flower.
Pharm. J. Indones. 2021, 18, 413–421. [CrossRef]
139. Mahdi-Pour, B.; Jothy, S.L.; Latha, L.Y.; Chen, Y.; Sasidharan, S. Antioxidant activity of methanol extracts of different parts of
Lantana camara. Asian Pac. J. Trop. Biomed. 2012, 2, 960–965. [CrossRef]
140. Clarkson, C.; Maharaj, V.J.; Crouch, N.R.; Grace, O.M.; Pillay, P.; Matsabisa, M.G. In vitro antiplasmodial activity of medicinal
plants native to or naturalised in South Africa. J. Ethnopharmacol. 2004, 92, 177–191. [CrossRef] [PubMed]
141. Sharma, M.; Sharma, P.D.; Bansal, M.P. Chemopreventive effect of Lantana camara leaf extract on 7,12-dimethylbenz[a]anthracene-
induced squamous cell carcinoma of skin in Swiss albino mice. Pharm. Biol. 2007, 45, 145–148. [CrossRef]
142. Arbiastutie, Y.; Marsono, D.; Hartati, M.S.; Purwanto, R. The potential of understorey plants from Gunung Gede Pangrango
National Park (West Java, Indonesia) as cervixs anticancer agents. Biodiversitas 2017, 18, 109–115. [CrossRef]
143. Ruslin; Yamin; Rahma, N.A.; Imawati; Rohman, A. UPLC MS/MS profile and antioxidant activities from nonpolar fraction of
patiwala (Lantana camara) leaves extract. Separations 2022, 9, 75. [CrossRef]
144. Erdoğan, P.; Üstündağ, S. Lantana camara Ekstraktının Patates güvesi [Phthorimae operculella (Zeller) (Lepidoptera: Gelechiidae)]’ne
İnsektisit Etkileri. Int. J. Sivas Univ. Sci. Technol. 2022, 1, 63–75.
145. Kamaraj, C.; Rahuman, A.A.; Bagavan, A.; Elango, G.; Zahir, A.A.; Santhoshkumar, T. Larvicidal and repellent activity of
medicinal plant extracts from Eastern Ghats of South India against malaria and filariasis vectors. Asian Pac. J. Trop. Med. 2011,
4, 698–705. [CrossRef] [PubMed]
146. Verma, R.K.; Verma, S.K. Phytochemical and termiticidal study of Lantana camara var. aculeata leaves. Fitoterapia 2006, 77, 466–468.
[CrossRef]
147. Barre, J.; Jenber, A.J. Evaluation of selected botanicals for the management of maize weevil (Sitophilus zeamais) on maize
(Zea mays L.) grain under laboratory condition in Gabilay District, Somaliland. Heliyon 2022, 8, e11859. [CrossRef]
Molecules 2025, 30, 851 50 of 51

