ArticlePDF Available

Technology Readiness Level Roadmap for Developing Innovative Herbal Medicinal Products

Authors:

Abstract and Figures

Despite the vast global botanical diversity, the pharmaceutical development of herbal medicinal products (HMPs) remains underexploited. Of over 370,000 described plant species, only a few hundred are utilized in HMPs. Most of these have originated from traditional use, and only a minority come from megadiverse countries. Exploiting the pharmacological synergies of the hundreds of compounds found in poorly studied plant species may unlock new therapeutic possibilities, enhance megadiverse countries’ scientific and socio-economic development, and help conserve biodiversity. However, extensive constraints in the development process of HMPs pose significant barriers to transforming this unsatisfactory socio-economic landscape. This paper proposes a roadmap to overcome these challenges, based on the technology readiness levels (TRLs) introduced by NASA to assess the maturity of technologies. It aims to assist research entities, manufacturers, and funding agencies from megadiverse countries in the discovery, development, and global market authorization of innovative HMPs that comply with regulatory standards from ANVISA, EMA, and FDA, as well as WHO and ICH guidelines.
Content may be subject to copyright.
Citation: Pagani, E.; Ropke, C.D.;
Soares, C.M.; Perez, S.A.C.; Benevides,
P.J.C.; Barbosa, B.S.; Carvalho, A.C.B.;
Behrens, M.D. Technology Readiness
Level Roadmap for Developing
Innovative Herbal Medicinal Products.
Pharmaceuticals 2024,17, 703. https://
doi.org/10.3390/ph17060703
Academic Editor: Ilkay Erdogan
Orhan
Received: 19 April 2024
Revised: 15 May 2024
Accepted: 17 May 2024
Published: 29 May 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
pharmaceuticals
Article
Technology Readiness Level Roadmap for Developing
Innovative Herbal Medicinal Products
Eduardo Pagani 1, 2, * , Cristina Dislich Ropke 2, Cristiane Mota Soares 3, Sandra Aurora Chavez Perez 3,
Paulo JoséCoelho Benevides 4,
Barbara Sena Barbosa
2
, Ana Cecilia Bezerra Carvalho
5
and Maria Dutra Behrens
6,
*
1Medical Department, Azidus Brasil, Valinhos 13271-130, SP, Brazil
2Centroflora Group, Innovation Department, Campinas 06460-040, SP, Brazil
3Project Management Office, Vice Direction of Education, Research and Innovation, Institute of Drug
Technology Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil
4Business Office, Oswaldo Cruz Foundation, Campo Grande 79081-746, MS, Brazil
5GMESP, Brazilian Health Regulatory Agency, Agência Nacional de Vigilância Sanitária (ANVISA),
Brasília 71205-050, DF, Brazil
6Natural Products Department, Vice Direction of Education, Research and Innovation, Institute of Drug
Technology Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil
*Correspondence: eduardo.pagani@azdcro.com.br (E.P.); maria.behrens@fiocruz.br (M.D.B.)
Abstract: Despite the vast global botanical diversity, the pharmaceutical development of herbal
medicinal products (HMPs) remains underexploited. Of over 370,000 described plant species, only
a few hundred are utilized in HMPs. Most of these have originated from traditional use, and
only a minority come from megadiverse countries. Exploiting the pharmacological synergies of
the hundreds of compounds found in poorly studied plant species may unlock new therapeutic
possibilities, enhance megadiverse countries’ scientific and socio-economic development, and help
conserve biodiversity. However, extensive constraints in the development process of HMPs pose
significant barriers to transforming this unsatisfactory socio-economic landscape. This paper proposes
a roadmap to overcome these challenges, based on the technology readiness levels (TRLs) introduced
by NASA to assess the maturity of technologies. It aims to assist research entities, manufacturers,
and funding agencies from megadiverse countries in the discovery, development, and global market
authorization of innovative HMPs that comply with regulatory standards from ANVISA, EMA, and
FDA, as well as WHO and ICH guidelines.
Keywords: phytotherapy; technology readiness level; herbal substance; herbal preparation; herbal
medicinal products; drug discovery; drug development
1. Introduction
Natural products (NPs) have historically been the primary source of lead compounds
in drug discovery [
1
]. Living organisms have developed metabolites for hundreds of
millions of years to improve their survival capacity, thus creating chemical diversity [
2
].
NP collections have increased chemical and steric complexity (e.g., more heterocycles and
chiral centers) and display better drug-like properties than their synthetic counterparts,
making them more useful for early-stage screenings [
3
5
]. More than 50% of all marketed
therapeutic small molecules are either NPs, NP derivatives, or synthetic molecules whose
pharmacophoric center was inspired by NPs, including some with traditional use [6,7].
Herbal preparations (HP) comprise hundreds of phytochemicals [
8
,
9
] that could
address medical needs through multitarget mechanisms [
10
13
]. The possible synergy of
multiple compounds, within a phytocomplex proposed decades ago, has been substantiated
by advancements in systems biology and “-omics” [14].
Herbal medicinal products (HMPs) are a niche of NPs [
15
]. It is estimated that up
to 80% of the world’s population relies on herbal medicines and medicinal plants as a
Pharmaceuticals 2024,17, 703. https://doi.org/10.3390/ph17060703 https://www.mdpi.com/journal/pharmaceuticals
Pharmaceuticals 2024,17, 703 2 of 23
primary source of healthcare [
16
], which can be beneficial in several non-communicable
diseases [
17
]. The global herbal market was USD 60 billion in 2010 and is expected to reach
USD 5 trillion by 2050 [
18
]. If properly managed, this market growth can generate income
and convert local populations into conservation agents [19].
The world has currently 377,749 accepted plant species, mostly occurring in megadi-
verse countries [
20
]. Only 15% have been characterized phytochemically and 6% pharma-
cologically [
21
,
22
]. The number of species used in HMPs registered by the Food and Drug
Administration (FDA), European Medicines Agency (EMA), and the Brazilian Health Reg-
ulatory Agency (ANVISA) is just a few hundred, and most like Ginkgo biloba,Panax ginseng,
and Echinacea purpurea have been known for centuries. This occurs despite regulations
that have existed for decades in Brazil, the EU, and the USA permitting the registration of
innovative HMPs.
Among the 17 megadiverse countries, only two, namely the USA and China, have a
consistent track record of pharmaceutical innovation. Encouraging the development of
innovative HMPs originating from other countries could potentially stimulate their scien-
tific and socio-economic progress, safeguard biodiversity, and introduce novel therapeutic
options for medical needs [2325].
The absence of innovation may partly stem from legal challenges. Sanitary regulations
differ across countries [
26
,
27
], and there exist legal uncertainties in the operationalization
of the Nagoya Protocol [
28
,
29
]. Brazil has addressed both issues. ANVISA has issued
a regulatory framework for HMPs aligned with EMA [
30
]. The Brazilian Biodiversity
Law [
31
] aligns with the Nagoya Protocol, offering legal certainty for the assessment of
genetic resources and traditional knowledge, and benefit sharing [
28
]. Also, attempts by
the European Community (EC) and the World Health Organization (WHO) provide models
for multinational harmonization [32].
However, despite these advancements, innovations from Brazil and other biodiverse
countries remain scarce, indicating that a significant factor may still be missing. The devel-
opment of HMPs is a lengthy and intricate process that relies on the effective management
of various capabilities and long-term funding. Furthermore, adaptations to previously
planned tasks are often necessary. Given these challenges, we believe that a comprehensive
yet adaptable roadmap outlining this process could assist research institutions, pharma-
ceutical companies, and funding bodies in establishing consistent, long-term partnerships
aimed at fostering innovation.
NASA proposed the technology readiness levels (TRLs) decades ago to guide man-
agers in assessing the aeronautical technology’s readiness and risks at specific development
key points [
33
,
34
]. The concept has spread over many innovation fields and is currently
used in pharmacology by development-fostering agencies [
35
39
]. TRLs fit very well in
the proposal of a roadmap providing hierarchized development concepts and enabling a
step-by-step approach.
Almost no peer-reviewed publication specifically covering TRLs in pharmaceutical
development is available, either for small molecules, HMPs, or biotechnological products.
Moreover, most published TRL scales have general concepts and examples, but lack descrip-
tors defining boundaries, leaving room for inconsistent subjective choices that might vary
among projects. Such a problem was detected and addressed in the chemical industry [
40
].
To face these challenges, we propose a TRL-guided HMP development roadmap
applicable to Brazilian innovations aiming at worldwide marketing. It is organized in a
step-by-step set of actions grouped into six domains, defined by clear boundaries, and
compliant with international guidelines.
This proposal applies to HMPs, based on herbal preparations (HPs) as active phar-
maceutical ingredients (API), and not to isolated or purified substances. It also applies to
plants not included in official monographs, regardless of previous traditional use. For the
development of HMPs based on traditional or well-established use, we refer the reader to
the following references [
41
47
]. Moreover, for the innovations addressed here, previous
ethnopharmacological information is helpful, but not mandatory.
Pharmaceuticals 2024,17, 703 3 of 23
2. Results and Discussion
Following the original Mankins style [
33
,
48
,
49
], we discuss each technology readiness
level (TRL) main attribute, citing examples and providing brief descriptions of associated
costs and funding sources. Table 1outlines the primary deliverables for each level defined
by Mankins, alongside our proposed adaptations for herbal medicinal products (HMPs).
Additionally, Table 1provides a visual summary of the key concepts and tasks associated
with each TRL as applied to the development of HMPs.
Table 1. Technology readiness level roadmap according to Mankins [
23
,
34
,
40
] and its adaptation to
herbal medicinal product development (HMP).
Mankins Basic principles observed and reported
TRL-1 HMP Pharmacological effect of an herbal preparation observed and reported
Mankins Technology concept and/or application formulated
TRL-2 HMP Technological application formulated and proof of concept planned
Mankins Analytical and experimental critical function and/or characteristic proof of concept
TRL-3 HMP Proof of concept of herbal preparation (API candidate)
Mankins Component and/or breadboard validation in a laboratory environment
TRL-4 HMP Optimization of herbal preparation in bench-scale and non-GLP validation
Mankins Component and/or breadboard validation in a relevant environment
TRL-5 HMP Optimization of herbal preparation in pilot-scale and GLP validation
Mankins System/subsystem model or prototype demonstration in a relevant environment (ground or space)
TRL-6 HMP Herbal medicinal product prototype completes phase I clinical trial
Mankins System prototype demonstration in a space environment
TRL-7 HMP Herbal medicinal product completes phase II clinical trial
Mankins Actual system completed and “flight qualified” through test and demonstration (ground or space)
TRL-8 HMP Herbal medicinal product completes phase III clinical trial, and the dossier is approved by one
regulatory authority
Mankins Actual system “flight-proven” through successful mission operations
TRL-9 HMP Herbal medicinal product pharmacovigilance and phase IV clinical trials are performed
2.1. TRLs for Herbal Medicinal Products
2.1.1. TRL-1
TRL-1 was described as “basic science envisioning an application” [
33
,
49
51
]. For
chemicals, it was summarized as a “concept” [
40
]. BARDA [
38
] describes it as “active
monitoring of scientific knowledge database”.
For HMPs, the starting point is the identification and reporting of an herbal substance
(HS) or herbal preparation’s (HP) pharmacologically relevant effect. This comes either
from an ethnopharmacological observation or laboratory experimentation. Both demand
envisioning a connection between an effect and a medical need [
35
]. Examples of these
two independent possibilities are: 1- an ethnopharmacological description of a plant
infusion with psychic effects, and 2- an experimental finding of vasodilating properties
of a plant extract. The associated costs typically involve academic research expenditures,
encompassing both experimental work and field-based ethnopharmacological surveys.
Funding for this stage is commonly provided by basic science funding agencies.
2.1.2. TRL-2
TRL-2 “formulates the technology concept or application, with no experimental proof
or detailed analysis supporting the conjecture” [
33
,
49
,
52
]. The R&D fostering agencies
agree on “developing hypotheses and experimental designs” [
35
38
]. Therefore, TRL-2
creates a project focused on a product.
For HMPs, we propose that TRL-2 devises a technological application and delivers a
proof-of-concept (PoC) research project based on the literature and experimental data. It
starts addressing issues on the HS and/or HP and chooses at least one experimental model
with translational capability. The literature review must be comprehensive and preferably
include some data produced by the responsible research group. This difference from the
Pharmaceuticals 2024,17, 703 4 of 23
original NASA concept is due to the higher dependence of pharmaceutical development
on previous experimental data to design a reliable PoC.
The progression of the two examples mentioned at TRL-1 involves the formulation of
research projects aimed at testing the antidepressant potential of the plant with psychologi-
cal effects or the antihypertensive potential of the extract with vasodilating properties. The
associated costs typically fall within the scope of small- or medium-sized academic projects
sponsored by basic-science funding agencies.
