Open access peer-reviewed chapter

Plant-Derived Compounds against Microbial Infections and Cancers

Written By

Gabin Thierry M. Bitchagno, Vaderament-A. Nchiozem-Ngnitedem, Nadine Tseme Wandji, Guy Cedric T. Noulala, Serge Alain T. Fobofou and Bruno Ndjakou Lenta

Submitted: 10 February 2020 Reviewed: 19 April 2020 Published: 04 June 2020

DOI: 10.5772/intechopen.92596

From the Edited Volume

Bioactive Compounds in Nutraceutical and Functional Food for Good Human Health

Edited by Kavita Sharma, Kanchan Mishra, Kula Kamal Senapati and Corina Danciu

Chapter metrics overview

843 Chapter Downloads

View Full Metrics

Abstract

Plants synthesize and preserve a variety of metabolites known as natural products. Many of them are easily extractable and can be used as starting material or chemical scaffolds for various purposes, especially in drug discovery. Numbers of reports have listed valuable candidates with privilege scaffolds currently in active development as drugs. New compounds with anticancer and antiinfective activities have been discovered recently, some presented these backbones. The present book chapter aims to highlight these findings from plants which can be considered valuable for the development of new drugs against malignant cells and infective diseases. Interest in anti-infective agents is increasing due to the resistance of microorganisms to existing drugs and newly emerging infectious diseases. This resistance is also, nowadays, associated to some forms of cancers. In addition, the value of plants as essential part in the health care pipeline in low- and middle-income countries is under consideration even though these countries are almost all surrounded by a rich and untapped biodiversity. People are always relying on “modern drugs and treatment” which is unfortunately not affordable to all. Therefore, the present compilation of data on plant-derived compounds can inspire the formulation of ameliorated traditional medicines (ATM) against the targeted diseases and the conservation of species.

Keywords

  • phytoconstituents
  • anticancer
  • antimicrobial
  • biological cutoff points
  • sesquiterpenoid lactones
  • phenolic compounds

1. Introduction

1.1 General statement

As any other organisms on Earth, plants are said to possess multi-functional properties. They constitute feedstock materials to feed people and are reputed for their uses in medicines [1, 2]. History of plants has been always related to that of Human. Reports said Human have always insured their primary health care by using plants [3, 4, 5]. Even with the discovery of technology leading to synthetic drugs with sometimes more efficiency, plants still remain ubiquitous and safe for health concerns.

Research currently overflows in the literature related to the chemistry and biology of plants. Interests focus on experimental validation of ethnopharmacological uses of certain herb and formulation of plant extracts for a sustainable health care [1, 2, 3, 4, 5]. Therefore, plants are ground, exhausted and evaluated for various biological activity including properties to inhibit the growth of or to kill microorganisms and tumor cell lines. However, both microorganism and cancer cells become more and more resistant and remain serious threats for life. As an example, resistance to penicillin used for the treatment of lung infection ranged from 0 to 51% around the World and between 8 and 65% Escherichia coli associated with urinary tract infections presented resistance to ciprofloxacin, another antibiotic (https://www.who.int/health-topics/antimicrobial-resistance). WHO took some measures to diagnose and eradicate the issue but the problem is still actual and present.

More than half of existing antibiotics and anticancers are from synthesis of which almost a quarter takes its origin in natural substances isolated from plants, marine organisms and microorganisms [6]. Nevertheless, plant supply extracts continue to play a relevant role in human beings daily life. Up to date data show that plant extracts are reputed in food science where they are used as dietary supplements [6, 7, 8]. This practice is prevalent in Europe and North America where the interest in plants and related materials is rising up. Despite the progress made in the field of the synthesis of active principles for the formulation of medicaments, people still rely on natural occurring drugs due to their safety and uniqueness. The list of valuable substances from plants cannot be exhaustive.

In ancient time, the discovery of salicin, an ortho-O-glucopyranosylphenylethanol, from Salix alba led to the development of the reputed anti-inflammatory agent aspirin [9, 10]. Morphine, a benzylisoquinoline alkaloid isolated from Papaver somniferum, is a painkiller quite known in medicine and which also exist under its derivatives, heroin and codeine [9, 10]. Another alkaloid namely quinine isolated from Cinchona succirubra has been for long employed to cure malaria and fever related ailments but since 2004, almost all antimalarial drugs in the markets is made up of artemisinin isolated from the Chinese medicinal plant Artemisia annua. Artemisinin is commercialized under various acronyms including arteether, artesunate or artemether [10, 11]. In other hand, the chemotherapy of breast cancer uses the drug taxol which is the commercial name for paclitaxel, a diterpene isolated from Taxus brevifolia. Other compounds like ingenol-3-angelate, from Euphorbia peplus, known under the acronym Ingenol mebutate or the L-histidine-derived alkaloid pilocarpine found in Pilocarpus jaborandi are also some drugs used against other form of cancer [12, 13, 14].

Days after days, we keep discovering the deeply wealth of our surrounded nature. Reports abound in the literature especially on valuable natural compounds in drug development [6, 7, 8]. Most of them highlight natural product scaffolds as building blocks to the development of other compounds through synthesis [6, 7, 8]. A list of priority backbones has even been proposed to lead the development of new drugs [6].

However, interest in health care could also be to find out natural occurring compounds with considerable effects and low toxicity which can be introduced in the actual pipelines of treatment of a disease. That is, the sensibility of the found natural substance is not strong enough to compete commercial drugs but could be proposed alongside prescribed medicines because of its safety and availability. This can actually help in low- or middle-income countries to face certain diseases and build up a sustainable health care system. One can question how useful was the discovery of artemisinin for indigenous people if they have to wait years for pharmaceuticals companies to manufacture the drugs before its consumption. The same interrogation can also be valid to other discovery from plants, e.g. taxol, michellamine B or vinblastine. The plant sources of these active substances are known but still people from villages are waiting for “modern medicines” to take care of their respective health problems.

1.2 Problematic of microbial infections nowadays: drug resistance and climate change

According to WHO microbial infections are said to be the second cause of death globally, with low- and middle-income countries bearing the greatest burden. They include bacteria or fungi but viruses and protozoa diseases are also listed in this category (https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance). Their origin preceded that of human life on earth [15]. In fact, human comes from successive mutations and evolution of bacteria [15]. Our body is made up of more than 100 trillion bacteria [16, 17, 18]. Some of them are useful in human life where they played a critical role in metabolism. However, greater percentage of them has been found to be harmful. By analogy to what is being said by believers, “you are dust and will return to dust,” one can also argue that “you are bacteria and will return bacteria.”

A lot of concerted effort has been put forward since the existence of mankind in trying to understand the biology of infective pathogens and their control. Some success has been achieved although there still more room for further research on this area. Through our constant manipulation and uses of these pathogens together with huge amount of chemicals, including drugs, we end up developing “new organisms” with different properties compared to their natural counterparts. In fact, the original pathogens start developing resistance to the drugs that were previously used for their eradication, making the problem worse [19, 20, 21, 22].

The question of resistance of pathogens to commercialized drugs relies on the living environment of these small organisms. A misconception of bacteria considered that they exist as individual organisms [23]. However, things are different. Bacteria accumulated in colonies to live. They generally stick on a surface and gathered to survive together [23, 24]. Such a constitution known as biofilm is made up of bacteria somehow wrapped in a certain liquid (extracellular matrix) with strange properties. The entire constitution acts as a safety membrane for bacteria. The so-described making-up of this living organism constitutes the first barrier to bacteria and therefore the first stage of resistance [25, 26]. Biofilm are quite distributed in hospitals and nursing homes. They are claimed in household and industrial pipes, biomaterials such as contact lenses, medical devices including implants and urinary catheters, as well as plant and animal tissues. Cases of bacteria resistance have exploded this last decade especially in those zones [23, 24, 25, 26]. This includes bacteria like Acinetobacter, Pseudomonas and various Enterobacteriaceae (Klebsiella, E. coli, Serratia and Proteus) which cause severe and often deadly infections such as bloodstream infections and pneumonia. Bacteria are carried over by devices such as ventilator systems and blood stream catheters. In high-income countries, 7% of all hospitalized people will contract some form of infection, including one in three people in intensive care units. In low- and middle-income countries, this figure rises to at least 10% of hospitalized people, and up to half of people in intensive care units, said the WHO (https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance).