148. Kayesth, S.; Gupta, K.K. Impact of Lantana camara hexane extract on survival, growth and development of Dysdercus koenigii
Fabricius (Heteroptera: Pyrrhocoriedae). Acta Ecol. Sin. 2018, 38, 187–192. [CrossRef]
149. Alghamdi, A.A.; Basher, N.S.H. Efficacy of leaves and flowers ethanol extracts of the invasive species Lantana camara Linn as a
mosquito larvicidal. Int. J. Mosq. Res. 2020, 7, 43–47.
150. Hemalatha, P.; Elumalai, D.; Janaki, A.; Babu, M.; Velu, K.; Velayutham, K. Larvicidal activity of Lantana camara aculeata against
three important mosquito species. J. Entomol. Zool. Stud. 2015, 3, 174–181.
151. Sharma, M.; Alexander, A.; Nakhate, K.T.; Nagwanshi, K.K.; Ajazuddin. Evaluation of the mosquito larvicidal potential and
comparative assessment of the juice of Lantana camara Linn and Ocimum gratissimum Linn. Exp. Parasitol. 2023, 249, 108521.
[CrossRef]
152. Kamatchi, P.A.C.; Maheswaran, R.; Ignacimuthu, S. Evaluation of larval toxicity of Lantana camara L. and Catharanthus roseus L.
against Culex quinquefasciatus say and Aedes aegypti L. Entomol. Ornithol. Herpetol. 2016, 5, 170.
153. Taha, K.A.; Osman, H.E.; Sidahmed, O.A.A. Larvicidal effects of some plant extracts against Anopheles arabiensis Patton Larvae
(Diptera: Culicidae). J. Sci. Technol. 2011, 12, 67–74.
154. Gindri, D.M.; Coelho, C.M.M.; Uarrota, V.G.; Rebelo, A.M. Herbicidal bioactivity of natural compounds from Lantana camara on
the germination and seedling growth of Bidens pilosa. Pesqui. Agropecu. Trop. 2020, 50, e57746. [CrossRef]
155. Tuyiringire, N.; Taremwa Mugisha, I.; Tusubira, D.; Munyampundu, J.P.; Mambo Muvunyi, C.; Vander Heyden, Y. In vitro
antimycobacterial activity of medicinal plants Lantana camara, Cryptolepis sanguinolenta, and Zanthoxylum leprieurii. J. Clin. Tuberc.
Other Mycobact. Dis. 2022, 27, 100307. [CrossRef] [PubMed]
156. Bangou, M.J.; Almaraz-Abarca, N.; Méda, N.T.R.; Zeba, B.; Kiendrebéogo, M.; Millogo-Rasolodimby, J. Polyphenolic composition
of Lantana camara and Lippia chevalieri, and their antioxidant and antimicrobial activities. Phytomedicine 2012, 4, 115–124.
157. Pradeep, B.V.; Tejaswini, M.; Nishal, P.; Pardhu, G.; Shylaja, S.; Kumar, K.C. Phytochemical screening and antimicrobial activities
of plant extract of Lantana camara. J. Environ. Biol. 2013, 34, 645–649.
158. El Gendy, S.N.; Ezzat, M.I.; EL Sayed, A.M.; Saad, A.S.; Elmotayam, A.K. HPLC-PDA-ESI-MS-MS analysis of acids content of
Lantana camara L. flower extract and its anticoagulant activity. Egypt. J. Chem. 2023, 66, 249–256.
159. Mengistu, G.; Engidawork, E.; Nedi, T. Evaluation of the antidiarrhoeal activity of 80% methanol extract and solvent fractions of
the leaves of Lantana camara Linn (Verbenaceae) in mice. Ethiop. Pharm. J. 2015, 31, 107–120. [CrossRef]
160. Sousa, E.O.; Miranda, C.M.B.A.; Nobre, C.B.; Boligon, A.A.; Athayde, M.L.; Costa, J.G.M. Phytochemical analysis and antioxidant
activities of Lantana camara and Lantana montevidensis extracts. Ind. Crops Prod. 2015, 70, 7–15. [CrossRef]
161. Suryati; Santoni, A.; Kartika, M.Z.; Aziz, H. Antioxidant activity and total phenolic content of ethyl acetate extract and fractions
of Lantana camara L. leaf. Der Pharma Chem. 2016, 8, 92–96.
162. Kalita, S.; Kumar, G.; Karthik, L.; Rao, K.V.B. In vitro antioxidant and DNA damage inhibition activity of aqueous extract of
Lantana camara L. (Verbenaceae) leaves. Asian Pac. J. Trop. Biomed. 2012, 2, 1675–1679. [CrossRef]
163. Kazmi, I.; Afzal, M.; Ali, B.; Damanhouri, Z.A.; Ahmaol, A.; Anwar, F. Anxiolytic potential of ursolic acid derivative-a stearoyl
glucoside isolated from Lantana camara L. (Verbanaceae). Asian Pac. J. Trop. Med. 2013, 6, 433–437. [CrossRef]
164. Parimoo, H.A.; Sharma, R.; Patil, R.D.; Sharma, O.P.; Kumar, P.; Kumar, N. Hepatoprotective effect of Ginkgo biloba leaf extract
on lantadenes-induced hepatotoxicity in guinea pigs. Toxicon 2014, 81, 1–12. [CrossRef] [PubMed]
165. Veraplakorn, V. In vitro micropropagation and allelopathic effect of lantana (Lantana camara L.). Agric. Nat. Resour. 2017, 51,
478–484. [CrossRef]
166. Motwani, G.S.; Golani, N.; Solanki, H.A. Allelopathic effects of aqueous leaf leachates of Lantana camara on Eichhornia crassipes.
Life Sci. Leaf 2013, 1, 83–90.
167. Saxena, M.K.; Gupta, J.; Singh, N. Allelopathic potential of callus extract of Lantana camara. Int. J. Recent Sci. Res. 2013, 4,
1628–1630.
168. Veraplakorn, V. Allelopathic hormesis and slow release of lantana (Lantana camara L.) callus extract. Agric. Nat. Resour. 2018, 52,
335–340. [CrossRef]
169. Nayak, B.S.; Raju, S.S.; Eversley, M.; Ramsubhag, A. Evaluation of wound healing activity of Lantana camara L.—A preclinical
study. Phytother. Res. 2009, 23, 241–245. [CrossRef]
170. Suryati, A.S.; Imelda, R.V.U. Cytotoxic and molecular docking potential of β-sitosterol isolated from Lantana camara leaves against
breast (T47D) and cervical cancer (HeLa) cell lines. Trop. J. Nat. Prod. Res. 2024, 8, 6911–6917.
171. Wang, J.; Lu, X.; Jing, Q.; Zhang, B.; Ye, J.; Zhang, H.; Xiao, Z.; Zhang, J. Spatiotemporal characterization of heavy metal and
antibiotics in the Pearl River Basin and pollutants removal assessment using invasive species-derived biochars. J. Hazard. 2023,
454, 131409. [CrossRef]
172. Surya Prakash, V.; Radhakrishnan, N.; Vasantha-Srinivasan, P.; Veeramani, C.; El Newehy, A.S.; Alsaif, M.A.; Al-Numair, K.S. In
silico analysis of selected nutrition rich fruit of Bunch berry (Lantana camara) constituents as human acetylcholinesterase (hAchE),
carbonic anhydrase II (hCA-II) and carboxylesterase 1 (hCES-1) inhibitory agents. Saudi J. Biol. Sci. 2023, 30, 103847. [CrossRef]
Molecules 2025, 30, 851 51 of 51

173. Patil, S.P.; Sahu, K.G.; Andhale, G.S. Lantana camara mediated green synthesis of metallic nanoparticles: A mini review. Inorg.
Chem. Commun. 2024, 160, 111853. [CrossRef]
174. Hussein, L.M.; Hasan, A.Y. Biosynthesis of zinc oxide nanoparticles by Lantana camara flowers extract characterization and their
antibacterial potential against pathogenic bacteria isolated from wounds infections. J. Pharm. Negat. 2022, 13, 451–458.
175. Hamdiani, S.; Shih, Y.F. Environmentally methods for the loaded of silver and zinc oxide nanoparticles on diatomite as a potential
nanofiller for industrial application. AIP Conf. Proc. 2023, 2619, 040008.
176. Negi, A.; Joshi, S.K.; Bhandari, N.S. Estimation of sorption-desorption characteristics of biosorbent of Lantana camara leaves for
removal of Pb (II) ions from wastewater. Environ. Monit. Assess. 2023, 195, 42. [CrossRef] [PubMed]

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.

You might also like