2.1.3. TRL-3
TRL-3 delivers an “analytical and experimental critical function and/or characteristic
proof of concept” [
33
,
49
,
52
]. The R&D fostering agencies agree on the “proof of concept”,
and BARDA adds identifying a target or candidate and demonstrating relevant
in vitro
activity [3538].
For HMPs, we propose that TRL-3 performs the R&D fostering agencies (PoC) on a
bench scale according to the research plan described in TLR-2. The results must justify the
progression to TRL-4 and may demand more than one assay. The first example progresses
to the performance of an
in vitro
test for serotonin reuptake and/or an animal model for
depression [
53
]. The second could be an
in vitro
vasodilation test and/or an animal model
for systemic arterial hypertension [
54
]. As mentioned above, the associated costs typically
fall within the scope of small- or medium-sized academic projects sponsored by basic
science funding agencies.
2.1.4. TRL-4
TRL-4 goes further in the validation. It must support the concept formulated earlier
and be consistent with the requirements of potential system applications [
33
,
49
,
52
]. Moor-
house also adds “credible for design and performance conditions” [
55
]. The R&D fostering
agencies focus on testing in a defined laboratory or animal model, and BARDA specifically
mentions “optimization” [3538].
For HMPs, we propose HP optimization on a bench scale and broad validation through
several independent assays. TRL-4 is the most laborious and complex stage up to this
point and may involve the work of different groups for years. It holds the first major
risk inflection point by testing optimized HPs in the most translational disease models.
Here, both examples converge to a chemically defined HP with characterization and
quantification of constituents with therapeutic activity or marker compound(s) and several
in vitro
and
in vivo
tests. The costs and timeliness rise substantially. The challenges
might be faced by academic centers alone, or in collaboration with manufacturers and/or
contract research organizations (CROs). The costs might be public-sponsored by large-
sized academic research projects and may start involving private resources from investors
and manufacturers.
2.1.5. TRL-5
TLR-5 performs “component or prototype validation in a development-relevant envi-
ronment” [33,34,49,50,52,55].
For HMPs, we define “component” as a candidate HP produced through optimized
and standardized processes. “Validation in a development-relevant environment” is
equated with conducting “Good Laboratory Practice (GLP) non-clinical studies” [
35
38
].
GLP testing commences only when the phytochemical profile of the HP candidate is well
defined, achieved through standardized preparation procedures and bio-guided studies.
Hence, TRL-5 encompasses two primary tasks: scaling up from the bench to the
pilot scale and conducting GLP animal testing. These activities are usually carried out in
specialized facilities, including CROs. Costs associated with TRL-5 are generally within the
same range as those of TRL-4. While some public funding sources may still be available,
the reliance on investors and manufacturers tends to increase.
Pharmaceuticals 2024,17, 703 5 of 23
2.1.6. TRL-6
TRL-6 refers to a “system/subsystem model or prototype demonstration in a relevant
environment (ground or space)” [33,34,49,50,52,55].
For both drugs and HMPs, the “relevant environment” corresponds to the “first-in-
human” testing in phase I clinical trials conducted under Good Clinical Practices (GCP),
typically conducted in a medical facility [
35
38
]. Costs escalate significantly due to the
manufacture of the candidate HMP for human use and GCP testing. While some public
funding sources may still be available, the reliance on investors, manufacturers, and banks
further increases.
2.1.7. TRL-7
TRL-7 is the “system prototype demonstration in the expected operational envi-
ronment” [
33
,
34
,
49
52
,
55
]. There is a clear parallel between the “expected operational
environment” and the phase II clinical trials conducted with patients with the condition
potentially treated by the HMP candidate [3538].
This TRL holds the second major risk inflection point because, in the case of success,
the risks of failure in further stages are reduced. In case of failure, the project shall be
aborted or deeply revised. The costs keep rising. Because the technological risks are still
high, some public funding might be expected, along with a higher amount of private capital
from investors, manufacturers, and banks.
2.1.8. TRL-8
In the TLR-8, the “actual system is completed and qualified through test and demon-
stration” [
33
,
34
,
49
52
,
55
]. For pharmaceutical development, it means that an Investiga-
tional New Drug (IND) is reviewed and approved by the regulatory agency.
The parallel with phase III clinical trials is evident. This stage is designed to generate
statistically significant evidence of HMP efficacy and safety in large patient populations
with the target disease [
35
38
]. The HMP used is that intended for marketing authorization.
Costs associated with TRL-8 are the highest of the development process. TRL-8 alone can
incur expenses equivalent to those of TRLs 1 to 7combined. However, risks could be lower
compared to TRL-7. Costs are typically privately sponsored by manufacturers, along with
contributions from investors and banks.
2.1.9. TRL-9
TRL-9 is an “actual system flight-proven through successful mission
operations” [
33
,
34
,
49
52
,
55
]. This was translated by “post-marketing studies and surveil-
lance of the HMP after approval for use” [
35
38
]. By definition, all technologies that
succeed in being applied in actual systems go eventually to TRL-9.
In certain instances, new formulations may be developed with higher or lower concen-
trations, slow-release properties, or tailored for pediatric use, necessitating additional clini-
cal trials (phase IV or post-marketing). Despite the potential inclusion of phase IV clinical
trials, costs typically decrease substantially and are usually sponsored by
the manufacturer.
2.2. TRL Domains and Boundaries
Given the complexity of HMP development, we established six technological domains
whose progression facilitated the delineation of the boundaries for technology readiness
levels (TRLs): herbal substance, herbal preparation, herbal medicinal product, analytical
development, non-clinical assays, and clinical trials. Table 2summarizes the deliverables
and main regulations applicable for HMPs at each level.
Pharmaceuticals 2024,17, 703 6 of 23
Table 2. Main deliverables of each TRL for each HMP development domain with main regulations
applicable.
HS HP HMP AD NCA CT
TLR1 Initial literature review and data collection
TLR-2
Addressing
botanical
inconsistencies
Comprehensive
literature review
Compatibility
with vehicles
Initial
parameters’
proposal
Comprehensive
literature
review
Comprehensive
literature
review
TLR-3
Certified
botanical
identification
Best extract under
current conditions
Advance
solubility and
compatibility
issues
Best characteri-
zation under
current
conditions
Optimal PoC
under current
conditions
TLR-4
Full traceability
supply-chain
development
Bench-scale
optimization and
semi-industrial
scale initiation
Advances
formulation for
pre-clinicals
Consistent char-
acterization
Efficacy and
safety
confirmation
and bio-guided
HP
optimization
Update
literature
review
TLR-5 Supply chain
consolidation
Semi-industrial
scale optimization
Advance
formulation for
human use
Validated
methods for HP
GLP tests for
phase I CT
authorization
Update
literature
review
TLR-6 Stability
assessment
GMP-compliant
batches
GMP-
compliant
batches
Validated
methods for HP
and HMP
GLP tests for
phase II CT
authorization
GCP phase I
clinical trial
TLR-7
Industrial scale
and GACP im-
plementation
GMP-compliant
industrial batches
GMP-
compliant
batches
Validated
methods for HP
and HMP
GLP tests for
long-term use
GCP phase II
clinical trials
TLR-8 GACP
compliance
GMP-compliant
industrial batches
GMP-certified
batches
Validated
methods for HP
and HMP
GLP tests for
long-term use
GCP phase III
clinical trials
TLR-9 GACP certified
GMP-compliant *
or certified
industrial batches
GMP-certified
batches
Validated
methods for HP
and HMP
GCP phase IV
trials and
pharmaco-
vigilance
HS: herbal substance; HP: herbal preparation; HMP: herbal medicinal product; AD: analytical development; NCA:
pre-clinical assays; CT: clinical trials; GACP: Good Agricultural and Collection Practices; GLPs: Good Laboratory
Practices; GCP: Good Clinical Practices; The Nagoya Protocol applies to the HSs and HPs in different forms in
different countries. * The demand for GMP-compliant or GMP-certified HP at TRL-8 varies in different countries.
2.2.1. Herbal Substance
HMPs’ TRLs 1 and 2rely on a comprehensive literature review, eventually supple-
mented by any original data provided by the responsible researcher. Older publications
can be valuable if they contain human data. However, these reports may suffer from miss-
ing, outdated, or unreliable botanical data, inconsistencies in subspecies, or phenotypical
variations. Researchers must diligently address these inconsistencies, striving to include all
potentially relevant information while highlighting sources of confusion and detailing how
these issues were resolved to ensure reliable botanical identification. Additionally, data on
species within the same genus may be useful in predicting the expected phytochemical and
biological profiles of the chosen plant [
56
]. TRL-2 also involves gathering information on
genetic resources and assessments related to traditional knowledge, in compliance with the
Nagoya Protocol [29].
For TRL-3 proof of concept (PoC), botanical identification must be conducted by a
certified botanist using a voucher sample, ideally including the flower, which should be de-
posited in an official herbarium. The collection process must be meticulously documented,
including GPS location, date, time, and weather conditions (preferably dry) at the time
Pharmaceuticals 2024,17, 703 7 of 23
of collection, as well as phenological information and the quantity collected. Records of
conservation conditions (light and humidity) are of utmost importance. The quantity stored
must be sufficient to facilitate counter-proof tests until the completion of TRL-4. Henceforth,
the species should not change, except for official naming updates. If there is a change in
the species, an impact analysis must be conducted to determine which phytochemical and
biological tests need to be repeated.
TRL-4 needs HS full traceability, following the procedures outlined for TRL-3. To
optimize conditions, gathering various batches of HSs from different locations and during
different seasons is essential. This process is time-consuming and progresses from TRLs
4to 7, making an early start crucial. Each batch must be of sufficient quantity and stored
appropriately to facilitate repeated assessments for counter-proof tests until the conclusion
of TRL-7.
TRL-4 also involves addressing the HS supply chain. Sustainable collection practices
are viable if the plant is abundant in the wild and collection methods do not harm the
environment. This approach may even yield environmental benefits by engaging rural
populations as conservation agents [
57
]. Agroecological approaches and organic agriculture
offer viable options as well. Strains can be selected, domesticated, and cultivated on a
small scale to initiate the development of a cultivar. Plant strains exhibiting the desired
metabolite profile and biological activity can be chosen for pilot-scale cultivation, typically
encompassing less than one hectare [58,59].
TRL-5 achieves consistency in production type (collection or cultivation), selects the
optimal strain, collection time, and implements effective conservation practices which are
paramount for scalability. This stage also initiates the development of Good Agricultural
and Collection Practices (GACP) and the qualification of suppliers. Rigorous control of
production’s initial stages is imperative, encompassing GACP and Good Herbal Processing
Practices (GHPP) [
60
,
61
]. From TRL-5 onwards, the HS must undergo quality controls to
ensure plant identification, chemical profiling, and detection of impurities (including other
materials and plants). Any deviations in the supply chain or composition need an impact
analysis on the product. Actions to increase the production scale commence at this stage,
continuing through TRL-7 or 8. Initial non-clinical GLP tests can be conducted with pilot
batches, while long-term pivotal tests and clinical trials are preferably performed using
industrial batches.
At TRL-6, the focus shifts to consolidating the supply chain and finalizing composition
tests. By TRL-7 the HS production scales up to industrial batches, ranging from hundreds of
kilograms to tons, depending on the equipment used. A qualification master plan becomes
necessary for phase II clinical trials [
62
,
63
], and Good Agricultural and Collection Practices
(GACP) must be implemented to provide HS batches for use in phase II and III clinical
trials [61].
At TRL-8, the raw materials utilized must be those intended for HMP registration.
Proof of compliance with GACP guidelines is essential. While formal GACP certification is
not mandatory in Brazil, it is required in the European Community to simplify assessment
and provide assurance to regulatory bodies [
61
]. The FDA mandates compliance with the
full GACP standard for an IND PH3 Investigational New Drug, as outlined in 21 CFR
321.23. Certification is only necessary for the New Drug Application phase [64].
2.2.2. Herbal Preparation
TRL-1 ideally provides some information on the extractive methods and the extract’s
composition. At TRL-2, a thorough literature review should encompass all available
information from scientific and patent databases concerning methodological variations and
hypotheses regarding active, toxic, and inert components. Initiating a bio-guided study
aimed at isolating and identifying or characterizing constituents with therapeutic activity
or markers is advisable at this stage. If conducted, high-throughput and high-content
screenings (HTS, HCS) of extracts or fractions commence here and continue throughout
TRL-3 [22,65].
Pharmaceuticals 2024,17, 703 8 of 23
At TRL-3, a PoC must use the best possible extract, considering the literature review,
the investigators’ capabilities, and the budget available. The quantity produced shall be
sufficient for repetitions and comparisons until the conclusion of TRL-4. The extractive
method must be thoughtfully described and justified. Preferably, the solvents should be
compatible with a future industrial scale-up; otherwise, some tests may need to be repeated.
HP candidates must undergo chemical characterization to the fullest extent possible with
the available resources. The bio-guided study should identify constituents with known
therapeutic activity, markers, or at least the active phytochemical classes.