The World is currently facing an unprecedented rising of temperature, leading to certain change in habits and behavior. Discussion are mainly focused on how it could have an impact on human life, as it’s getting colder and colder when it is the cold season or more and more hot when it is the hot season or even hot when it is supposed to be cold and vice versa. Politics in every country recommend adopting new habits to stop the rising up of the earth temperature. In the meantime, it seems that no one is caring about changes occurring at microorganism scale. It comes out that changes occurring in microorganisms due to climate change are not so important for us. And yet we should beware at least, the occurrences of the new viruses Ebola and Corona these last years should make us change our mind on how the World is changing. New microorganisms would be discovered, and the existing microorganisms could mutate to more harmful organisms. Fortunately, almost the same changes are also expected in plant kingdom even though the global warming and deforestation contribute also to plant extinctions. It is also awaited that new metabolites are being made as a result of new biosynthetical routes. These metabolites could either be directly active against pathogenic microorganisms or inspire new synthetical routes in laboratories to reach new drugs and medicines.

1.3 Standard antibacterial and anticancer cutoff points

Discussions are ongoing in the literature between scientists to clearly established standards for a substance to be considered for further steps in drugs development. Established standards are rare or inaccessible. The Kuete’s group proposed standards of evaluating antimicrobial or anticancer properties of secondary metabolites derived from plants. They established that the antimicrobial activity of a crude extract can be considered significant when it MIC is below 100 μg/mL, moderate when between 100 and 625 μg/mL and low when more than 625 μg/mL. For pure compounds, the activity is considered significant when the MIC is below 10 μg/mL, moderate when between 10 μg/mL < MIC<100 μg/mL or low when greater than 100 μg/mL. One can notice that these standards are not considering the MIC of standard commercial drug references. We are not saying that the comparison of obtained MIC with those of the commercial drug is not necessary but the abovementioned cut-off points precise the ranges for a plant substance to be considered as valuable in drug discovery. Likewise, an extract is said to possess a good cytotoxicity if the IC50 values are below 4 μg/mL, modererate when 4 < IC50 < 20 μg/mL/10 < IC50 < 50 μM and low with IC50 above 100 μg/mL (250 μM) [14, 15]. While for cancer cell lines the activity of the pure compound is consider strong when IC50 < 10 μM [20, 21].

Another point of constant intensive discussion remains the relative low activity of most plant extracts and related constituents against microbial and cancer strains. Their activities are sometimes hundred- or thousand-fold less than the sensitivity of existing drugs. Some scientists find these activities not significant enough to be considered for clinical trials as a phytochemical substance should show comparable sensitivity with the commercial drugs. However, knowing that most cancer treatments are based on chemotherapy which is, as known, as harmful as the disease, can natural substances replace synthetic compounds? Likewise, one can also question infectious disease treatments in the same words. In what extent can we clearly consider a plant based products in drug development with respect to their activity against strains of bacteria or tumor cell lines? As mentioned above, the objective is not to compete with existing drugs developed with many expenditures but simply to select valuable extracts and phytoconstituents which could be used alongside actual treatments because of their safety and availability status especially for less developing countries.

1.4 World sustainable development goals related to the field

In September 2015, the UN adopted a list of 17 goals for a better life on the planet with emphasis on the quality of life for posterity (https://sustainabledevelopment.un.org/?menu=1300). Globally it is recognized that ending poverty and other deprivations must go hand-in-hand with strategies that improve health and education, reduce inequality, and spur economic growth, by tackling climate change and working to preserve our oceans and forests are essential for our future as human beings. In the health sector, people should commit themselves to promote healthy lively hood and well-being for all at all ages. These are objectives stated in the Sustainable Development Goal number 3 of the list. Targets within this goal include ending the preventable deaths of newborns and children and ensuring access to effective medicines to all.

However, we are still living in a place where basic infections (malaria, typhoid, diarrhea, cholera and others) can cause death; a World where medicines are too expensive and inaccessible to everyone; a region where people have to walk more than 10 Km to expect treatment in a hospital or a World with increasing political and economic crisis. Nowadays, one should also highlight the increasing resistance of microbes and other pathogens to existing drugs and the occurrence of new strains of bacteria and virus. The former has been related to the overuse and misuse of drugs which modify the living pathogens environment making them used to it, thus developing tolerances to the used drugs.

One of the alternatives to tackle these challenging issues remain natural remedies and drugs. Many sources are being investigated but plants remain the most exploited. Substances from plants are quantitative, affordable, reachable and biologically recognized and easily metabolized by other organisms. They are environmentally friendly and can thus be promoted ever. Numbers of reports are available in the literature, highlighting the antimicrobial and anticancer properties of phytoextracts and products. Extracts can then be standardized and proposed to our fellow population to alleviate the cost of various and diverse drugs available in the markets.

1.5 Rationale of this survey

The present research literature aims to review recent plant compounds reported for their anticancer or antimicrobial properties which constitute valuable candidates to drug development. Our survey covers research reported from 2010. We only listed compounds with MICs or IC50s < 10 μg/mL for a molarity scale ranging from 10 6 –20 μM. Activities of extracts were not highlighted herein. Both sensitive and resistant strains were checked out without restriction.

Advertisement

2. Plant-based secondary compounds with antimicrobial properties

Infective diseases are one of the most common illnesses in the World. They are currently the main concern on earth due to the ongoing Coronavirus (Covid-19) outbreak. Some pathogens spread out in animals and much of them are not known so far. But, at one moment or at another, due to our growing familiarization with wild animals, pathogens can spread within Human kingdom. Research are constantly been done to contain the diseases and come over the pathogens. Most of them are based on drug discovery, one of the oldest fields of Human concern so far. Plants constitute the main source of drugs although interests have moved to bioactive microbial constituents in the last decades mainly against microbial infectious. Owing to the rich biodiversity in our planet, the search for bioactive compounds from untapped natural resources is among the important ongoing projects.

One of the main constituents of plants with pronounced therapeutic interests against infective diseases are volatile oil. They are found in almost every organ of a plant but are said to be present in high extent in fruits and seeds. The composition of essential oil consists of monoterpenes and sesquiterpenes paired with aromatic compounds and lightweight esters, fatty acids, alcohols, ketones and aldehydes. Some examples include γ-terpinene, carvacrol, p-cymene, thymol, linalool, α-terpinene, limonene, eucalyptol, geranyl propionate and α- and β-pinene [27]. Owing to their high hydrophobicity, essential oil are said to impair the cell membrane of microbes, increase their membrane permeability and decrease their cytoplasmic pH [28]. The so-described abilities explained their significant activity against bacteria and fungi including resistant strains like Staphylococcus sp. and Pseudomonas sp. with MIC values approaching 0.01 μg/mL [27]. Volatile oil play also an essential role in protecting and even preventing biofilm development which is very important as presented above [29]. However, the same lipophilicity capacity of essential oil, relevant for their good antiinfective properties, constitutes also their main bottlenecks in drug development because essential oil present a low bioavailability. But when isolated, some of their constituents are water soluble e.g. 1.25 mg/mL at 25°C for carvacrol (https://pubchem.ncbi.nlm.nih.gov/compound/carvacrol#section=Solubility) or 1.59 mg/mL at 25°C (https://pubchem.ncbi.nlm.nih.gov/compound/6549) for linalool and are being studied and used as excipient in drug formulation. Essential oil are however reputed in therapies which promoted local application, inhalation or bath modes of treatment like aromatherapy [27, 56]. They are associated to numerous of ailments including depression, indigestion, headache, insomnia, muscular pain, respiratory problems, skin ailments, swollen joints or urine complications [56]. Unlike essential oil, secondary metabolites mainly found in the solid part of a plant extract can present significant activities with considerable bioavailability and hence, constitute the main research object in natural product domain.

Since 2010 at least 44 metabolites have been reported with MIC values below 10 μg/mL. Phenolic compounds (118, 2026, 2835, 41) were the group of compounds mostly active among the metabolites found. Besides, benzophenanthridine (27), steroids (3637, 40), pentacyclic triterpenoids (3839, 4344) and diterpenoids (19, 42) have been found active against various infective strains (Table 1 and Figure 1). Some of the strains studied are among the microbes listed by WHO as highly harmful and needed new drugs.

Family Species (part) Compound name Test microorganisms (MIC in μg/mL) Refs.
Guttiferae Garcinia mangostana Mangostin A (1) MRSA (6.25 μg/mL), VRE (3.13 μg/mL) [30]
Garcinia cowa (fruits) B. cereus (0.5 μg/mL), B. subtilis (0.25 μg/mL), M. luteus (1.0 μg/mL) [31]
Garcinia mangostana Mangostin Y (2) MSSA (6.25 μg/mL), MRSA (3.13 μg/mL), VRE (6.25 μg/mL), VSE (6.25 μg/mL) [30]
Garcinia cowa (stem barks) Cowanol (3) MRSA SK1 (2 μg/mL), S. aureus (8 μg/mL) [32]
Cowagarcinone E (4) MRSA SK1 (8 μg/mL)
Garciniacowone (5) MRSA SK1 (2 μg/mL), S. aureus (2 μg/mL)
Cowanin (6) MRSA SK1 (4 μg/mL)
Garcinia cowa (fruits) B. subtilis (4 μg/mL), M. luteus (4 μg/mL) [31]
Garcinia cowa (fruits) Garcicowanone A (7) B. cereus (0.25 μg/mL), B. subtilis (2 μg/mL), M. luteus (4 μg/mL),
9-Hydroxycalabaxanthone (8) B. cereus (8 μg/mL), B. subtilis (2 μg/mL), M. luteus (4 μg/mL),
B-mangostin (9) B. cereus (0.25 μg/mL), B. subtilis (4 μg/mL)
Cowagarcinone E (10) B. cereus (4 μg/mL), B. subtilis (4 μg/mL), M. luteus (8 μg/mL)
Rubraxanthone (11) B. cereus (2 μg/mL), B. subtilis (1 μg/mL), M. luteus (2 μg/mL)
Garcinia smeathmannii (stem barks) 1,3,5,8-Tetrahydroxy-2- (3-methy but-2-enyl)-4- (3,7-dimethylocta-2,6- dienyl)xanthone (12) E. faecalis (8 μg/mL) [33]
Cheffouxanthone (13) E. faecalis (8 μg/mL)
Ananixanthone (14) E. faecalis (2 μg/mL)
Clusiaceae Allanblackia gabonensis (fruits) Morelloflavone (15) ATCC8739 (8 μg/mL) [34]
Myristicaceae Pycnanthus angolensis (roots) Pycnanthulignene A (16) MRSA (9.8 μg/mL) [35]
3,4-Dimethoxy-3′,4′- methylenedioxy-7,7′- epoxylignan (17) M. smegmatis (9.8 μg/mL)
4,5-Dimethoxy-3′,4′- methylenedioxy-2,7′- cycloligna-7,7′-diene (18) M. tuberculosis (9.8 μg/mL)
Dioscoreaceae Dioscorea bulbifera (Bulbil) Bafoudiosbulbins C (19) M. smegmatis ATCC700084, M. tuberculosis ATCC27294 and M. tuberculosis MTCS2 (8 μg/mL) [36]
Clusiaceae Garcinia nobilis (stem bark) 4-Prenyl-2-(3,7-dimethyl- 2,6-octadienyl)- 1,3,5,8tetrahydroxyxantho ne (20) M. tuberculosis ATCC27294 and M. tuberculosis MTCS2 (8 μg/mL) [37]
Moraceae Dorstenia manii (roots) Dorsmanin C (21) P. aeruginosa PA 01 and E. coli ATCC 10536 (4 μg/mL) [38]
Dorsmanin F (22) P. aeruginosa PA 01 and E. coli ATCC 10536 (4 μg/mL), K. pneumonia PA01 (8 μg/mL)
Dorsmanin E (23) Candida albicans TCC9002 (8 μg/mL)
Ficus exasperata (stem bark) (S)-(−) Oxypeucedanin hydrate (24) B. cereus (9.76 μg/mL) [39]
(R)-(+) Oxypeucedanin hydrate (25)
Trilepisium madagascariense (stem bark) Dihydrokaempferol (26) E. coli ATCC8739 (8 μg/mL) [40]
Rutaceae Fagara tessmannii (roots) Bergenin (27) E.AG102 and K. pneumoniae ATCC 11296 (4 μg/mL)
E. coli ATCC 8739, K. pneumoniae ATCC 11296, K. pneumoniae KP 55, P. stuartii PS 299645 and P. aeruginosa PA01 (8 μg/mL)
[41]
Hypericaceae Harungana madagascariensis (bark) Ferruginin (28) E. coli ATCC 10536, K. pneumoniae K2 and E. cloacae BM 67 (4 μg/mL)
E. aerogenes ATCC 13048, E. aerogenes EA 294, P. aeruginosa PA01 and K. pneumoniae KP 55 (8 μg/mL)
K. pneumonia ATCC 11296, E. cloacae BM 47 and E. coli ATCC 8739, E. aerogenes ATCC 13048, K. pneumoniae KP 55, P. stuartii (8 μg/mL)
[42]
Fabaceae Erythrina sigmoidea (leaves) Neobavaisoflavone (29) E. coli ATCC 8739, E. cloacae ECC 169, K. pneumoniae KP 55, P. stuartii NAE16 and P. aeruginosa PA01 (8 μg/mL)
P. stuartii ATCC 29916, E. cloacae BM 47 (4 μg/mL)
[43]
Moraceae Milicia excels (roots and leaves) 2-(3,5-Dihydroxyphenyl) benzofuran-5,6-diol (30) E. coli ATCC 8739, K. pneumonia ATCC 11296, E. cloacae BM 47 (4 μg/mL)
E. coli AG 102 and K. pneumoniae KP 55 (8 μg/mL)
[44, 45, 46]
Candidone (31)
Myristicaceae Myristica fragrans (seeds) 3′,4′,7-Trihydroxyflavone (32) E. coli ATCC 8739 (8 μg/mL)
P. stuartii ATCC (199645) (4 μg/mL)
[47, 48]
Hypericaceae Hypericum roeperianum (stem Bark) 1,4,6,7-Tetrahydroxyxanthone (33) P. aeruginosa PA01 (2 μg/mL) [49]
Fabaceae Entada abyssinica (leaves) Entadanin (34) S. typhimurium (1.56 μg/mL) [50]
Quercitrin (35) S. typhimurium (3.12 μg/mL)
Meliaceae Pseudocedrela kotschyi (stem bark) 3,4-Secotirucalla- 4(28),7,24-trien-3,21- dioic acid (36) S. aureus (4 μg/mL) [51]
3,4-Secotirucalla- 4(28),7,24-trien-3,21- dioic acid (36) and 3- methyl ester 3,4- secotirucalla-4(28),7,24- trien-3,21-dioic (37) (1:1) S. aureus (8 μg/mL)
Rubiaceae Crossopteryx febrifuga (stem bark) l8-epi-3β-D- Glucopyranosylurs- l2,20(30)diene-27,28- dioic acid (38) K. pneumoniae ATCC11296 (8 μg/mL) [52]
Lamiaceae Leucosceptrum canum (aerial part) 4-En-3-keto-stigmasterol (39) M. luteus (9.5 μg/mL) [53]
Stigmast-5-en-3-acetate (40) M. luteus (4.2 μg/mL)
4′,5,7-Trihydroxy-6 methoxyflavone (41) M. luteus (6.5 μg/mL)
Leucoperoxyterpene (42) M. luteus (5.4 μg/mL) and S. minor (5.4 μg/mL)
Caryophyllaceae Silene rubella (aerial part) Oleanolic acid (43) VRE (6.36 μg/mL) [54]
Fabaceae Entada abyssinica (leaves) Ursolic acid (44) B. cereus (6.25 μg/mL) [55]

Table 1.

Examples of plant-based natural products with significant antiinfective properties.

Methicillin-resistant Staphylococcus aureus (MRSA), vancomycin resistant Enterococcus (VRE), methicillin-sensitive Staphylococcus aureus (MSSA), vancomycin-sensitive Enterococcus (VSE), Staphylococcus aureus (S. aureus), Bacillus subtilis (B. subtilis), Bacillus cereus (B. cereus), Micrococcus luteus (M. luteus), Mycobacteria smegmatis (M. smegmatis), Mycobacteria tuberculosis (M. tuberculosis), Pseudomonas aeruginosa (P. aeruginosa), Escherichia coli (E. coli), Klebsiella pneumonia (K. pneumonia), Candida albicans (C. albicans), Bacillus cereus (B. cereus), Escherichia coli (E. coli), Klebsiella pneumoniae (K. pneumoniae), Providencia stuartii (P. stuartii), Pseudomonas aeruginosa (P. aeruginosa), Enterobacter cloacae (E. cloacae), Enterobacter aerogenes (E. aerogenes), Salmonella typhimurium (S. typhimurium), Staphylococcus aureus (S. aureus), Micrococcus luteus (M. luteus), Streptococcus minor (S. minor), Vancomycin resistant Enterococcus (VRE).

Figure 1.

Bioactive compounds against infective bacteria and fungi.

Advertisement

3. Plant-based compounds with anticancer features

The chemistry and biology of plants to fight against malignant cells are wide and diverse. Various classes of metabolites have been reported to possess valuable anticancer properties. As a recall, taxol, one of the mostly used anticancer drug in chemotherapy, is a complex diterpene-based metabolite; vinblastine, used in the therapy of various cancer as well, is made up of terpenic indol-type alkaloids and artemisinin or parthenolide actually in active clinical trials for cancer drugs possess a sesquiterpenoid lactone backbone. Since 2010, more than 72 compounds have been reported with considerable antiproliferative activity against different cancer cell lines with IC50 ranging from 0.001–10 μM.