At TRL-4, the focus is on optimizing extractive conditions at a bench scale to maximize
active components while minimizing inert or potentially toxic ones [
66
,
67
]. This includes
testing various equipment, solvents, techniques, and drying methods [
68
70
]. The extracts
undergo evaluation through a series of biological assays to confirm their pharmacological
activity [
69
,
71
]. TRL-4 is considered complete only when the chemical profile of the herbal
preparation (HP) candidate is optimized at the bench scale through bio-guided studies.
Additionally, attention is given to HP stability. Adequate samples must be stored for
repeated testing until TRL-7 or 8. The entire process must be thoroughly described, and
storage conditions must be traceable and auditable.
At TRL-5, the HP candidate production is scaled up to pilot or semi-industrial quan-
tities. The challenge lies in maintaining or improving the characteristics selected in the
previous TRLs. The pilot process must be robust, reproducible, and capable of delivering a
stable product. This lays the foundation for implementing Good Manufacturing Practices
(GMP) for the HP, which are required for regulatory-compliant Good Laboratory Practice
(GLP) preclinical studies. Typically, this process is carried out by companies specialized
in HP manufacture and should adhere to GMP, or in some countries, be GMP-certified.
Development of the extractive method is finalized with at least three pilot batches, each
comprising at least five kilograms of dry extract, accompanied by the respective analytical
reports [16,47,72].
The industrial batches are developed during TRL-6 and TRL-7. The phases I and II
clinical trials can be run with either pilot or industrial batches manufactured in compliance
with GMP, with validation and certification processes initiated [
38
,
73
]. The TRL-8 phase III
clinical trial must be performed with GMP-certified industrial batches identical to those
intended for licensing [
74
]. Information on batch-to-batch variations and the demonstration
that the API (HP) is safe and effective within the tolerated variability is required [74].
Herbal Preparation Concepts in Europe and Brazil
In Europe and Brazil, extracts can be classified into three categories, according to
the European Pharmacopoeia 11th edition [
41
,
75
] and the Brazilian Pharmacopoeia 6th
edition [76]:
Standardized extracts (i), in which constituents with therapeutic activity are known.
The amount of native or genuine (without excipients) extract is variable, adjusted within the
range defined for the active component—the adjustment is carried out with inert excipients
or by blending production batches with a higher or lower content of active constituents,
resulting in a variable amount of native HP (e.g., Atropa belladonna). Quantified extracts (ii),
which are adjusted within a defined range of compounds (active markers), whose relation to
the activity is proved through clinical trials, and, therefore, generally accepted to contribute
to the therapeutic activity. The amount of native (genuine) extract is constant and the
adjustment can only be achieved by blending extract batches with the same specification
and based on a constant amount of native extract (e.g., Ginkgo biloba). Other extracts
(iii) are mainly defined by their manufacturing process (state of the drug to be extracted,
solvent, extraction conditions) as well as their specification. There is no adjustment for a
constituent or group of constituents since the active substance is the native extract, whose
amount is constant. The constituents with known therapeutic activity or markers are not
known. Therefore, analytical markers are indicated, and their contents are batch-specific,
Pharmaceuticals 2024,17, 703 9 of 23
informative, or recommended to have a minimum content referred to (e.g., Valeriana
officinalis). In the case of traditional use, the extraction process should not change.
2.2.3. Herbal Medicinal Product
This domain encompasses administration route, formulation, intellectual property
(IP), and partnerships. TRL-2 and TRL-3 focus on HP solubility and compatibility with
vehicles for both
in vitro
and
in vivo
testing. In addition to academic sources, conducting a
preliminary review of patent databases is advisable to assess freedom to operate.
At TRL-4, the physical form and administration route of the HP are defined, and
formulation development commences after gathering information on API solubility and
stability [
77
]. Seeking professional advice on intellectual property is advisable to ensure
freedom to operate and explore opportunities for patenting formulations that enhance API
delivery and stability. For academic projects, this is an opportune time to explore potential
partnerships with manufacturers.
At TRL-5, GLP tests are conducted with a formulation as close as possible to that
intended for human use, considering animal testing peculiarities. Solid formulations for
oral use, such as pills and capsules, cannot be directly administered to animals. Instead,
GLP guidelines for oral toxicity studies permit administration via gavage, dissolved in
drinking water, or mixed with food [
78
]. Additionally, Brazilian guidelines allow oral tests
to be conducted with the HP [
79
]. Typically, partnerships with manufacturers are already
in operation, and intellectual property (IP) issues have been addressed.
TRL-6 phase I and TRL-7 phase II clinical trials must be performed with the HMP
manufactured in compliance with GMP and stability tests compatible with the clinical
trials’ duration. The investigational product manufacture happens in a GMP-like envi-
ronment but is not necessarily GMP-certified [
38
,
73
]. Still, there might be variations in
the dose/concentration of the product that goes to the market. If the claim is based on
traditional information, traditional doses and concentrations must be followed.
At TRL-8, phase III clinical trials are conducted with the GMP-certified formulation in-
tended for marketing authorization, ensuring stability throughout the study
duration [16,38,73].
If there were differences between the formulation used in phase II and phase III clinical
trials, the impact on safety and efficacy must be addressed and the modifications justified.
For dossier submission, stability tests must be finalized for at least three HMP batches in
their packaging. The Nagoya Protocol mandates notification of the final product’s genetic
resources and associated traditional knowledge assessment [29].
At TRL-9, the HMP is finalized. However, it is feasible to develop new formulations
for alternative routes, varying concentrations, slow-release, or pediatric use, which typically
require new clinical trials for marketing authorization.
2.2.4. Analytical Development
TRL-2 proposes parameters for HS and HP characterization. For known HSs, the best
sources are official pharmacopoeias or compendia [
45
,
46
,
62
,
76
,
80
83
]. Each country has its
list of official legally binding pharmacopoeias. For innovative HS, the pharmacopoeias and
scientific publications provide insights on standards to follow. HS characterization typically
includes descriptions of the plant part(s) used, purity, chemical content, contaminants,
etc. [
84
86
]. The HP candidate usually contains substances or chemical classes associated
with therapeutic or toxic effects, which can serve as positive and negative markers for HP
optimization and quality control. It is recommended to perform a bio-guided isolation of
bioactive compounds, especially when commercial standards are unavailable.
At TRL-3, analytical methods must be employed for HS and HP characterization by
techniques such as thin-layer chromatography (TLC), high-performance liquid chromatog-
raphy (HPLC), gas chromatography (GC), and mass spectrometry (MS). A fingerprint
chromatogram with all peaks and times, along with the identification of some components,
is recommended.
Pharmaceuticals 2024,17, 703 10 of 23
TRL-4 requires HPs’ comprehensive qualitative and quantitative characterization
by different methods produced from several HS batches. Bio-guided identification of
active compounds and markers progresses, alongside high-throughput screening (HTS)
or high-content screening (HCS), if applicable. The analysis should identify the major
chromatographic peaks. Similar objectives can be achieved through MS virtual libraries.
Hyphenated methods such as HPLC-PDA, UHPLC-PDA-MS, HS-SPME-GC-MS, or GC-
FID/MS are increasingly used [
87
89
]. Knowledge of positive and negative markers should
increase through bio-guided studies aimed at compound isolation, especially for species
lacking traditional use or literature data. All HPs tested thereafter must have analytical
reports in the CTD format. ANVISA and EMA have published guidelines adapted to
HMPs [9092].
TRL-5 needs protocols for HS and HP quality control. The analytical methods must
be either pharmacopoeic or independently developed and validated addressing speci-
ficity/selectivity, linearity, working range, precision, accuracy, detection/quantification
limits, and robustness [
81
,
93
,
94
]. In Brazilian regulations, these methods can replace the
phytochemical assays for quality control, when technically justified [95].
From TRL-6 onwards, the requirements expand to include the evaluation of formula-
tions for human use, as well as stability testing. At this stage, active ingredients or markers
with known therapeutic effects are identified, and all analytical methods must adhere to
pharmacopoeial standards or be thoroughly validated. Additionally, a comprehensive
chromatographic profile (fingerprint) is established, alongside stringent analysis of po-
tential contaminants, including pesticides, heavy metals, adulterants, microorganisms,
and toxins. These analyses must conform to the regulatory requirements of the target
country for licensing, primarily following official pharmacopoeias. For TRL-8, stability
testing is conducted on products in their final packaging. TRL-9 uses the methodologies
established in previous stages, applying them to every industrial batch. New analytical
methods are developed or adapted as necessary to accommodate any new formulations
eventually introduced.
2.2.5. Non-Clinical Assays
The purpose and number of non-clinical assays for innovative HMPs depend on the
documented human exposure. In all cases, TRL-1 relies on some previous data. TRL-2
performs a comprehensive literature review, including all relevant
in vitro
and
in vivo
data on HSs, HPs, and HMPs from the same species. In some cases, data on other species
from the same genus may also be helpful. The review embraces decades of information in
pharmacopoeias, scientific journals, theses, books, websites, congress abstracts, and others.
If available, it is advantageous to include data produced by the development team.
The PoC experiment characterizes TRL-3. It includes
in vitro
assays of relevant targets
and/or
in vivo
disease models. Appropriated negative and positive controls are mandatory.
The assay(s) choice shall be justified, based on the potential for extrapolation to humans,
and described in detail, including allometric dose transposal. The report(s) must address
data quality and sufficiency to progress to the next level.
At TRL-4, assays increase in quantity, quality, and translational capability. The hy-
potheses on active compounds and MoA advance, helping efficacy and safety assess-
ments [
96
].
In vitro
, multitarget screening helps to elucidate the MoA and discloses off-
target effects [9799].
Extracts, fractions, and pure compounds must be tested to identify those participating
in the biological effects. The impact of minor and unknown components must be addressed.
A set of tests shall be developed for bio-guiding the HP optimization. These should be easy
to run (preferably
in vitro
) and may evolve as knowledge accumulates. Dose–response
curves for different HPs and active isolated compounds shall be produced. Confirmatory
assays examining the same and other related pathways and disease models in different
species are advisable [
100
102
]. Collaboration between groups mastering different methods
is encouraged.
Pharmaceuticals 2024,17, 703 11 of 23
Whenever structural information is available, fast and low-cost in silico tests are also
recommended. Its relevance grows as quantitative structure–activity relationship (QSAR)
modeling and chemical databases improve [
69
,
103
]. Also, some tests integrating responses
of various compounds in mixtures are under development [104106].
The careful selection, performance, interpretation, and reporting of the best models
are of foremost importance. Animal model predictions are often not confirmed in clinical
trials [
107
109
]. Therefore, the potential for extrapolating the conclusions to humans
shall be reviewed and deepened. Experiments must address aspects of efficacy, toxicity,
and capability to reach the target tissues/organs (PK). The report should discuss the
experiments’ strengths and weaknesses for translation to human outcomes. This includes
the relevance of the experimental models, the allometric dose transposal, and comparisons
with the controls.
The authors recommend performing the “killer experiment” as soon as possible after
the PoC, at the end of TRL-3 or the beginning of TRL-4. It aims at “killing” an unviable
technology before significant resources are spent. Its performance builds trust that, once
optimized, the technology will be competitive in the market [110].
TRL-5 focuses on regulatory-demanded GLP animal safety and toxicology
tests [78,79,111].
A peculiarity of HMPs is the possibility of waiving some non-clinical tests if sufficient data
from other sources are available [112,113].
For HMPs with a history of human use, the requirement for GLP toxicology studies is
determined by how closely the new HP matches the one previously used by humans. The
comparison encompasses composition, clinical application, exposure (dose and duration),
and the frequency and severity of any known adverse reactions [
16
,
79
]. Nonetheless,
genotoxicity and reproductive toxicity, whose risks are difficult to detect clinically should
be addressed, but exemptions apply [
79
,
114
,
115
]. The potential interaction with other
medicines also needs to be addressed [116,117].
For innovative HMPs, with no prior human exposure, most regulatory agencies
demand the same set of GLP toxicological tests required for small molecules, starting at
TRL-5 and finishing at TRL-7 or TRL-8 [79,118].
At TRL-5, tests needed for phase I clinical trial authorization include cytotoxic-
ity, genotoxicity, local tolerance (if applicable), acute toxicity, short-term repeated dose
toxicity, and safety evaluations of the cardiovascular, respiratory, and central nervous
systems [16,79,111,119].
TRL-6 encompasses tests necessary for phase II clinical trial authorization, specifically
repeated dose toxicity and reproductive toxicity studies [
62
,
111
]. Additional tests may be
required like the HMP batch-to-batch quality control biological test according to the United
States Pharmacopeia, chapters 1032 and 1033, required by the FDA [
120
,
121
]. Its development
starts at TRL-4 and finishes at TRL-8. Another example is the pharmacokinetic (PK) and
pharmacodynamic data (PD) demanded by the EMA for standardized HPs, but not for
quantified and other HPs. PK and PD data may be difficult to obtain if the effect depends
on mixtures but must be obtained in standardized extracts for at least one constituent with
known therapeutic activity. Also, the FDA states that “if feasible, chemical constituents
of a drug product that contribute to toxicity or pharmacology should be assessed in the
pharmacokinetic/toxicokinetic studies” [
62
]. These PK and PD data are not required for
HMPs by most other regulatory agencies [16,79,92].
If applicable, TRL-7 performs the tests demanded for continuous or long-term use
such as chronic toxicity. Carcinogenicity is required if there is evidence of genotoxicity, of
carcinogenicity in repeated dose toxicity, or if the composition holds a structure related to
a known carcinogen [
79
,
111
,
122
]. These must start before the phase III clinical trials and
have enough interim data to support the duration of these trials. Complete data will be
required for HMP licensing [62].
At TRLs 7 and 8, regulators accept in silico testing for assessing the safety of minor
components in standardized extracts. This method is particularly valuable when the
Pharmaceuticals 2024,17, 703 12 of 23
molecular structure is known and traditional toxicological
in vivo
or
in vitro
tests are
impractical [123125].
Allometric Dose Conversion
Certain biological effects occur
in vitro
and
in vivo
at doses exceeding those feasible
for human use, making the findings clinically irrelevant. Allometric dose conversion
is employed to achieve comparable target tissue concentrations across different species.
For an accurate calculation, it is advised to adjust the methodologies used for isolated
substances to HMPs [126,127].
Killer Experiment
The “killer experiment” is aimed at stopping a project with low chances of success
before consuming significant resources. If a well-designed and well-conducted killer
experiment does not demonstrate the project’s unfeasibility, this is a compelling argument
for its continuity [110]. Examples of “killer experiments” adapted for HMPs are:
a.
Demonstration of low efficacy in doses suitable for humans after allometric calculation
in the gold-standard model of the target disease.
b.
Demonstration of efficacy/toxicity balance worse than a comparator under develop-
ment or already in the market, even after optimization. Note: if the MoA is different
from the available comparators, the product may still be viable, even if the potency
is lower or the toxicity is higher, because it might work in cases not responsive to
the comparator.
c.
Demonstration that a component responsible for a large part of the effect does not
reach an effective concentration in the target tissue/organ in a dose suitable for
humans after formulation optimization.
d.
Demonstration of active components’ instability, except for unmet medical needs. For
these cases, the search for stable related compounds is advisable.
e. Demonstration of manufacturing costs incompatible with the therapeutic indication.
2.2.6. Clinical Trials
Clinical trials for INDs have robust international regulations, which are out of the
scope of this paper [
118
,
128
]. The specific demands for HMPs depend on the degree of
innovation. In all cases, TRL-2 performs a comprehensive review of all available sources,
looking for traditional use, pharmacovigilance, clinical trials, accidental intoxications, and
other reports on human use. Literature-based comparisons with existing drugs for the
same indication are advisable to reinforce the medical utility of the proposed innovation.
The review must be updated at TRL-5, before starting clinical trials.
For innovative HMPs, with no prior human exposure, most regulatory agencies follow
the IND regulations [
118
]. In cases with reliable data on prior human use, some IND
requirements could be exempt. The sponsor is recommended to contact the regulatory
authority to obtain advice on the clinical development plan [62].
At TRL-6, phase I clinical trials address aspects of safety and PK for the constituents
with known therapeutic activity. The FDA and EMA have specific recommendations
applicable to first-in-human studies which are worth consulting [
73
,
129
]. For HMPs,
whenever enough information is available from traditional use, phase I clinical trials in
normal volunteers are usually waived. A substantial prior human use of the HMP generally
conveys confidence that similar amounts are safe for small numbers of carefully monitored
subjects in phase II trials [
16
,
62
]. However, the FDA recommends getting PK parameters to
achieve the same objectives of clinical pharmacology studies for nonbotanical drugs, if the
major active constituent(s) in a botanical product is(are) known [62].
At TRL-7, phase II clinical trials explore efficacy, safety, and dose-finding in small
populations. If the phase I clinical trial is waived and phase II is the first-in-human test,
consult the FDA and EMA-specific regulations [
73
,
129
]. This is one of the most critical
stages of pharmaceutical development, often referred to as the “valley of death”, where
Pharmaceuticals 2024,17, 703 13 of 23
most clinical failures are seen [
97
]. Although generally regarded as a PoC in humans, the
phase II clinical trials do not necessarily produce statistically significant results. Instead,
they provide information suggestive of efficacy that enables the sample size calculation
for the confirmatory phase III clinical trials. Moreover, TRL-7 must generate dose-ranging
data. Some regulatory agencies demand dose-ranging studies if the scientific literature
does not contain valid data [
16
]. The FDA demands the dose selection rationale to be based
on experimental human PK data, regardless of previous marketing experience [62].
At TRL-8, phase III clinical trials confirm efficacy and safety in broader patient pop-
ulations. These trials are designed to yield statistically significant results for predefined
variables through predetermined statistical methods (estimands) [
130
]. They also expand
the safety evaluations to larger cohorts to uncover rare adverse events [
16
,
113
]. Achiev-
ing marketing authorization from at least one regulatory agency signifies the completion
of TRL-8 criteria. Therefore, it is recommended for sponsors to engage with regulatory
agencies before initiating any phase III trial [
62
]. At TRL-9, the sponsor may conduct
phase IV clinical trials to further elucidate the HMP’s therapeutic profile or to assess
new formulations.
Special Issues on Clinical Trials of Herbal Medicinal Products
Although considered the gold standard for randomized-controlled clinical trials, the
use of a placebo is especially challenging for HMP development [
131
]. The possibilities
for placebos include excipients and other pharmacologically inert ingredients. However,
HMPs usually have typical organoleptic properties such as taste, odor, and appearance that
might be difficult to mask [
132
]. A possibility is using other botanical materials with no
pharmacological activity. However, concerns about an unsuspected activity might always
arise. Another possibility is a double-dummy design that randomizes participants to take
either the active treatment “1” and the placebo of the treatment “2”, or the placebo of the
treatment “1” and the active treatment “2” [
133
]. The FDA understands that it might be
difficult to select an ideal placebo for HMPs and encourages the sponsor to consult with
the agency beforehand [62].
Another challenge is the adjuvant treatment of serious conditions to which the FDA
recommends the “add-on to standard care versus standard care” [
62
]. In Brazil, according
to the Brazilian National Health Council, the placebo may be used only in cases where
no recognized helpful treatment is available. Also, for these cases, the add-on design
is recommended.
To minimize different understandings, the WHO published the guideline Operational
guidance: Information needed to support clinical trials of herbal products”. This should be
reviewed before designing the trials alongside the national regulations of the country
where the HMP is intended to be licensed [16].
Ethnopharmacological Information, Traditional Use
The research on NPs has historically been guided by ethnopharmacology. The level
of information spans from an anecdotal mention of use to long-term, high-quality phar-
macovigilance documentation with detailed descriptions of the HP, therapeutic effects,
recommended doses, and adverse events. Whenever available, no human data shall be ne-
glected. The FDA, ANVISA, and EMA demand the submission of all information available
on prior human exposure [62].
Ethnopharmacology can be a development starter or accelerator. In some cases, it
starts a research program to confirm and clarify the pharmacological potential. In other
cases, it justifies skipping some non-clinical assays and clinical trials after scaling-up issues
have been addressed. If consistent information on HP use by humans is available, it might
even be unethical to submit animals or humans to IND-directed studies [112,113].
However, handling ethnopharmacological information holds many challenges. The
first is species identification. Many historical records only mention common names, which
can be attributed to multiple plants across different botanical families, complicating ac-
Pharmaceuticals 2024,17, 703 14 of 23
curate identification. The next challenge is the HP description, including the part of the
plant used, extractive methods, route of administration, and doses, among others. Once
these challenges are overcome, the value of the ethnopharmacological source is given by
the richness and reliability of the information on human effects. Traditional communities
may employ terminology vastly different from that of Western medicine, potentially com-
plicating the interpretation of therapeutic effects. However, if clinical manifestations are
documented, it may be feasible to translate these into terms recognizable within Western
medical frameworks [
134
]. This can be the case for more consolidated and documented al-
ternative medical systems such as Ayurveda, Traditional Chinese Medicine, Unani, Siddha,
and other herbal medicines [113].
Moreover, according to the FDA [
62
], “when the rationale for developing certain
botanical drug products is based on prior clinical experience in alternative medical systems
e.g., Ayurveda,
Traditional Chinese Medicine, Unani, Siddha, and other herbal medicine
and pharmacognosy textbooks), the sponsor may propose to incorporate traditional prac-
tices into their clinical protocols. For example, patients may be selected or grouped based
on alternative medical practice and treated with specific botanical regimens accordingly,
or the final dosage form may be prepared by individual patients according to traditional
Chinese or Indian methods”.
3. Materials and Methods
The TRLs for HMPs were based on the original NASA concepts from aeronautical
development [
33
,
34
,
48
52
,
55
,
135
]. A search was conducted on the Web of Science, PubMed,
Embase, and Scopus databases using MeSH descriptors and keywords related to technology
readiness levels (TRLs) in pharmaceutical development, identifying
360 references.
After
removing duplicates, 262 articles were analyzed, and 12 were selected for further consid-
eration based on the specific mention of TRL in any biological
field [40,136146].
None
specifically addressed TRLs in pharmaceutical development, including small molecules,
biotechnological products, and herbal medicinal products (HMPs) for human use. The
closest were in chemistry [
40
], drugs and vaccines for animal health [
145
], and the bioin-
dustry [
146
]. Some R&D agencies’ websites have adapted technology readiness level (TRL)
concepts to pharmaceutical development, serving as both starting points and founda-
tions upon which we built our concepts for HMPs [
35
38
]. Drawing from these sources
and our professional experience, we applied technology readiness level (TRL) concepts
to herbal medicinal products (HMPs), creating a list of milestones to be reached at each
level. These milestones were categorized into six domains and aligned with relevant
guidelines from the ANVISA, EMA, FDA, the International Council for Harmonisation of
Technical Requirements for Pharmaceuticals for Human Use (ICH), the Organisation for
Economic Co-operation and Development (OECD), and the World Health Organization
(WHO). Considering the extensive European expertise in HMP regulation, we embraced
the terminology established by the EMA. The correspondence of EMA terminology to that
of the FDA and ANVISA is in Table 3.
Table 3. Terminology correspondence between EMA, FDA, and ANVISA.
EMA Term [31] FDA Term [48] ANVISA Term [63] EMA Definition
Herbal
substance
Botanical drug
substance Droga vegetal
Mainly whole, fragmented, or cut plants, plant parts,
algae, fungi, and lichen in an unprocessed, usually
dried form, but sometimes fresh. Certain exudates
that have not been subjected to a specific treatment are
also considered to be herbal substances. Herbal
substances are precisely defined by the plant part used
and the botanical name according to the binomial
system (genus, species, variety, and author) [114].
Pharmaceuticals 2024,17, 703 15 of 23
Table 3. Cont.
EMA Term [31] FDA Term [48] ANVISA Term [63] EMA Definition
Herbal preparation Botanical drug
preparation Derivado vegetal
Preparations are obtained by subjecting herbal
substances to treatments such as extraction,
distillation, expression, fractionation, purification,
concentration, or fermentation. These include
comminuted or powdered herbal substances, tinctures,
extracts, essential oils, expressed juices, and processed
exudates [115].
Herbal medicinal
product
Botanical drug
product Fitoterápico
Any medicinal product, exclusively containing as
active ingredients one or more herbal substances or
one or more herbal preparations, or one or more such
herbal substances in combination with one or more
such herbal preparations [116].
4. Concluding Remarks
This TRL-based roadmap is primarily designed to facilitate collaboration among
Brazilian scientists, manufacturers, and funding agencies in the development of innovative
herbal medicinal products (HMPs) targeting global markets. However, it could also be
beneficial for developers in other megadiverse countries looking to navigate similar paths.
Biodiversity along with ethnopharmacological information are still very underexploited
assets from megadiverse countries for drug discovery and development [
147
150
]. Most mar-
keted HMPs are based on European plants or traditional Chinese
medicine [151,152].
After the
Nagoya Protocol, some large pharmaceutical companies have scaled back their bioprospect-
ing activities [
1
,
22
,
153
]. This presents a potential window of opportunity for research
institutions in megadiverse countries.
The challenges are many. The pharma R&D processes are not mature in tropical
countries’ manufacturers. HMPs lack regulatory harmonization, and some legal uncertain-
ties from the Nagoya protocol still remain. Also, the HMPs’ R&D is more complex than
synthetic small molecules because the HP composition is susceptible to variations in plant
strain, place and season of cultivation/collection, and extraction and storage conditions.
This roadmap proposal is an effort to overcome some of these challenges. It should be
viewed as a general guidance meant to be tailored to each unique development scenario,
acknowledging that every case is distinct and has its own set of specific characteristics.