These compounds are distributed in homoisoflavonoids (4560), isoprenylflavonoids (6163, 6870), stilbenoids (6467), xanthones (7172), benzophenanthridines (7382), Amaryllidaceae-type alkaloids (8386), cardenolides (8790) and sesquiterpenoid lactones (91117). Their respective sensibility toward tumor cell lines are depicted in Table 2 and their respective structures in Figure 2. As expected, sesquiterpenoid lactones were the most exploited metabolites. They are reputed for their ability to induce apoptosis in cancer cell lines with good selectivity. Homoisoflavonoids were the second most important group of compounds found to exhibit high cytotoxicity herein. The interest in this class of metabolites for anticancer solution is most likely related to their potency as inhibitor of angiogenesis both in vitro and in vivo, without showing any toxicity [80]. On the other hand, benzophenanthridines are reputed for their bioavailability because they contained more often ionic bond besides their bioactivity. Their mode of action in cancer therapy includes either the inhibition of mitosis via a reaction of the imine bond with the sulfhydryl nucleophile in protein and enzyme or the enzymatic activities of DNA Topoisomerase I and Topoisomerase II by implantation into DNA molecules to retard the fast proliferation of tumor cells [81].

Family Species (part) Compound name Cancer cell lines (IC50) Refs.
Asparagaceae Bellevalia eigii (bulbs) 5,7,3′-triihydroxy-4′- methoxy
Homoisoflavanone (45)
MDA-MB-435 (1.0 μM) [57]
5,3′-dihydroxy-4′,7,8- trimethoxy
Homoisoflavanone (46)
MDA-MB-435 (1.1 μM)
7-O-methyl-3′- hydroxypunctatin (47) MDA-MB-435 (4–6 μM)
Bellevalia flexuosa (bulbs) 3′-hydroxy-3,9- dihydroeucomin (48) MDA-MB-435 (1–6 μM); MDA-MB- 231 (9–5 μM) [58]
Asparagaceae Urginea depressa (whole plant) Urgineanin A (49) H522-T1 (0–071 μM); A2780 (0.32 μM); A2058 (0.068 μM) [59]
Urgineanin B (50) H522-T1 (6.78 μM); A2780 (3.4 μM)
Urgineanin C (51) H522-T1 (0.74 μM); A2780 (1.35 μM), A2058 (0.69 μM)
Urgineanin D (52) H522-T1 (0.43 μM); A2780 (0.35 μM), A2058 (0.38 μM)
Urgineanin E (53) A2780 (1.44 μM)
Urgineanin F (54) A2780 (2.3 μM)
Convallariaceae Ophiopogon japonicus (tubers) Homoisopogon A (55) KB (0.51 μM); LU-1 (0.66 μM); SK- Mel-2 (0.66 μM) [60]
Ophiopogon japonicus (roots) 5,7,4′-Trihydroxy-3′-
methoxy-6,8-
dimethylhomoisoflavanone (56)
A549 (6.40 μM) [61]
Methylophiopogonanone B (57) A549 (0.84 μM)
Methylophiopogonanone A (58) A549 (1.66 μM)
Liliaceae Scilla persica (bulbs) Scillapersicene (59) AGS (8.4 μM) [62]
Amaryllidaceae Crinum zeylanicum (whole plant) Ungeremine (60) CCRF-CEM (4.89 μM); MDA-MB - 231-pcDNA (5.47 μM); MDA-MB- 231-BCRP (3.67 μM); HCT116 (p53 +/+) (6.45 μM)
HCT116 (p53 /) (7.06 μM); U87MG (5.38 μM)
[63]
Euphorbiaceae Macaranga balansae (fruits) 6,8-Diprenyl-4-methyl- naringenin (61) Pan C1 (7.89 μM) [64]
(2S)-6-Farnesylnaringenin (62) P388 (3.27 μg/mL)
6-Farnesyl-3′,4′,5,7-tetrahydroxy flavanone (63) P388 (2.61 μg/mL)
Macaranga triloba (inflorescences) HeLa (1.3 μg/mL), HL-60 (3.3 μg/mL) [65]
Euphorbiaceae Macaranga tanarius (fruits) Vedelianin (64) KB (0.050 μM), MCF-7 (0.050 μM) [66]
Schweinfurthin E (65) KB (0.050 μM), MCF-7 (0.030 μM)
Schweinfurthin F (66) KB (0.10 μM), MCF-7 (0.12 μM)
Schweinfurthin H (67) KB (0.26 μM)
Thelypteridaceae Cyclosorus parasiticus (leaves) Parasiticin C (68) SW1990 (2.33 μM), MDA-MB-231 (4.88 μM),
MCF-7 (4.16 μM), HepG2 (1.6 μM), A.549 (5.50 μM), ALLSIL (6.06 μM)
[67]
2′,4′-Dihydroxy-6′-methoxy-3′,5′-Dimethylchalcone (69) SW1990 (6.64 μM),
MDA-MB-231 (9.67 μM),
MCF-7 (8.49 μM), HepG2 (2.82 μM), A549 (7.89 μM), ALLSIL (9.50 μM)
Moraceae Artocarpus obtusus (stem bark) Pyranocycloartobiloxanthone A (70) HL60 (0.5 μg/mL), K562 (2.0 μg/mL) [68]
Guttiferae Calophyllum soulattri (stem bark) Soulattrin (71) Raji (1.01 μM/mL), LS174T (1.25 μM/mL), IMR-32 (0.27 μg/mL), SK-MEL-28 (0.57 μM/mL). [69]
Guttiferae Garcinia xanthochymus (stem bark) 1,3,5,6-T etrahydroxy-4,7,8-tri(3-methylbut-2-enyl)xanthone (72) PC-3 (6.8 μM) [70]
Papaveraceae Macleaya microcarpa (roots) Maclekarpine A (73) BGC-823 (0.7 μM) [71]
Maclekarpine C (74) HCT-8 (1.9 μM), Bel-7402 (2.1 μM), A2780 (1.6 μM), A549 (3.4 μM)
Maclekarpine D (75) HCT-8 (1.9 μM), BGC-823 (0.2 μM), A2780 (2.0 μM)
Maclekarpine E (76) BGC-823 (0.1 μM)
6-Methoxydihydrochelerythrine
(77)
HCT-8 (1.1 μM), Bel-7402 (0.9 μM) BGC-823 (0.8 μM), A2780 (1.8 μM)
Dihydrosanguinarine (78) HCT-8 (1.3 μM), Bel-7402 (2.3 μM) BGC-823 (0.1 μM), A2780 (2.1 μM)
Dihydrochelerythrine (79) HCT-8 (1.4 μM), BGC-823 (0.4 μM), A2780 (3.5 μM)
6-Butoxydihydrochelerythrine (80) HCT-8 (1.7 μM), Bel-7402 (1.3 μM) BGC-823 (0.7 μM) A2780 (1.8 μM),
Papaveraceae Bis[6-(5,6-dihydrochelerythrinyl)] ether (81) HCT-8 (1.6 μM), Bel-7402 (2.1 μM) BGC-823 (0.1 μM), A2780 (1.6 μM),
6-Methoxydihydrosanguinarine (82) HCT-8 (0.5 μM), Bel-7402 (0.5 μM) BGC-823 (0.6 μM), A2780 (0.5 μM), A549 (0.6 μM)
Amaryllidaceae Zephyranthes candida (whole plant) N-methylhemeanthidine Chloride (83) HL-60 (0.91 μM), K562 (1.0 μM), A549 (1.1 μM), HepG2 (1.5 μM), HT-29 (1.2 μM) [72]
Hemeanthamin (84) HL-60 (1.4 μM), K562 (2.5 μM), A549 (2.5 μM), HepG2 (4.8 μM), HT-29 (2.1 μM)
Lycorine (85) HL-60 (1.6 μM), K562 (2.3 μM), A549 (1.9 μM), HepG2 (3.7 μM), HT-29 (3.2 μM)
N-phenethylcrinasiadine (86) HL-60 (1.6 μM), K562 (2.3 μM), A549 (1.9 μM), HepG2 (3.7 μM), HT-29 (3.2 μM)
Asparagaceae Bellevalia flexuosa (bulbs) Urginin B (87) A2780 (0.011 μM), A2058 (0.060 μM), H522-T1 (0.044 μM) [59]
Urginin C (88) A2780 (0.041 μM), A2058 (0.076 μM), H522-T1 (0.051 μM)
14β-Bydroxy-19β-oxobufa-4,20, 22-trienolide-3β-O-β-D-gluco pyranoside (89) A2780 (0.024 μM), A2058 (0.048 μM), H522-T1 (0.034 μM)
14β-Hydroxybufa-4,20,22-trien olide-3β-O-{α-L- rhamnopyranosyl-[(1 → 4)-β- D-glucopyranosyl]-(1 → 3)-α-L-rhamnopyranoside} (90) A2780 (0.111 μM), A2058 (0.18 μM), H522-T1 (0.11 μM)
Asteraceae Leptocarpha rivularis Leptocarpin (91) DU-145 (2.0 μM), PC-3 (4.5 μM), HT-29 (3.8 μM), MCF7 (3.1 μM), MDA-MB-231 (6.4 μM), CCD 841 CoN (5.2 μM) [73]
Smallanthus sonchifolius (leaves) Enhydrin (92) CCRF-CEM (3.6 μM) [74]
Uvedalin (93) CCRF-CEM (9.2 μM)
Polymatin B (94) CCRF-CEM (0.8 μM), CEM-ADR5000 (1.3 μM), MIA-PaCa-2 (3.7 μM)
Sonchifolin (95) CCRF-CEM (3.1 μM), CEM-ADR5000 (3.1 μM), MIA-PaCa-2 (7.4 μM)
8β-Angeloyloxy-9α-hydroxy-14-oxo-acanthospermolide (96) CCRF-CEM (2.2 μM), CEM-ADR5000 (6.7 μM), MIA-PaCa-2 (8.9 μM)
Fluctuanin (97) CCRF-CEM (0.6 μM), CEM-ADR5000 (1.4 μM), MIA PaCa-2 (4.4 μM)
Ambrosia cumanensis (aerial parts) 2,3-Dehydropsilostachyn C (98) Jurkat (6.0 μM), U937 (8.0 μM) [75]
Sonchus palustris (roots) 15-p-Hydroxyphenylacetyllactucin (99) CEM (5.1 μM), BJ (9.8 μM) [76]
15-p-Methoxyphenylacetyllactucin (100) CEM (3.9 μM), BJ (8.4 μM)
Compositae Carpesium abrotanoides (whole plant) Caroguaianolide A (101) MDA-MB-231 (7.96 μM) [77]
Caroguaianolide B (102) MDA-MB-231 (4.25 μM), HGC-2 (6.47 μM)
Carpesium abrotanoides (whole plant) Caroguaianolide C (103) MDA-MB-231 (2.67 μM), HGC-2 (4.83 μM)
Akihalin (104) MDA-MB-231 (4.83 μM), HGC-2 (7.35 μM)
4β-Hydroxy,l0β-hydroperoxyl,5αh,7αh,8βh-guaia-1,11(13)- dien-8α,12- olide (105) MDA-MB-231 (5.79 μM)
4α-Hydroxy-1βh-guaia-9,11(13)-dien-12,8α-olide (106) MDA-MB-231 (4.07 μM), HGC-2 (8.95 μM)
(3ar,4as,5S,7as,8S,9ar)-5-Hydroxy-4a,8-dimethyl-3-methylen-decahy droazuleno[6,5-b]furan- 2(3H)-on (107) MDA-MB-231 (5.32 μM)
Carpesium faberi (whole plant) Guaianodilactones A (108) CCRF-CEM (9.13 μM) [78]
Guaianodilactones C (109) CCRF-CEM (4.74 μM)
Guaianodilactones B (110) CCRF-CEM (2.03 μM)
Asteraceae Inula japonica (aerial part) Neojaponicone B (111) Jurkat (5.9 μM), 6 T-CEM (4.4 μM) [79]
Inulanolide E (112) Jurkat (5.5 μM), 6 T-CEM (4.6 μM)
Inulanolide A (113) Jurkat (5.8 μM), 6 T-CEM (4.3 μM)
Japonicone Q (114) Jurkat (3.3 μM), 6 T-CEM (2.7 μM)
Japonicone N (115) Jurkat (2.5 μM), 6 T-CEM (2.4 μM)
Japonicone S (116) Jurkat (4.5 μM), 6 T-CEM (3.3 μM)
Japonicone A (117) Jurkat (3.1 μM) 6 T-CEM (2.2 μM)

Table 2.

Examples of significant secondary metabolites with antiproliferative properties.

Melanoma (MDA-MB-435, A2058 and SK-Mel-2); human non-small-cell lung (H522-T1), ovarian cancer (A2780), human epidermoid carcinoma (KB), human lung adenocarcinoma (LU-1), breast (MDA-MB-231), gastric (AGS), human myeloid leukemia (K562), human gastric (SGC-7901), human-lung-tumor (A549). Murine leukemia (P-388), human pancreatic (Pan C1 and SW1990), human cervical carcinoma (Hela), mouse leukemia (P388), human leukemia (HL-60 and ALL-SIL), mouth epidermal carcinoma cells (KB), breast cancer (MCF-7 and MDA-MB-231) lung cancer (A549), hepatocellular carcinoma (HepG2), human promyelocytic leukemia (HL60), human chronic myeloid leukemia (K562), B-lymphocyte (Raji), colon carcinoma (LS174T), human neuroblastoma (IMR-32), skin carcinoma (SK-MEL-28), colon (HCT-8), liver (Bel-7402), stomach (BGC-823), ovarian (A2780), lung (A549). Myeloid leukemia (HL-60 and K562), lung (A549), hepatocellular carcinoma (HepG2), colon (HT-29), ovarien (A2780), melanoma (A2058), non-small-cell lung cancer (H522-T1), lymphoblastic leukemia cell line (CCRF-CEM), resistant T-cell leukemia cell line (CEM-ADR5000), the pancreatic, carcinoma cell line (MIA PaCa-2)–, and on peripheral blood mononuclear cells (PBMC) from healthy human subjects, HeLa (cervical carcinoma), Jurkat (T-cell leukemia), and U937 (monocytic leukemia) cell lines, human acute lymphoblastic leukemia (CEM), and normal human skin fibroblasts (BJ), human breast (MDA-MB-231), human gastric (HGC-27), human leukemia (CCRF-CEM).

Figure 2.

Bioactive compounds against cancer cell lines.

Advertisement

4. Natural products in active development for drug discovery

Knowing the categories of compounds which has been screened and choose for clinical trials is quite important. It can help redefine our objectives and outlines in research. However, such information is not accessible easily. Almost all pharmaceutical makers keep this information for private uses. Nevertheless, available reports before 2010 on valuable compounds in development for cancer therapy for instance can continue to be used and analyze. There are privilege structures with unique structurally subunits which confer to drugs distinctive therapeutic affinities to a biological system. These core molecules include β-lactam unit like in penicillin; cyclopentanoperhydrophenanthrene fragment like in testosterone; pyrone, coumarins, isoflavone, or chalcone moieties and alkaloids like quinoline, isoquinoline or indole units.

As an example, since the large-scale screening for anticancer agents launched in the USA in 1960, more than 3000 sesquiterpene lactones have been reported. Most of them are with cytotoxic properties. Sesquiterpene lactones are well-known for their ability to bind sulfhydryl-containing peptides, mainly in proteins, presented as important route in well- programed death of a cell [82, 83, 84, 85]. This property and other have raised up interests in this class of compounds. Many members of this class are currently in clinical trials for drug development including parthenolide, artemisinin or thapsigargin among others.

Another most important class of phytocompounds in cancer therapy is phenolic compounds. Members of these classes of metabolites are reputed in caspase activation causing apoptosis in tumor cell lines. Research found that furanocoumarins for instance in grapefruit showed significant effects towards breast cancer, the second World leading cause of cancer-related death among Women [86]. In the same line, coumarine-type of compounds known as calanolides, isolated from Calophyllum species have been found to be active against lymphoblastic cells infected with HIV-1 [87]. They are currently in clinical trials Phase II to drug development. Likewise, all other phenolic compounds listed above can also undergo similar interactions with cancer cells. Anthraquinones, and quinones, in general form the basic core of many anticancer drugs known as anthracyclines. Resveratrol, a stilbene-like metabolite, is being continuously checked to explain issues encountered during laboratory trials against cancer in animal model. However, association of resveratrol with established anticancer drugs like clofarabine has been proved against mesothelioma cell lines [88].

Advertisement

5. Conclusion

The World is facing an unprecedented drastic climate change that impacts negatively not only on human beings but also plants. New metabolism routes have surely emerged leading to compounds with unprecedented structures for some and with relevant bioactivities for others. However, nothing is being done to take advantages of this wealth for our health care always relying on “modern drugs.” We should start exploring ways to use natural products with anticancer effects along with standard chemotherapy treatments to increase potency while reducing side effects of actual drugs. This strategy is currently being used in the USA. We highlighted relevant bio-sensibility of some compounds and they should now be investigated as main constituents to a standardization process of their respective plant extracts. The present survey can also help researchers in developing countries working on plants, to re-focus their research works.

Advertisement

Acknowledgments

The authors would like to acknowledge the Yaounde-Bielefeld Graduate School of Natural Products with Antiparasite and Antibacterial Activities (YaBiNaPA, www.yabinapa.de) for the research stay granted at the University of Bielefeld in Germany under the project No. 57316173.