As previously noted, the established TRL scales lack boundary indicators [
28
,
113
].
This was the pioneers’ proposal and the main reason for TRLs’ universality and application
to many diverse technologies. Nevertheless, this allows the classification of the same project
in different TRLs, depending on subjective judgments. To avoid this ambiguity, besides the
TRLs transposal, we created a list of indicators organized into six domains that progress
with the TRLs. The domains help project management and allow more precise indicators.
Due to the universality of the TRL concepts, the transposal consistently met the HMP’s
R&D milestones. Aeronautical and pharmaceutical developments share some similarities.
Both start with material properties, followed by a proposal of application. However,
one important difference was the requirement of experimental data at TRL-2 to design
the PoC. This was due to the larger dependence of pharmacology on experimentation.
TRL-3 was kept as the PoC proposed by Mankins. This crucial step concerns a low-cost
experiment (or group of experiments) that needs to be convincing enough to justify the
project’s progression.
TRL-4 is very important for the project’s success and its first major risk inflection
point. For HMPs, the “validation in laboratory environment” proposed by Mankins was
translated as the complex, long, expensive, and laborious bench-scale HP optimization.
In our opinion, the lack of innovation in HMPs is mostly due to insufficient optimization.
Several projects have progressed unoptimized to GLP testing or even to clinical trials
and went back to initial stages or were abandoned afterward, with inconsistent results.
Pharmaceuticals 2024,17, 703 16 of 23
Therefore, we equate TRL-4 with the first “valley of death” [
154
]. It is the gap between
the fundamental research and the regulatory-demanded non-clinical and clinical tests that
finalize the innovation process. This gap still endures, despite the availability of non-
refundable funding specifically directed to innovation, perhaps due to the unawareness of
its importance and complexity [155].
The killer experiment is a good managerial principle to avoid spending scarce re-
sources on wrong projects. It reduces technological risk and justifies the scaling-up invest-
ment. In case of failure, it might be necessary to abort or to improve the HP candidate,
meaning that fundamental science is lacking. Therefore, in some cases, it might drive the
interruption of the technological aspect to stimulate fundamental science. It should be
envisioned early on at TRL-2 and executed at TRL-3 to TRL-4. Every innovative project
achieving TRL-4 demands broader pharmacological knowledge. Hereon, the more the
project matures and the closer it gets to the market, the more information will be needed to
minimize the clinical trials’ risks. These studies are generally carried out in research centers
and shall not be confused with the technological project itself but establish a relationship of
reciprocal benefits. In this way, while a technological project progresses to higher TRLs,
phytochemical and pharmacological scientific studies can feed it with valuable information
and receive suggestions that can motivate new research lines.
TRL-5 has two important goals: scaling up and validating the HP. Considering this,
the “relevant environment” proposed by Mankins was translated as the GLP condition. It
is also time to improve the manufacturing standards to face the clinical trials’ demands.
TRL-6 has another remarkable similarity between aeronautic and pharmacological
development—the first flight and the first human test demand substantial preceding
validations. Additionally, the object of development is called by Mankins: “component” at
TRL-5, “system/subsystem” at TRL-6, “prototype” at TRL-7, and “actual system” at TRL-8.
This bears close similarity with the HMP. At TRL-5, the “component” is the HP and, at
TRL-6 and TRL-7, the prototype and system/subsystem correspond to the evolving HMP
formulation to be administered to humans. At TRL-8, the “actual system” corresponds to
the HMP that goes to the market. In either case, models could be considered one-order-of-
magnitude approximations for performance [55].
Mankins also stated that “not all technologies undergo TRLs 6 and 7”, but, by defini-
tion, all go through TRL-8, which completes the “system development” [
52
]. For HMPs,
those projects with previous human data can skip at least TRL-6, and in the case of previ-
ously established doses according to traditional use, TRL-7 could also be skipped, but, by
definition, all successful projects must have approval by at least one regulatory authority,
which characterizes the TRL-8 completion.
TRL-9 holds an important difference between HMPs and astronautic development.
Except for new formulations demanding new clinical trials, no changes to the HMP or
manufacturing chain are allowed.
As a final remark, the TRLs do not provide insight into the uncertainty in pursuing
further maturation levels. Therefore, they are not substitutes for individualized expert
assessments of each project. Instead, they provide tools to guide the expert assessment. For
risk assessment, an additional metric is needed: the R&D degree of difficulty as described
elsewhere [34,52].
Future Implications
This TRL-based roadmap was created to enhance HMP development in megadiverse
countries with scientific rigor and regulatory compliance. It facilitates streamlined manage-
ment and resource allocation, fostering trust among scientists, manufacturers, and funding
agencies in the planning and execution of HMP development projects. We anticipate that it
will promote sustainable practices and catalyze therapeutic breakthroughs from underuti-
lized plant species, thereby increasing the introduction of innovative HMPs. The future
application of this roadmap in concrete cases will reveal areas for potential improvements.
Pharmaceuticals 2024,17, 703 17 of 23
Author Contributions: Conceptualization, E.P., C.D.R., C.M.S., S.A.C.P., B.S.B., A.C.B.C., and M.D.B.;
investigation, E.P., C.D.R., C.M.S., S.A.C.P., P.J.C.B., B.S.B., and M.D.B.; methodology, E.P., C.D.R.,
C.M.S., S.A.C.P., and M.D.B.; validation, E.P., C.D.R., C.M.S., A.C.B.C., and M.D.B.; writing—original
draft, E.P.; writing—review and editing, E.P., C.D.R., C.M.S., S.A.C.P., P.J.C.B., B.S.B., A.C.B.C., and
M.D.B. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: The raw data supporting the conclusions of this article will be made
available by the authors on request.
Acknowledgments: The authors wish to thank Henry Suzuki (Axonal) and Ana Claudia Dias
(ABIFINA) for their important contributions to the intellectual property of herbal medicinal products,
Rodrigo Rocha Secioso de Sá(FINEP) for contributions regarding the funding of research and
development projects, Carlos Eduardo Vitor for information about final product development, and
Emerson Queiroz (UniGe) for important contributions about the international herbal medicinal
products R&D scenario.
Conflicts of Interest: The authors declare that the research was conducted in the absence of any
commercial or financial relationships that could be construed as potential conflicts of interest.
References
1.
Li, J.W.; Vederas, J.C. Drug discovery and natural products: End of an era or an endless frontier? Science 2009,325, 161–165.
[CrossRef] [PubMed]
2.
Firn, R.D.; Jones, C.G. Natural products—A simple model to explain chemical diversity. Nat. Prod. Rep. 2003,20, 382–391.
[CrossRef] [PubMed]
3.
Davison, E.K.; Brimble, M.A. Natural product derived privileged scaffolds in drug discovery. Curr. Opin. Chem. Biol. 2019,52, 1–8.
[CrossRef] [PubMed]
4.
Larsson, J.; Gottfries, J.; Muresan, S.; Backlund, A. ChemGPS-NP: Tuned for navigation in biologically relevant chemical space.
J. Nat. Prod. 2007,70, 789–794. [CrossRef] [PubMed]
5.
Bruder, M.; Polo, G.; Trivella, D.B.B. Natural allosteric modulators and their biological targets: Molecular signatures and
mechanisms. Nat. Prod. Rep. 2020,37, 488–514. [CrossRef] [PubMed]
6.
Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019.
J. Nat. Prod. 2020,83, 770–803. [CrossRef] [PubMed]
7. Drews, J. Drug discovery: A historical perspective. Science 2000,287, 1960–1964. [CrossRef] [PubMed]
8.
Efferth, T.; Koch, E. Complex interactions between phytochemicals. The multi-target therapeutic concept of phytotherapy. Curr.
Drug Targets 2011,12, 122–132.
9.
Fibigr, J.; Satinsky, D.; Solich, P. Current trends in the analysis and quality control of food supplements based on plant extracts.
Anal. Chim. Acta 2018,1036, 1–15. [CrossRef] [PubMed]
10.
Koeberle, A.; Werz, O. Multi-target approach for natural products in inflammation. Drug Discov. Today 2014,19, 1871–1882.
[CrossRef]
11.
Stasilowicz, A.; Tomala, A.; Podolak, I.; Cielecka-Piontek, J. Cannabis sativa L. as a Natural Drug Meeting the Criteria of a
Multitarget Approach to Treatment. Int. J. Mol. Sci. 2021,22, 778. [CrossRef]
12.
Stockfleth, E.; Meyer, T. The use of sinecatechins (polyphenon E) ointment for treatment of external genital warts. Expert Opin.
Biol. Ther. 2012,12, 783–793. [CrossRef] [PubMed]
13.
Noor, F.; Tahir Ul Qamar, M.; Ashfaq, U.A.; Albutti, A.; Alwashmi, A.S.S.; Aljasir, M.A. Network Pharmacology Approach for
Medicinal Plants: Review and Assessment. Pharmaceuticals 2022,15, 572. [CrossRef] [PubMed]
14.
Caesar, L.K.; Cech, N.B. Synergy and antagonism in natural product extracts: When 1 + 1 does not equal 2. Nat. Prod. Rep. 2019,
36, 869–888. [CrossRef] [PubMed]
15.
WHO. WHO Traditional Medicine Strategy: 2014–2023; Institutional repository for information sharing; World Health Organization:
Geneva, Switzerland, 2013.
16.
WHO. Operational Guidance: Information Needed to Support Clinical Trials of Herbal Products; Institutional repository for information
sharing; World Health Organization: Geneva, Switzerland, 2005.
17.
Liang, Z.; Hu, H.; Li, J.; Yao, D.; Wang, Y.; Ung, C.O.L. Advancing the Regulation of Traditional and Complementary Medicine
Products: A Comparison of Five Regulatory Systems on Traditional Medicines with a Long History of Use. Evid. Based Complement.
Altern. Med. 2021,2021, 5833945. [CrossRef] [PubMed]
18.
Jadhav, C.A.; Vikhe, D.N.; Jadhav, R.S. Global and domestic market of herbal medicines: A review. Res. J. Sci. Technol. 2020,12,
327–330. [CrossRef]
Pharmaceuticals 2024,17, 703 18 of 23
19.
Grabher, C. A Governança e a Sustentabilidade Do Extrativismo Do Jaborandi Na Amazônia e Transição Para o Cerrado e a
Caatinga. In Faculdade de Ciências Econômicas. Programa de Pós-Graduação em Desenvolvimento Rural; Universidade Federal do Rio
Grande do Sul: Porto Alegre, Brazil, 2015.
20. WFO. World Flora Online. 2024. Available online: https://www.worldfloraonline.org/ (accessed on 4 January 2024).
21.
Theodoridis, S.; Drakou, E.G.; Hickler, T.; Thines, M.; Nogues-Bravo, D. Evaluating natural medicinal resources and their
exposure to global change. Lancet Planet. Health 2023,7, e155–e163. [CrossRef] [PubMed]
22.
Atanasov, A.G.; Waltenberger, B.; Pferschy-Wenzig, E.M.; Linder, T.; Wawrosch, C.; Uhrin, P.; Temml, V.; Wang, L.; Schwaiger, S.;
Heiss, E.H.; et al. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnol. Adv.
2015,33, 1582–1614. [CrossRef] [PubMed]
23.
Seile, B.P.; Bareetseng, S.; Koitsiwe, M.T.; Aremu, A.O. Indigenous Knowledge on the Uses, Sustainability and Conservation of
African Ginger (Siphonochilus aethiopicus) among Two Communities in Mpumalanga Province, South Africa. Diversity 2022,14,
192. [CrossRef]
24.
Pasdaran, A.; Naychov, Z.; Batovska, D.; Kerr, P.; Favre, A.; Dimitrov, V.; Aneva, I.; Hamedi, A.; Kozuharova, E. Some European
Gentiana Species Are Used Traditionally to Cure Wounds: Bioactivity and Conservation Issues. Diversity 2023,15, 467. [CrossRef]
25.
Balkrishna, A.; Singh, R.; Gohel, V.; Arora, S.; Dev, R.; Bhattacharya, K.; Varshney, A. Enteric-Coated Cologrit Tablet Exhibit
Robust Anti-Inflammatory Response in Ulcerative Colitis-like In-Vitro Models by Attuning NFkappaB-Centric Signaling Axis.
Pharmaceuticals 2022,16, 63. [CrossRef]
26.
Pan American Health Organization. Country Profiles. 2024. Available online: https://tcim-globalsummit.bvsalud.org/
regulations-and-policies/ (accessed on 4 January 2024).
27.
Geck, M.S.; Cristians, S.; Berger-Gonzalez, M.; Casu, L.; Heinrich, M.; Leonti, M. Traditional Herbal Medicine in Mesoamerica:
Toward Its Evidence Base for Improving Universal Health Coverage. Front. Pharmacol. 2020,11, 1160. [CrossRef]
28.