Advertisement

Conflict of interest

The authors declare no conflict.

References

  1. 1. Bitchagno GTM, Schüffler A, Simo IK, Krumb M, Tane P, Opatz T. Neo-clerodane diterpenoids from Conyza pyrrhopappa Sch.Bip. ex A. Natural Product Research. 2019. DOI: 10.1080/14786419.2019.1690490
  2. 2. Bitchagno MGT, Tankeo BS, Tsopmo A, Mpetga SDJ, Tchinda TA, Fobofou TSA, et al. Lemairones A and B: Two new antibacterial tetraflavonoids from the leaves of Zanthoxylum lemairei (Rutaceae). Phytochemistry Letters. 2015;14:1-7. DOI: 10.1016/j.phytol.2015.08.012
  3. 3. Nganou BK, Mbaveng AT, Fobofou SAT, Fankam AG, Bitchagno GTM, Mpetga JDS, et al. Furoquinolines and dihydrooxazole alkaloids with cytotoxic activity from the stem bark of Araliopsis soyauxii. Fitoterapia. 2019;133:193-199. DOI: 10.1016/j.fitote.2019.01.003
  4. 4. Mbaveng AT, Bitchagno GTM, Kuete V, Tane P, Efferth T. Cytotoxicity of ungeremine towards multi-factorial drug resistant cancer cells and induction of apoptosis, ferroptosis, necroptosis and autophagy. Phytomedecine. 2019;60:152832. DOI: 10.1016/j.phymed
  5. 5. Sonfack G, Tchinda CF, Simo IK, Bitchagno GTM, Nganou BK, Çelik I, et al. Saponin with antibacterial activity from the roots of Albizia adianthifolia. Natural Product Research. 2019. DOI: 10.1080/14786419.2019.1672689
  6. 6. Barreiro EJ. Privileged scaffolds in medicinal chemistry: An introduction. In: Bräse S, editor. Privileged Scaffolds in Medicinal Chemistry: Design, Synthesis, Evaluation. RSC Drug Discovery Series No. 50: Cambridge; 2015. p. 476. DOI: 10.1039/9781782622246-00001
  7. 7. Butler MS, Robertson AAB, Cooper MA. Natural product and natural product derived drugs in clinical trials. Natural Product Reports. 2014;31:1612-1661. DOI: 10.1039/C4NP00064A
  8. 8. Butler MS. Natural products to drugs: Natural product derived compounds in clinical trials. Natural Product Reports. 2005;22:162-195. DOI: 10.1039/B514294F
  9. 9. Dias DA, Urban S, Roessner U. A historical overview of natural products in drug discovery. Metabolites. 2012;2:303-336. DOI: 10.3390/metabo2020303
  10. 10. Der Marderosian A, Beutler JA, editors. The Review of Natural Products. 2nd ed. Seattle, WA, USA: Facts and Comparisons; 2002. p. 794
  11. 11. Newman DJ, Cragg GM. Natural products as sources of new drugs over the last 25 years. Journal of Natural Products. 2007;70:461-477. DOI: 10.1021/np068054v
  12. 12. Kedei N, Lundberg DJ, Toth A, Welburn P, Garfield SH, Blumberg PM. Characterization of the interaction of ingenol 3-angelate with protein kinase C. Cancer Research. 2004;64:3243-3255. DOI: 10.1158/0008-5472.CAN-03-3403
  13. 13. Ogbourne SM, Suhrbier A, Jones B. Antitumour activity of ingenol 3-angelate: Plasma membrane and mitochondrial disruption and necrotic cell death. Cancer Research. 2004;64:2833-2839. DOI: 10.1158/0008-5472.CAN-03-2837
  14. 14. Aniszewski T. Alkaloids—Secrets of life. In: Aniszewski T, editor. Alkaloid Chemistry, Biological Significance, Applications and Ecological Role. Amsterdam: The Netherlands: Elsevier Science; 2007. p. 334
  15. 15. Hartman H, Matsuno K. The Origin and Evolution of the Cell. Singapore: World Scientific Publishing Co Pte Ltd; 1993. p. 446. DOI: 10.1142/9789814536219
  16. 16. American Academy of Microbiology FAQ: Human Microbiome. Archived 31 December 2016 at the Wayback Machine
  17. 17. Rosner JL. Ten times more microbial cells than body cells in humans? Microbe. 2014;9:47. DOI: 10.1128/microbe.9.47.2
  18. 18. Abbott A. Scientists bust myth that our bodies have more bacteria than human cells. Nature. 2016. DOI: 10.1038/nature.2016.19136
  19. 19. Abreu AC, McBain AJ, Simões M. Plants as sources of new antimicrobials and resistance-modifying agents. Natural Product Reports. 2012;29:1007-1021. DOI: 10.1039/c2np20035
  20. 20. Kuete V. Potential of Cameroonian plants and derived products against microbial infections: A review. Planta Medica. 2010;76:1479-1491. DOI: 10.1055/s-0030-1250027
  21. 21. Kuete V, Efferth T. African flora has the potential to fight multidrug resistance of cancer. BioMed Research International. 2015;2015:914813. DOI: 10.1155/2015/914813
  22. 22. Cowan MM. Plant products as antimicrobial agents. Clinical Microbiology Reviews. 1999;12:564-582. DOI: 10.1128/CMR.12.4.564
  23. 23. Watnick P, Kolter R. Biofilm, city of microbes. Journal of Bacteriology. 2000;182:2675-2679. DOI: 10.1128/jb.182.10.2675-2679.2000
  24. 24. O’Toole GA, Kolter R. Initiation of biofilm formation in Pseudomonas fluorescens WCS365 proceeds via multiple, convergent signalling pathways: A genetic analysis. Molecular Microbiology. 1998;28:449-461. DOI: 10.1046/j.1365-2958.1998.00797.x
  25. 25. Briandet R, Herry J, Bellon-Fontaine M. Determination of the van der Waals, electron donor and electron acceptor surface tension components of static Gram-positive microbial biofilms. Colloids and Surfaces. B, Biointerfaces. 2001;21:299-310. DOI: 10.1016/S0927-7765(00)00213-7
  26. 26. Takahashi H, Suda T, Tanaka Y, Kimura B. Cellular hydrophobicity of listeria monocytogenes involves initial attachment and biofilm formation on the surface of polyvinyl chloride. Letters in Applied Microbiology. 2010;50:618-625. DOI: 10.1111/j.1472-765X.2010.02842.x
  27. 27. Man A, Santacroce L, Jacob R, Mare A, Man L. Antimicrobial activity of six essential oils against a group of human pathogens: A comparative study. Pathogen. 2019;8:15. DOI: 10.3390/pathogens8010015
  28. 28. Lambert RJW, Skandamis PN, Coote PJ, Nychas G-JE. A study of the minimum inhibitory concentration and mode of action of oregano essential oil, thymol and carvacrol. Journal of Applied Microbiology. 2001;91:453-462. DOI: 10.1046/j.1365-2672.2001.01428.x
  29. 29. Saad IK, Hassan L, Ghizlane Z, Hind M, Adnane R. Carvacrol and thymol components inhibiting Pseudomonas aeruginosa adherence and biofilm formation. African Journal of Microbiology Research. 2011;5:3229-3232. DOI: 10.5897/AJMR11.275
  30. 30. Sakagami Y, Thevanesam V, Programme NP, Lanka S, Lanka S, Sakagami Y. Antibacterial activity of xanthones from Garcinia mangostana (L.) and their structure – Activity relationship studies. Natural Product Research. 2013;27:938-941. DOI: 10.1080/14786419.2012.678348
  31. 31. Auranwiwat C, Trisuwan K, Saiai A, Pyne SG, Ritthiwigrom T. Antibacterial tetraoxygenated xanthones from the immature fruits of Garcinia cowa. Fitoterapia. 2014;98:179-183. DOI: 10.1016/j.fitote.2014.08.003
  32. 32. Siridechakorn I, Phakhodee W, Ritthiwigrom T, Promgool T. Antibacterial dihydrobenzopyran and xanthone derivatives from Garcinia cowa stem barks. Fitoterapia. 2012;83:1430-1434. DOI: 10.1016/j.fitote.2012.08.006
  33. 33. Fouotsa H, Lannang AM, Dzoyem JP, Tatsimo SJN, Neumann B, Eloff JN, et al. Antibacterial and antioxidant Xanthones and Benzophenone from Garcinia smeathmannii. Planta Medica. 2015;81:594-599. DOI: 10.1055/s-0035-1545841