Heinrich, M.; Scotti, F.; Andrade-Cetto, A.; Berger-Gonzalez, M.; Echeverria, J.; Friso, F.; Garcia-Cardona, F.; Hesketh, A.; Hitziger,
M.; Maake, C.; et al. Access and Benefit Sharing Under the Nagoya Protocol-Quo Vadis? Six Latin American Case Studies
Assessing Opportunities and Risk. Front. Pharmacol. 2020,11, 765. [CrossRef] [PubMed]
29.
Convention on Biological Diversity, United Nations. Nagoya Protocol on Access to Genetic Resources and the Fair and Equitable Sharing
of Benefits Arising from Their Utilization to the Convention on Biological Diversity; Convention on Biological Diversity, United Nations:
Rio de Janeiro, Brazil; New York, NY, YSA, 2011.
30.
Castro Braga, F. Brazilian traditional medicine: Historical basis, features and potentialities for pharmaceutical development.
J. Tradit. Chin. Med. Sci. 2021,8, S44–S50. [CrossRef]
31.
Presidência da República. Lei No 13.123, de 20 de Maio de 2015; Diário Oficial; Republica Federativa do Brasil IN; Governo do
Brasil: Brasília, Brazil, 2015.
32.
Knoess, W.; Wiesner, J. The Globalization of Traditional Medicines: Perspectives Related to the European Union Regulatory
Environment. Engineering 2019,5, 22–31. [CrossRef]
33. Mankins, J.C. Technology Readiness Level: A White Paper; ResearchGate: Berlin, Germany, 1995.
34. Héder, M. From NASA to EU: The evolution of the TRL scale in Public Sector Innovation. Innov. J. 2017,22, 1–23.
35.
BIRAC. 1. Drugs (Including Drug Delivery). BIRAC-TRLs: Technology Readiness Levels by BIRAC Across Areas under
Biotechnology 2018. Available online: https://www.birac.nic.in/webcontent/birac_trl_doc1_drug_12_09_2018.pdf (accessed on
16 May 2024).
36.
IA. Description of Technology Readiness Levels (TRL). tia.org.za. 2019. Available online: https://www.tia.org.za/core/uploads/
2019/12/TRL-1.pdf (accessed on 17 November 2022).
37.
Notander, G. Technology Readiness Levels—TRL—NASA’s Contribution to Horizon 2020. 2020. Available online: https:
//www.wmahsn.org/storage/resources/documents/EIT_Health_KIC_A_guide_to_TRL-EIT_health.pdf (accessed on 5 July
2023).
38.
MedicalCountermeasures.gov. Integrated Technology Readiness Levels (TRLS) for Medical Countermeasure Products (Drugs and
Biologics); MedicalCountermeasures.gov: Bethesda, MD, USA, 2024.
39.
European Commission. Horizon 2020 Work Programme 2018–2020; General Annexes; European Commission: Brussels, Belgium,
2019.
40.
Buchner, G.A.; Stepputat, K.J.; Zimmermann, A.W.; Schomäcker, R. Specifying Technology Readiness Levels for the Chemical
Industry. Ind. Eng. Chem. Res. 2019,58, 6957–6969. [CrossRef]
41.
European Medicines Agency (Ed.) Guideline on Declaration of Herbal Substances and Herbal Preparations in Herbal Medicinal
Products/Traditional Herbal Medicinal Products; EMA: Amsterdam, The Netherlands, 2010.
42.
EMA. European Union Monographs and List Entries. Human Regulatory Herbal Products 2022; The Committee on Herbal
Medicinal Products (HMPC) Compiles and Assesses Scientific Data on Herbal Substances, Preparations and Combinations
with a Focus on Safety and Efficacy. This Work Supports the Harmonisation of the European Market: National Competent
Authorities Are Able to Refer to One Unique Set of Information on a Herbal Substance or Preparation When Evaluating
Marketing Applications. Available online: https://www.ema.europa.eu/en/human-regulatory/herbal-products/european-
union-monographs-list-entries (accessed on 4 January 2023).
43. European Medicines Agency (Ed.) Herbal Medicinal Products; EMA: Amsterdam, The Netherlands, 2024.
Pharmaceuticals 2024,17, 703 19 of 23
44.
European Commission. A Guideline on Summary of Product Characteristics (SmPC) Revision 2; European Commission: Brussels,
Belgium, 2009.
45.
EMA. Guideline on Specifications: Test Procedures and Acceptance Criteria for Herbal Substances, Herbal Preparations and Herbal Medicinal
Products/Traditional Herbal Medicinal Products; EMA: Amsterdam, The Netherlands, 2022.
46.
EMA. Guideline on Quality of Herbal Medicinal Products/Traditional Herbal Medicinal Products; EMA: Amsterdam, The Netherlands,
2022.
47. WHO. WHO Guidelines on Good Herbal Processing Practices for Herbal Medicines; WHO: Geneva, Switzerland, 2018.
48.
Mankins, J.C. Research & Development Degree of Difficulty (R&D3)—A White Paper; Economics Web Institute: Washington, DC, USA,
1998.
49.
Mankins, J.C. Approaches to strategic research and technology (R&T) analysis and road mapping. Acta Astronaut. 2002,51, 3–21.
50. Mankins, J.C. Technology readiness assessments: A retrospective. Acta Astronaut. 2009,65, 1216–1223. [CrossRef]
51.
Banke, J. Technology Readiness Levels Demystified. Aeronautics. 2010. Available online: https://www.nasa.gov/topics/
aeronautics/features/trl_demystified.html (accessed on 20 July 2022).
52. Mankins, J.C. Technology readiness and risk assessments: A new approach. Acta Astronaut. 2009,65, 1208–1215. [CrossRef]
53.
Hao, Y.; Ge, H.; Sun, M.; Gao, Y. Selecting an Appropriate Animal Model of Depression. Int. J. Mol. Sci. 2019,20, 4827. [CrossRef]
54.
Lerman, L.O.; Kurtz, T.W.; Touyz, R.M.; Ellison, D.H.; Chade, A.R.; Crowley, S.D.; Mattson, D.L.; Mullins, J.J.; Osborn, J.; Eirin,
A.; et al. Animal Models of Hypertension: A Scientific Statement From the American Heart Association. Hypertension 2019,73,
e87–e120. [CrossRef] [PubMed]
55.
Moorhouse, D.J. Detailed Definitions and Guidance for Application of Technology Readiness Levels. J. Aircr. 2002,39, 190–192.
[CrossRef]
56.
Krizkovska, B.; Kumar, R.; Rehorova, K.; Sykora, D.; Dobiasova, S.; Kucerova, D.; Tan, M.C.; Linis, V.; Oyong, G.; Ruml, T.;
et al. Comparison of Chemical Composition and Biological Activities of Eight Selaginella Species. Pharmaceuticals 2020,14, 16.
[CrossRef] [PubMed]
57.
Brazbio. Partnerships for a Better Wolrd. 2022. Brazbio Was Founded in 2018 as a Joint Venture between Centroflora Group, a
Brazilian Company That Operates in the Production and Commercialization of Pharmaceutical Extracts from Natural Ingredients,
and Givaudan, a Swiss Company and Global Leader in the Production of Flavors and Fragrances. Available online: http:
//brazbio.com/en/our-company/ (accessed on 20 July 2022).
58.
Canter, P.H.; Thomas, H.; Ernst, E. Bringing medicinal plants into cultivation: Opportunities and challenges for biotechnology.
Trends Biotechnol. 2005,23, 180–185. [CrossRef] [PubMed]
59.
CORDIS. Replacement of Contentious Inputs in Organic Farming Systems. Horizon 2020. 2018. Available online: https:
//cordis.europa.eu/article/id/442664-new-agroecological-approaches-to-scale-up-organic-farming (accessed on 16 May 2024).
60.
WHO. Fifty-First Report of the WHO Expert Committee on Specifications for Pharmaceutical Preparations; WHO technical report series;
no. 1003; WHO Library Cataloguing-in-Publication Data: Geneva, Switzerland, 2017.
61.
WHO (Ed.) Guidelines on Good Agricultural and Collection Practices (GACP) for Medicinal Plants; in iris.com; WHO: Geneva,
Switzerland, 2003.
62.
FDA. Botanical Drug Development Guidance for Industry; U.S. Department of Health and Human Services Food and Drug Adminis-
tration Center for Drug Evaluation and Research (CDER), Ed.; FDA: Silver Spring, MD, USA, 2016.
63.
EMA. Guideline on Good Agricultural and Collection Practice (GACP) for Starting Materials of Herbal Origin; EMEA/HMPC/246816/2005;
HMPC, Ed.; European Medicines Agency/Herbal Medicinal Products Committee: Amsterdam, The Netherlands, 2006; pp. 1–11.
64. FDA. 21CFR312.23; FDA: Silver Spring, MD, USA, 2023.
65.
Maitra, U.; Stephen, C.; Ciesla, L.M. Drug discovery from natural products—Old problems and novel solutions for the treatment
of neurodegenerative diseases. J. Pharm. Biomed. Anal. 2022,210, 114553. [CrossRef] [PubMed]
66.
Jităreanu, A.; Trifan, A.; Vieriu, M.; Caba, I.-C.; Mâr
t
,
u, I.; Agoroaei, L. Current Trends in Toxicity Assessment of Herbal Medicines:
A Narrative Review. Processes 2022,11, 83. [CrossRef]
67.
Quan, N.V.; Dang Xuan, T.; Teschke, R. Potential Hepatotoxins Found in Herbal Medicinal Products: A Systematic Review. Int. J.
Mol. Sci. 2020,21, 5011. [CrossRef] [PubMed]
68.
Ramesh, P.; Palaniappan, A. Terminalia arjuna, a Cardioprotective Herbal Medicine-Relevancy in the Modern Era of Pharmaceuti-
cals and Green Nanomedicine—A Review. Pharmaceuticals 2023,16, 126. [CrossRef]
69.
Djehiche, C.; Benzidane, N.; Djeghim, H.; Tebboub, M.; Mokrani, E.H.; Mebrek, S.; Messaoudi, M.; Bensouici, C.; Alsalme, A.;
Cornu, D.; et al. Exploring the Therapeutic Potential of Ammodaucus leucotrichus Seed Extracts: A Multi-Faceted Analysis of
Phytochemical Composition, Anti-Inflammatory Efficacy, Predictive Anti-Arthritic Properties, and Molecular Docking Insights.
Pharmaceuticals 2024,17, 385. [CrossRef]
70. Madia, V.N.; De Angelis, M.; De Vita, D.; Messore, A.; De Leo, A.; Ialongo, D.; Tudino, V.; Saccoliti, F.; De Chiara, G.; Garzoli, S.;
et al. Investigation of Commiphora myrrha (Nees) Engl. Oil and Its Main Components for Antiviral Activity. Pharmaceuticals
2021,14, 243. [CrossRef]
71.
Gaurav, H.; Yadav, D.; Maurya, A.; Yadav, H.; Yadav, R.; Shukla, A.C.; Sharma, M.; Gupta, V.K.; Palazon, J. Biodiversity,
Biochemical Profiling, and Pharmaco-Commercial Applications of Withania somnifera: A Review. Molecules 2023,28, 1208.
[CrossRef] [PubMed]
72. WHO (Ed.) WHO Guidelines on Good Manufacturing Practices for (GMP) for Herbal Medicines; WHO: Geneva, Switzerland, 2007.
Pharmaceuticals 2024,17, 703 20 of 23
73.
FDA. Guidance for Industry CGMP for Phase 1 Investigational Drugs Administration; U.S. Department of Health and Human Services
Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research
(CBER) Office of Regulatory Affairs (ORA), Ed.; FDA: Silver Spring, MD, USA, 2008.
74.
Bodeker, G.; Ong, C.-K.; Grundy, C.; Burford, G.; Shein, K. WHO Global Atlas of Traditional, Complementary and Alternative Medicine;
WHO Centre for Health Development: Kobe, Japan, 2005.
75.
Council of Europe; European Directorate for the Quality of Medicines & HealthCare. European Pharmacopoeia (Ph. Eur.), 11th ed.;
Conuncil of Europe: Strasbourg, France, 2023.
76.
ANVISA. Farmacopéia Brasileira. In Volume 2 Plantas Medicinais; Agência Nacional de Vigilância Sanitária: Brasília, Brazil, 2022.
77.
Nicolai, M.; Mota, J.; Fernandes, A.S.; Pereira, F.; Pereira, P.; Reis, C.P.; Robles Velasco, M.V.; Baby, A.R.; Rosado, C.; Rijo, P.
Assessment of the Potential Skin Application of Plectranthus ecklonii Benth. Pharmaceuticals 2020,13, 120. [CrossRef] [PubMed]
78.
OECD. OECD Test No. 423: Acute Oral Toxicity—Acute Toxic Class Method; OECD Guidelines for the Testing of Chemicals, Section
4; OECDiLibrary: Paris, France, 2002. [CrossRef]
79. ANVISA. Estudos Não Clinicos Necessários ao Desenvolvimento de Medicamentos Fitoterápicos; ANVISA: Brasília, Brazil, 2019.
80. WHO (Ed.) Quality Control Methods for Herbal Materials; WHO: Geneva, Switzerland, 2011.
81. USP. Herbal Medicines Compendium; United States Pharmacopeial Convention, Ed.; USP: North Bethesda, MD, USA, 2024.
82.
AOACInternational. Official Methods of Analysis, 22nd ed.; AOACInternational: Rockville, MD, USA, 2023. Available online:
https://www.aoac.org/official-methods-of- analysis/ (accessed on 4 July 2023).
83.
Medicines & Healthcare Products Regulatory Agency (Ed.) The British Pharmacopoeia Quality Standards; MHRA: London, UK,
2024.
84.
Choudhary, N.; Sekhon, B.S. An overview of advances in the standardization of herbal drugs. J. Pharm. Educ. Res. 2011,2, 55–70.
85.
Fitzgerald, M.; Heinrich, M.; Booker, A. Medicinal Plant Analysis: A Historical and Regional Discussion of Emergent Complex
Techniques. Front. Pharmacol. 2019,10, 1480. [CrossRef]
86.
Muyumba, N.W.; Mutombo, S.C.; Sheridan, H.; Nachtergael, A.; Duez, P. Quality control of herbal drugs and preparations: The
methods of analysis, their relevance and applications. Talanta Open 2021,4, 100070. [CrossRef]
87.
Kim, M.K.; Park, S.C.; Park, G.; Choi, E.; Ji, Y.; Jang, Y.P. Analytical quality by design methodology for botanical raw material
analysis: A case study of flavonoids in Genkwa Flos. Sci. Rep. 2021,11, 11936. [CrossRef] [PubMed]
88.
Indrayanto, G. Recent Development of Quality Control Methods for Herbal Derived Drug Preparations. Nat. Prod. Commun.
2018,13, 1599–1606. [CrossRef]
89.
Huang, S.; Chen, J.; Li, W.; Song, S.; Li, X.; Yu, H.; Long, F.; Chen, R.; Bao, X.; Chan, K.; et al. Comparison of Volatile Compositions
among Four Related Ligusticum chuanxiong Herbs by HS-SPME-GC-MS. Processes 2023,11, 196. [CrossRef]
90.
ICH. M4: The Common Technical Document. 2003. Available online: https://www.ich.org/page/ctd (accessed on 16 May 2024).
91.
ANVISA. Guia Para Organização do Documento Técnico Comum (CTD) Para o Registro e Pós-Registro de Medicamentos; ANVISA:
Brasília, Brazil, 2019.
92.
EMA (Ed.) Guideline on the Use of the CTD Format in the Preparation of a Registration Application for Traditional Herbal Medicinal
Products; EMA: Amsterdam, The Netherlands, 2016.
93.
Agência Nacional de Vigilância Sanitária—Anvisa (Ed.) RDC 166/2017 Estabelece Critérios Para a Validaçãode Métodos Analíticos;
ANVISA: Brasília, Brazil, 2017.
94.
Sarma, N.; Upton, R.; Rose, U.; Guo, D.A.; Marles, R.; Khan, I.; Giancaspro, G. Pharmacopeial Standards for the Quality Control
of Botanical Dietary Supplements in the United States. J. Diet. Suppl. 2023,20, 485–504. [CrossRef]
95.
ANVISA. Resolução da Diretoria Colegiada—RDC N
26, in Dispõe Sobre o Registro de Medicamentos Fitoterápicos e o Registro e a
Notificação de Produtos Tradicionais Fitoterápicos; ANVISA: Brasília, Brazil, 2014.
96.
Borgonetti, V.; Les, F.; Lopez, V.; Galeotti, N. Attenuation of Anxiety-like Behavior by Helichrysum stoechas (L.) Moench Methanolic
Extract through Up-Regulation of ERK Signaling Pathways in Noradrenergic Neurons. Pharmaceuticals 2020,13, 472.
97.
Zurdo, J. Developability assessment as an early de-risking tool for biopharmaceutical development. Pharm. Bioprocess. 2013,1,
29–50. [CrossRef]
98.
Lin, A.; Giuliano, C.J.; Palladino, A.; John, K.M.; Abramowicz, C.; Yuan, M.L.; Sausville, E.L.; Lukow, D.A.; Liu, L.; Chait, A.R.;
et al. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci. Transl. Med. 2019,11,
eaaw8412I. [CrossRef]
99.
Tuohongerbieke, A.; Wang, H.; Wu, J.; Wang, Z.; Dong, T.; Huang, Y.; Zhu, D.; Sun, D.; Tsim, K.W.K. Xiao Cheng Qi Decoction, an
Ancient Chinese Herbal Mixture, Relieves Loperamide-Induced Slow-Transit Constipation in Mice: An Action Mediated by Gut
Microbiota. Pharmaceuticals 2024,17, 153. [CrossRef] [PubMed]
100.
Parish, T.
In vitro
drug discovery models for Mycobacterium tuberculosis relevant for host infection. Expert Opin. Drug Discov.
2020,15, 349–358. [CrossRef] [PubMed]
101.
Sinha, S.; Vohora, D. Drug Discovery and Development. In Pharmaceutical Medicine and Translational Clinical Research; Academic
Press: Cambridge, MA, USA, 2018; pp. 19–32.
102.
Franco, C.H.; Alcantara, L.M.; Chatelain, E.; Freitas-Junior, L.; Moraes, C.B. Drug Discovery for Chagas Disease: Impact of
Different Host Cell Lines on Assay Performance and Hit Compound Selection. Trop. Med. Infect. Dis. 2019,4, 82. [CrossRef]
[PubMed]
Pharmaceuticals 2024,17, 703 21 of 23
103.
Mokhtar, F.A.; Selim, N.M.; Elhawary, S.S.; Abd El Hadi, S.R.; Hetta, M.H.; Albalawi, M.A.; Shati, A.A.; Alfaifi, M.Y.; Elbehairi,
S.E.I.; Fahmy, L.I.; et al. Green Biosynthesis of Silver Nanoparticles Using Annona glabra and Annona squamosa Extracts with
Antimicrobial, Anticancer, Apoptosis Potentials, Assisted by In Silico Modeling, and Metabolic Profiling. Pharmaceuticals 2022,15,
1354. [CrossRef] [PubMed]
104.
Pereira, F.; Aires-de-Sousa, J. Computational Methodologies in the Exploration of Marine Natural Product Leads. Mar. Drugs
2018,16, 236. [CrossRef]
105.
Loiodice, S.; Nogueira da Costa, A.; Atienzar, F. Current trends in in silico,
in vitro
toxicology, and safety biomarkers in early
drug development. Drug Chem. Toxicol. 2019,42, 113–121. [CrossRef] [PubMed]
106.
Rim, K.T. In silico prediction of toxicity and its applications for chemicals at work. Toxicol. Environ. Health Sci. 2020,12, 191–202.
[CrossRef] [PubMed]
107.
Van Norman, G.A. Limitations of Animal Studies for Predicting Toxicity in Clinical Trials: Is it Time to Rethink Our Current
Approach? JACC Basic Transl. Sci. 2019,4, 845–854. [CrossRef]
108.
Robinson, N.B.; Krieger, K.; Khan, F.M.; Huffman, W.; Chang, M.; Naik, A.; Yongle, R.; Hameed, I.; Krieger, K.; Girardi, L.N.; et al.
The current state of animal models in research: A review. Int. J. Surg. 2019,72, 9–13. [CrossRef]
109.
Laman, J.D.; Kooistra, S.M.; Clausen, B.E. Reproducibility Issues: Avoiding Pitfalls in Animal Inflammation Models. Methods Mol.
Biol. 2017,1559, 1–17. [PubMed]
110.
Owens, P.K.; Raddad, E.; Miller, J.W.; Stille, J.R.; Olovich, K.G.; Smith, N.V.; Jones, R.S.; Scherer, J.C. A decade of innovation in
pharmaceutical R&D: The Chorus model. Nat. Rev. Drug Discov. 2015,14, 17–28. [PubMed]
111.
ICH. Guidance on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Phar-
maceuticals M3(R2) Current Step 4 Version. International Conference on Harmonization; ICH. 2009. Available online:
https://www.ema.europa.eu/en/documents/scientific-guideline/ich-guideline-m3r2-non-clinical-safety-studies-conduct-
human-clinical-trials-and-marketing-authorisation-pharmaceuticals-step-5_en.pdf (accessed on 16 May 2024).
112.
Directive 2004/24/EC of the European Parliament and of the Council. Off. J. Eur. Union 2004,L 136, 85–90. Available online:
https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2004:136:0085:0090:en:PDF (accessed on 16 May 2024).
113.
WHO. General Guidelines for Methodologies on Research and Evaluation of Traditional Medicine; institutional repository for information
sharing; WHO: Geneva, Switzerland, 2000.
114.
EMA. Guideline on Non-Clinical Documentation in Applications for Marketing Authorisation/Registration of Wellestablished and Traditional
Herbal Medicinal Products; EMEA/HMPC/32116/2005 Rev.1; Committee on Herbal Medicinal Products (HMPC): Amsterdam,
The Netherlands, 2018.
115.
da Rocha, C.F.; Flexa, C.N.N.; de Souza, G.C.; Pereira, A.C.M.; Carvalho, H.O.; do Nascimento, A.L.; de Jesus Vasconcelos, N.J.P.;
da Silva, H.R.; Carvalho, J.C.T. Acute and Reproductive Toxicity Evaluation of Ormona((R)) SI and Ormona((R)) RC-Two New
Nutraceuticals with Geranylgeraniol, Tocotrienols, Anthocyanins, and Isoflavones-In Adult Zebrafish. Pharmaceuticals 2022,15,
1434. [CrossRef]
116.
WHO (Ed.) Key Technical Issues of Herbal Medicines with Reference to Interaction with Other Medicines; WHO: Geneva, Switzerland,
2021.
117.
Mazzari, A.L.; Milton, F.; Frangos, S.; Carvalho, A.C.; Silveira, D.; de Assis Rocha Neves, F.; Prieto, J.M.
In vitro
Effects of Four
Native Brazilian Medicinal Plants in CYP3A4 mRNA Gene Expression, Glutathione Levels, and P-Glycoprotein Activity. Front.
Pharmacol. 2016,7, 265. [CrossRef] [PubMed]
118.
FDA. The Drug Development Process. 2018. Available online: https://www.fda.gov/patients/learn-about-drug-and-device-
approvals/drug-development-process (accessed on 20 July 2023).
119.
Steinmetz, K.L.; Spack, E.G. The basics of preclinical drug development for neurodegenerative disease indications. BMC Neurol.
2009,9(Suppl. 1), S2. [CrossRef] [PubMed]
120. USP. <1033> Biological Assay Validation; IPQ International Pharmaceutical Quality: Washington, DC, USA, 2022.
121.
USP. <1032> Design and Development of Biological Assays; IPQ International Pharmaceutical Quality: Washington, DC, USA, 2022.
[CrossRef]
122.
OECD. OECD Test Guideline 451: Carcinogenicity Studies; OECD Publishing: Paris, France, 2009. Available online: https:
//www.oecd.org/env/test-no-451-carcinogenicity-studies- 9789264071186-en.htm (accessed on 16 May 2024).
123.
Kovarich, S.; Cappelli, C.I. Use of In Silico Methods for Regulatory Toxicological Assessment of Pharmaceutical Impurities.
Methods Mol. Biol. 2022,2425, 537–560. [PubMed]
124.
Little, J.G.; Marsman, D.S.; Baker, T.R.; Mahony, C. In silico approach to safety of botanical dietary supplement ingredients
utilizing constituent-level characterization. Food Chem. Toxicol. 2017,107, 418–429. [CrossRef] [PubMed]
125.
Myatt, G.J.; Ahlberg, E.; Akahori, Y.; Allen, D.; Amberg, A.; Anger, L.T.; Aptula, A.; Auerbach, S.; Beilke, L.; Bellion, P.; et al. In
silico toxicology protocols. Regul. Toxicol. Pharmacol. 2018,96, 1–17. [CrossRef] [PubMed]
126.
Nair, A.; Morsy, M.A.; Jacob, S. Dose translation between laboratory animals and human in preclinical and clinical phases of drug
development. Drug Dev. Res. 2018,79, 373–382. [CrossRef]
127.
Peters, S.A.; Petersson, C.; Blaukat, A.; Halle, J.P.; Dolgos, H. Prediction of active human dose: Learnings from 20 years of Merck
KGaA experience, illustrated by case studies. Drug Discov. Today 2020,25, 909–919. [CrossRef]
128. ICH. ICH: E6 (R2): Guideline for Good Clinical Practice—Step 5; International Conference on Harmonization: London, UK, 2016.
Pharmaceuticals 2024,17, 703 22 of 23
129.