  34. 34. Nganou BK, Konga IS, Fankam AG, Bitchagno GTM, Sonfack G, Nayim P, et al. Guttiferone BL with antibacterial activity from the fruits of Allanblackia gabonensis. Natural Product Research. 2018;6419:1-9. DOI: 10.1080/14786419.2018.1465424
  35. 35. Nono ECN, Mkounga P, Kuete V, Marat K, Hultin PG, Nkengfack AE. Pycnanthulignenes A-D, antimicrobial cyclolignene derivatives from the roots of Pycnanthus angolensis. Journal of Natural Products. 2010;73:213-216. DOI: 10.1021/np9007393
  36. 36. Kuete V, Nono ECN, Mkounga P, Marat K, Hultin PG, Nkengfack AE. Antimicrobial activities of the CH2Cl2–CH3OH (1:1) extracts and compounds from the roots and fruits of Pycnanthus angolensis (Myristicaceae). Natural Product Research. 2010;25:432-443. DOI: 10.1080/14786419.2010.522577
  37. 37. Kuete V, Teponno RB, Mbaveng AT, Tapondjou AL, Meyer JJM, Barboni L, et al. Antibacterial activities of the extracts, fractions and compounds from Dioscorea bulbifera. BMC Complementary and Alternative Medicine. 2012;12:228. DOI: 10.1186/1472-6882-12-228
  38. 38. Fouotsa H, Mbaveng AT, Mbazoa CD, Nkengfack AE, Farzana S, Iqbal C, et al. Antibacterial constituents of three Cameroonian medicinal plants: Garcinia nobilis, Oricia suaveolens and Balsamocitrus camerunensis. BMC Complementary and Alternative Medicine. 2013;13:81. DOI: 10.1186/1472-6882-13-81
  39. 39. Mbaveng AT, Kuete V, Ngameni B, Beng VP, Ngadjui BT, Meyer JJM, et al. Antimicrobial activities of the methanol extract and compounds from the twigs of Dorstenia mannii (Moraceae). BMC Complementary and Alternative Medicine. 2012;12:83. DOI: 10.1186/1472-6882-12-83
  40. 40. Dongfack MDJ, Lallemand M-C, Kuete V, Mbazoa CD, Wansi JD, Dufat H, et al. A new Sphingolipid and Furocoumarins with antimicrobial activity from Ficus exasperate. Chemical & Pharmaceutical Bulletin. 2012;60:1072-1075. DOI: 10.1248/cpb.c12-00279
  41. 41. Ango YP, Kapche DWFG, Kuete V, Ngadjui BT, Bezabih M, Abegaz BM. Chemical constituents of Trilepisium madagascariense (Moraceae) and their antimicrobial activity. Phytochemistry Letters. 2012;5:524-528. DOI: 10.1016/j.phytol.2012.05.006
  42. 42. Tankeo SB, Damen F, Awoufack MD, Mpetga J, Tane P, Eloff JN, et al. Antibacterial activities of the methanol extracts, fractions and compounds from Fagara tessmanii. Journal of Ethnopharmacology. 2015;169:275-279. DOI: 10.1016/j.jep.2015.04.041
  43. 43. Tankeo SB, Damen F, Sandjo LP, Tane P, Kuete V. Antibacterial activities of the methanol extracts, fractions and compounds from Harungana madagascariensis. Journal of Ethnopharmacology. 2016;190:100-105. DOI: 10.1016/j.jep.2016.06.005
  44. 44. Djeussi DE, Sandjo LP, Noumedem JAK, Omosa LK, Ngadjui BT, Kuete V. Antibacterial activities of the methanol extracts and compounds from Erythrina sigmoidea against Gram-negative multi-drug resistant phenotypes. BMC Complementary and Alternative Medicine. 2015;15:453. DOI: 10.1186/s12906-015-0978-8
  45. 45. Mbaveng AT, Sandjo LP, Tankeo SB, Ndifor AR, Pantaleon AA, Nagdjui BT, et al. Antibacterial activity of nineteen selected natural products against multi-drug resistant Gram-negative phenotypes. Springerplus. 2015;4:823. DOI: 10.1186/s40064-015-1645-8
  46. 46. Ouete JLN, Sandjo LP, Kapche DDW, Yeboah SO, Mapitse P, Abegaz BM, et al. Excelsoside: A new benzylic diglycoside from the leaves of Milicia excelsa. Zeitschrift für Naturforschung. 2014;69:271-275. DOI: 10.5560/ZNC.2014-0087
  47. 47. Ouete JLN, Sandjo LP, Kapche DDW, Liermann JC, Opatz T, Simo IK, et al. A new flavone from the roots of Milicia excelsa (Moraceae). Zeitschrift für Naturforschung. Section C. 2013;68:259-263. DOI: 10.5560/ZNC.2013.68c0259
  48. 48. Nganou BK, Konga IS, Fankam AG, Bitchagno GTM, Sonfack G, Nayim P, et al. Guttiferone BL with antibacterial activity from the fruits of Allanblackia gabonensis. Natural Product Research. 2018;33:2638-2646. DOI: 10.1080/14786419.2018.1465424
  49. 49. Dzotam JK, Simo IS, Bitchagno GTM, Celik I, Sandjo LP, Tane P, et al. In vitro antibacterial and antibiotic modifying activity of crude extract, fractions and 3′,4′,7-trihydroxyflavone from Myristica fragrans Houtt against MDR Gram-negative enteric bacteria. BMC Complementary and Alternative Medicine. 2018;18:15. DOI: 10.1186/s12906-018-2084-1
  50. 50. Damen F, Demgne OMF, Bitchagno GTM, Celik I, Mpetga JDS, Tankeo SB, et al. A new polyketide from the bark of Hypericum roeperianum Schimp. (Hypericaceae). Natural Product Research. 2019. DOI: 10.1080/14786419.2019.1677655
  51. 51. Mambou CS, Nono RN, Chouna JR, Tamokou JDD, Nkeng-Efouet-Alango P, Sewald N. Antibacterial secotirucallane triterpenes from the stem bark of Pseudocedrela kotschyi. Zeitschrift für Naturforschung. 2018;73:241-246. DOI: 10.1515/znc-2017-0207
  52. 52. Chouna JR, Tamokou JDD, Nkeng-Efouet-Alango P, Lenta BN, Sewald N. Antimicrobial triterpenes from the stem bark of Crossopteryx febrifuga. Zeitschrift für Naturforschung. 2015;70:169-173. DOI: 10.1515/znc-2014-4168
  53. 53. Devkota KP, Lenta BN, Wansi JD, Sewald N. Antibacterial constituents from Leucosceptrum canum. Phytochemistry Letters. 2009;3:24-28. DOI: 10.1016/j.phytol.2009.10.007
  54. 54. Hussein IA, Srivedavyasasri R, El-Hela AA, Mohammad AI, Ross SA. Antimicrobial secondary metabolites from Silene rubella growing in Egypt. Journal of Biomedical and Pharmaceutical Research. 2019;8. DOI: 10.32553/jbpr.v8i6.694
  55. 55. Dzoyem JP, Melong R, Tsamo AT, Tchinda AT, Kapche DGWF, Ngadjui BT, et al. Cytotoxicity, antimicrobial and antioxidant activity of eight compounds isolated from Entada abyssinica (Fabaceae). BMC Research Notes. 2017;10:118. DOI: 10.1186/s13104-017-2441-z
  56. 56. Ali B, Al-Wabel NA, Shams S, Ahamad A, Khan SA, Anwar F. Essential oils used in aromatherapy: A systemic review. Asian Pacific Journal of Tropical Biomedicine. 2015;5:601-611. DOI: 10.1016/j.apjtb.2015.05.007
  57. 57. Alali F, El-elimat T, Albataineh H, Al-balas Q, Al-gharaibeh M, Falkinham JO, et al. Cytotoxic Homoisoflavones from the bulbs of Bellevalia eigii. Journal of Natural Products. 2015;78:1708-1715. DOI: 10.1021/acs.jnatprod.5b00357
  58. 58. El-elimat T, Rivera-chávez J, Burdette JE, Czarnecki A, Alhawarri MB, Al-gharaibeh M, et al. Cytotoxic homoisoflavonoids from the bulbs of Bellevalia flexuosa. Fitoterapia. 2018;127:201-206. DOI: 10.1016/j.fitote.2018.02.022
  59. 59. Dai Y, Harinantenaina L, Brodie PJ, Goetz M, Shen Y, Tendyke K, et al. Antiproliferative Homoisoflavonoids and Bufatrienolides from Urginea depressa. Journal of Natural Products. 2013;76:865-872. DOI: 10.1021/np300900a
  60. 60. Dang NH, Chung ND, Tuan HM, Hiep NT, Dat NT. Cytotoxic Homoisoflavonoids from Ophiopogon japonicus tubers. Biological & Pharmaceutical Bulletin. 2016;65:204-207. DOI: 10.1248/cpb.c16-00743
  61. 61. Zhou C, Zou L, Mo J, Wang X, Yang B, He Q. Homoisoflavonoids from Ophiopogon japonicus. Helvetica Chimica Acta. 2013;96:1397-1405. DOI: 10.1002/hlca.201200493
  62. 62. Ghoran SH, Saeidnia S, Babaei E, Kiuchi F, Hussain H, Plants M, et al. Scillapersicene: A new homoisoflavonoid with cytotoxic activity from the bulbs of Scilla persica. Natural Product Research. 2015;30:1309-1314. DOI: 10.1080/14786419.2015.1054286
  63. 63. Mbaveng AT, Bitchagno GTM, Kuete V, Tane P, Thomas E. Phytomedicine cytotoxicity of ungeremine towards multi-factorial drug resistant cancer cells and induction of apoptosis , ferroptosis, necroptosis and autophagy. Phytomedicine. 2019;60:152832. DOI: 10.1016/j.phymed.2019.152832
  64. 64. Doan H, Mai T, Toan TP, Huu GT, Le TN, Thi V, et al. New flavonoid and stilbene derivatives from the fruits of Macaranga balansae. Natural Product Research. 2019. DOI: 10.1080/14786419.2019.1587425
  65. 65. Zakaria I, Ahmat N, Jaafar FM, Widyawaruyanti A. Flavonoids with antiplasmodial and cytotoxic activities of Macaranga triloba. Fitoterapia. 2012;83:968-972. DOI: 10.1016/j.fitote.2012.04.020
  66. 66. Mai T, Doan H, Nguyen TL, Van TT, Vu VN, Phi TD, et al. Cytotoxic phenolic compounds from fruit glandular trichomes of Macaranga tanarius. Journal of Analytical Methods in Chemistry. 2019;2019:2917032. DOI: 10.1155/2019/2917032
  67. 67. Wei H, Zhang X, Wu G, Yang X, Pan S, Wang Y, et al. Chalcone derivatives from the fern Cyclosorus parasiticus and their anti-proliferative activity. Food and Chemical Toxicology. 2013;60:147-152. DOI: 10.1016/j.fct.2013.07.045
  68. 68. Hashim NM, Rahmani M, Cheng G, Ee L, Sukari MA, Yahayu M, et al. Antiproliferative activity of xanthones isolated from Artocarpus obtusus. BioMed Research International. 2012;2012:130627. DOI: 10.1155/2012/130627
  69. 69. Mah SH, Cheng G, Ee L, Teh SS, Sukari MA. Antiproliferative xanthone derivatives from Calophyllum inophyllum and Calophyllum soulattri. Pakistan Journal of Pharmaceutical Sciences. 2012;28:425-429
  70. 70. Ji F, Li Z, Liu G, Niu S, Zhao N, Liu X, et al. Xanthones with Antiproliferative effects on prostate cancer cells from the stem bark of Garcinia xanthochymus. Natural Product Communications. 2012;7:53-56. DOI: 10.1177/1934578X1200700119
  71. 71. Deng A-J, Qin H-L. Cytotoxic dihydrobenzophenanthridine alkaloids from the roots of Macleaya microcarpa. Phytochemistry. 2010;71:816-822. DOI: 10.1016/j.phytochem.2010.02.007
  72. 72. Luo Z, Wang F, Zhang J, Li X, Zhang M, Hao X. Cytotoxic alkaloids from the whole plants of Zephyranthes candida. Journal of Natural Products. 2012;75:2113-2120. DOI: 10.1021/np3005425
  73. 73. Bosio C, Tomasoni G, Martínez R, Olea AF, Villena J. Cytotoxic and apoptotic effects of leptocarpin, a plant-derived sesquiterpene lactone, on human cancer cell lines. Chemico-Biological Interactions. 2015;242:415-421. DOI: 10.1016/j.cbi.2015.11.006
  74. 74. Ford CD, Ulloa JL, Catalán CAN, Grau A, Martino VS, Muschietti LV, et al. The sesquiterpene lactone polymatin B from Smallanthus sonchifolius induces different cell death mechanisms in three cancer cell lines. Phytochemistry. 2015;117:332-339. DOI: 10.1016/j.phytochem.2015.06.020
  75. 75. Jimenez-usuga S, Malafronte N, Cotugno R, Leo MD, Osorio E, Tommasi ND. New sesquiterpene lactones from Ambrosia cumanensis Kunth. Fitoterapia. 2016;113:170-174. DOI: 10.1016/j.fitote.2016.07.019
  76. 76. Shulha O, Çiçek SS, Piccolella S, Rárová L, Strnad M, Sönnichsen F, et al. Sesquiterpene lactones from Sonchus palustris L. (Asteraceae, Cichorieae). Phytochemistry. 2020;170:112196. DOI: 10.1016/j.phytochem.2019.112196
  77. 77. Carpesium L, Wang L, Qin W, Tian L, Zhang X, Lin F, et al. Caroguaianolide A-E, five new cytotoxic sesquiterpene lactones from Carpesium abrotanoides L. Fitoterapia. 2016;127:349-355. DOI: 10.1016/j.fitote.2018.03.015
  78. 78. Xu X-K, Ye J, Chen L, Zhang W, Yang Y, Li H. Four new isomeric sesquiterpene lactone dimers from Carpesium faberi. Tetrahedron Letters. 2015;56:6381-6384. DOI: 10.1016/j.tetlet.2015.09.127
  79. 79. Xu X-Y, Sun P, Guo D, Liu X, Liu J, Hu L. Cytotoxic sesquiterpene lactone dimers isolated from Inula japonica. Fitoterapia. 2015;101:218-223. DOI: 10.1016/j.fitote.2015.01.011
  80. 80. Shim JS, Kim JH, Lee JY, Kim SN, Kwon HJ. Anti-angiogenic activity of a homoisoflavanone from Cremastra appendiculata. Planta Medica. 2004;70:171-173. DOI: 10.1055/s-2004-815496
  81. 81. Han N, Yang Z, Liu Z, Liu H, Yin J. Research progress on natural Benzophenanthridine alkaloids and their pharmacological functions: A review. Natural Product Communications. 2016;11:1181-1188. DOI: 10.1177/1934578X1601100838
  82. 82. Garcia-Pineres AJ, Castro V, Mora G, Schmidt TJ, Strunck E, Pahl HL, et al. Cysteine 38 in p65/NF-kappaB plays a crucial role in DNA binding inhibition by sesquiterpene lactones. The Journal of Biological Chemistry. 2001;276:39713. DOI: 10.1074/jbc.M101985200
  83. 83. Ghantous A, Gali-Muhtasib H, Vuorela H, Saliba NA, Darwiche N. What made sesquiterpene lactones reach cancer clinical trials? Drug Discovery Today. 2010;15:668-678. DOI: 10.1016/j.drudis.2010.06.002
  84. 84. Kwok BH, Koh B, Ndubuisi MI, Elofsson M, Crews CM. The anti-inflammatory natural product parthenolide from the medicinal herb feverfew directly binds to and inhibits IkappaB kinase. Chemistry & Biology. 2001;8:759. DOI: 10.1016/s1074-5521(01)00049-7
  85. 85. Zunino SJ, Ducore JM, Storms DH. Parthenolide induces significant apoptosis and production of reactive oxygen species in high-risk pre-B leukemia cells. Cancer Letters. 2007;254:119-127. DOI: 10.1016/j.canlet.2007.03.002
  86. 86. Hung W-L, Suh JH, Wang Y. Chemistry and health effects of furanocoumarins in grapefruit. Journal of Food and Drug Analysis. 2017;25:71-83. DOI: 10.1016/j.jfda.2016.11.008
  87. 87. Kashman Y, Gustafson KR, Fuller RW, Cardellin JH, McMahon JB, Currens MJ, et al. The calanolides, a novel HIV inhibitory class of coumarin derivatives from the tropical rainforest tree, Calophyllum lanigerum. Journal of Medicinal Chemistry. 1992;35:2735-2743. DOI: 10.1021/jm00093a004
  88. 88. Lee YJ, Lee YJ, Im JH, Won SY, Kim YB, Cho MK, et al. Synergistic anti-cancer effects of resveratrol and chemotherapeutic agent clofarabine against human malignant mesothelioma MSTO-211H cells. Food and Chemical Toxicology. 2012;52:61-68. DOI: 10.1016/j.fct.2012.10.06

Written By

Gabin Thierry M. Bitchagno, Vaderament-A. Nchiozem-Ngnitedem, Nadine Tseme Wandji, Guy Cedric T. Noulala, Serge Alain T. Fobofou and Bruno Ndjakou Lenta

Submitted: 10 February 2020 Reviewed: 19 April 2020 Published: 04 June 2020