EMA (Ed.) Guideline on Strategies to Identify and Mitigate Risks for First-in-Human and Early Clinical Trials with Investigational
Medicinal Products; EMEA/CHMP/SWP/28367/07 Rev., 1; EMA: Amsterdam, The Netherlands, 2017.
130.
ICH. Addendum on Estimands and Sensitivity Analysis in Clinical Trials to the Guideline on Statistical Principles for Clinical
Trials E9(R1). ICH Expert Working Group, Ed.; 2019. Available online: https://database.ich.org/sites/default/files/E9-R1_Step4
_Guideline_2019_1203.pdf (accessed on 16 May 2024).
131.
Cañigueral, S.; Tschopp, R.; Ambrosetti, L.; Vignutelli, A.; Scaglione, F.; Petrini, O. The Development of Herbal Medicinal
Products. Pharm. Med. 2012,22, 107–118. [CrossRef]
132.
Zhang, X.; Tian, R.; Zhao, C.; Tang, X.; Lu, A.; Bian, Z. Placebo design in WHO-registered trials of Chinese herbal medicine need
improvements. BMC Complement. Altern. Med. 2019,19, 299. [CrossRef] [PubMed]
133.
Raus, K.; Pleschka, S.; Klein, P.; Schoop, R.; Fisher, P. Effect of an Echinacea-Based Hot Drink Versus Oseltamivir in Influenza
Treatment: A Randomized, Double-Blind, Double-Dummy, Multicenter, Noninferiority Clinical Trial. Curr. Ther. Res. Clin. Exp.
2015,77, 66–72. [CrossRef]
134.
Pagani, E.; Santos, J.F.L.; Rodrigues, E. Culture-Bound Syndromes of a Brazilian Amazon Riverine population: Tentative correspon-
dence between traditional and conventional medicine terms and possible ethnopharmacological implications.
J. Ethnopharmacol.
2017,203, 80–89. [CrossRef] [PubMed]
135.
Tomaschek, K.; Olechowski, A.; Eppinger, S.; Joglekar, N. A Survey of Technology Readiness Level Users. INCOSE Int. Symp.
2016,26, 2101–2117. [CrossRef]
136.
Sutherland, I.; Ignatova, S.; Hewitson, P.; Janaway, L.; Wood, P.; Edwards, N.; Harris, G.; Guzlek, H.; Keay, D.; Freebairn, K.; et al.
Scalable Technology for the Extraction of Pharmaceutics (STEP): The transition from academic knowhow to industrial reality.
J. Chromatogr. A 2011,1218, 6114–6121. [CrossRef] [PubMed]
137.
Wu, C.; Wang, B.; Zhang, C.; Wysk, R.A.; Chen, Y.W. Bioprinting: An assessment based on manufacturing readiness levels. Crit.
Rev. Biotechnol. 2017,37, 333–354. [CrossRef] [PubMed]
138.
Lakkireddy, H.R.; Bazile, D.V. Nano-carriers for drug routeing—Towards a new era. J. Drug Target. 2019,27, 525–541. [CrossRef]
[PubMed]
139.
Allwardt, V.; Ainscough, A.J.; Viswanathan, P.; Sherrod, S.D.; McLean, J.A.; Haddrick, M.; Pensabene, V. Translational Roadmap
for the Organs-on-a-Chip Industry toward Broad Adoption. Bioengineering 2020,7, 112. [CrossRef]
140.
de Araújo e Silva, R.; Santa Brígida, A.I.; de Freitas Rosa, M.; da Silva Neto, R.M.; Spinosa, W.A.; Benício de SáFilho, E.; Brito de
Figueirêdo, M.C. An approach for implementing ecodesign at early research stage: A case study of bacterial cellulose production.
J. Clean. Prod. 2020,269, 122245. [CrossRef]
141.
Daniotti, S.; Re, I. Marine Biotechnology: Challenges and Development Market Trends for the Enhancement of Biotic Resources in
Industrial Pharmaceutical and Food Applications. A Statistical Analysis of Scientific Literature and Business Models. Mar. Drugs
2021,19, 61. [CrossRef] [PubMed]
142.
Pfeifer, K.; Ergal, I.; Koller, M.; Basen, M.; Schuster, B.; Rittmann, S.K.R. Archaea Biotechnology. Biotechnol. Adv. 2021,47, 107668.
[CrossRef] [PubMed]
143.
Jenie, R.P.; Suryana, Y.; Pambudi, S.; Widayanti, T.; Irzaman, I.; Nurdin, N.M.; Dahrul, M.; Iskandar, J.; Kurniawan, A.; Siskandar,
R.; et al. General protocol for ethical conforming development for non-invasive blood biomarker measurement optical device.
AIP Conf. Proc. 2021,2320, 050023.
144.
Becker, C.R.; Majeed, A.; Crispin, A.; Knez, A.; Schoepf, U.J.; Boekstegers, P.; Steinbeck, G.; Reiser, M.F. CT measurement of
coronary calcium mass: Impact on global cardiac risk assessment. Eur. Radiol. 2005,15, 96–101. [CrossRef] [PubMed]
145.
Arnouts, S.; Brown, S.; de Arriba, M.L.; Donabedian, M.; Charlier, J. Technology Readiness Levels for vaccine and drug
development in animal health: From discovery to life cycle management. Front. Vet. Sci. 2022,9, 1016959. [CrossRef] [PubMed]
146.
Smanski, M.J.; Aristidou, A.; Carruth, R.; Erickson, J.; Gordon, M.; Kedia, S.B.; Lee, K.H.; Prather, D.; Schiel, J.E.; Schultheisz, H.;
et al. Bioindustrial manufacturing readiness levels (BioMRLs) as a shared framework for measuring and communicating the
maturity of bioproduct manufacturing processes. J. Ind. Microbiol. Biotechnol. 2022,49, kuac022. [CrossRef] [PubMed]
147.
Myers, N.; Mittermeier, R.A.; Mittermeier, C.G.; da Fonseca, G.A.; Kent, J. Biodiversity hotspots for conservation priorities. Nature
2000,403, 853–858. [CrossRef] [PubMed]
148.
Braga, F.C. Paving New Roads Towards Biodiversity-Based Drug Development in Brazil: Lessons from the Past and Future
Perspectives. Rev. Bras. Farmacogn. 2021,31, 505–518. [CrossRef]
149.
Yuan, H.; Ma, Q.; Ye, L.; Piao, G. The Traditional Medicine and Modern Medicine from Natural Products. Molecules 2016,21, 559.
[CrossRef]
150.
Kim, J.K.; Kim, K.H.; Shin, Y.C.; Jang, B.H.; Ko, S.G. Utilization of traditional medicine in primary health care in low- and
middle-income countries: A systematic review. Health Policy Plan. 2020,35, 1070–1083. [CrossRef]
151.
Bilia, A.R.; Costa, M.D.C. Medicinal plants and their preparations in the European market: Why has the harmonization failed?
The cases of St. John’s wort, valerian, ginkgo, ginseng, and green tea. Phytomedicine, 2021; 81, 153421.
152.
Wang, M.; Franz, G. The role of the European Pharmacopoeia (Ph Eur) in Quality Control of Traditional Chinese Herbal Medicine
in European Member States. World J. Tradit. Chin. Med. 2015,1, 5–15. [CrossRef]
153.
Franzen, S.R.; Chandler, C.; Lang, T. Health research capacity development in low and middle income countries: Reality or
rhetoric? A systematic meta-narrative review of the qualitative literature. BMJ Open 2017,7, e012332. [CrossRef] [PubMed]
Pharmaceuticals 2024,17, 703 23 of 23
154.
Calza, F.; Ferretti, M.; Panetti, E.; Parmentola, A. Moving drug discoveries beyond the valley of death: The role of innovation
ecosystems. Eur. J. Innov. Manag. 2020,24, 1184–1209. [CrossRef]
155.
Trivella, D.B.B.; Bruder, M.C.P.; Oliveira, F.C.B.; Porcaro, R.; Rustiguel, J.K.; Ribeiro, L.B.; Felicio, R.d.; Cunha, M.G.d.; Nascimento,
A.F.Z.; Zeri, A.C.M.; et al. Descoberta de fármacos a partir de produtos naturais e a abordagem Molecular Power House (MPH).
Rev. Fitos 2022,16 (Suppl. 2), 176–192. [CrossRef]
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.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Ammodaucus leucotrichus exhibits promising pharmacological activity, hinting at anti-inflammatory and anti-arthritic effects. This study investigated seed extracts from Ammodaucus leucotrichus using methanol and n-hexane, focusing on anti-inflammatory and anti-arthritic properties. The methanol extract outperformed the n-hexane extract and diclofenac, a reference anti-inflammatory drug, in trypsin inhibition (85% vs. 30% and 64.67% at 125 μg/mL). For trypsin inhibition, the IC50 values were 82.97 μg/mL (methanol), 202.70 μg/mL (n-hexane), and 97.04 μg/mL (diclofenac). Additionally, the n-hexane extract surpassed the methanol extract and diclofenac in BSA (bovine serum albumin) denaturation inhibition (90.4% vs. 22.0% and 51.4% at 62.5 μg/mL). The BSA denaturation IC50 values were 14.30 μg/mL (n-hexane), 5408 μg/mL (methanol), and 42.30 μg/mL (diclofenac). Gas chromatography–mass spectrometry (GC–MS) revealed 59 and 58 secondary metabolites in the methanol and n-hexane extracts, respectively. The higher therapeutic activity of the methanol extract was attributed to hydroxyacetic acid hydrazide, absent in the n-hexane extract. In silico docking studies identified 28 compounds with negative binding energies, indicating potential trypsin inhibition. The 2-hydroxyacetohydrazide displayed superior inhibitory effects compared to diclofenac. Further mechanistic studies are crucial to validate 2-hydroxyacetohydrazide as a potential drug candidate for rheumatoid arthritis treatment.
Article
Full-text available
Xiao Cheng Qi (XCQ) decoction, an ancient Chinese herbal mixture, has been used in treating slow-transit constipation (STC) for years. The underlying action mechanism in relieving the clinical symptoms is unclear. Several lines of evidence point to a strong link between constipation and gut microbiota. Short-chain fatty acids (SCFAs) and microbial metabolites have been shown to affect 5-HT synthesis by activating the GPR43 receptor localized on intestinal enterochromaffin cells, since 5-HT receptors are known to influence colonic peristalsis. The objective of this study was to evaluate the efficacy of XCQ in alleviating clinical symptoms in a mouse model of STC induced by loperamide. The application of loperamide leads to a decrease in intestinal transport and fecal water, which is used to establish the animal model of STC. In addition, the relationship between constipation and gut microbiota was determined. The herbal materials, composed of Rhei Radix et Rhizoma (Rhizomes of Rheum palmatum L., Polygonaceae) 55.2 g, Magnoliae Officinalis Cortex (Barks of Magnolia officinalis Rehd. et Wils, Magnoliaceae) 27.6 g, and Aurantii Fructus Immaturus (Fruitlet of Citrus aurantium L., Rutaceae) 36.0 g, were extracted with water to prepare the XCQ decoction. The constipated mice were induced with loperamide (10 mg/kg/day), and then treated with an oral dose of XCQ herbal extract (2.0, 4.0, and 8.0 g/kg/day) two times a day. Mosapride was administered as a positive drug. In loperamide-induced STC mice, the therapeutic parameters of XCQ-treated mice were determined, i.e., (i) symptoms of constipation, composition of gut microbiota, and amount of short-chain fatty acids in feces; (ii) plasma level of 5-HT; and (iii) expressions of the GPR43 and 5-HT4 receptor in colon. XCQ ameliorated the constipation symptoms of loperamide-induced STC mice. In gut microbiota, the treatment of XCQ in STC mice increased the relative abundances of Lactobacillus, Prevotellaceae_UCG_001, Prevotellaceae_NK3B31_group, Muribaculaceae, and Roseburia in feces and decreased the relative abundances of Desulfovibrio, Tuzzerella, and Lachnospiraceae_ NK4A136_group. The levels of SCFAs in stools from the STC group were significantly lower than those the control group, and were greatly elevated via treatment with XCQ. Compared with the STC group, XCQ increased the plasma level of 5-HT and the colonic expressions of the GPR43 and 5-HT4 receptor, significantly. The underlying mechanism of XCQ in anti-constipation could be related to the modulation of gut microbiota, the increase in SCFAs, the increase in plasma 5-HT, and the colonic expressions of the GPR43 and 5-HT4 receptor. Our results indicate that XCQ is a potent natural product that could be a therapeutic strategy for constipation.
Article
Full-text available
Wound care will always be among the main tasks in all surgical specialties. Several medicinal plants have proven efficacy to cure wounds. Ethnobotanical research and ethnopharmacological research have virtually endless potential to find new lead compounds. The aim of this research review is to assess the potential of some Gentiana species as sources of promising active compounds to support wound healing. Gentians are among the most popular medicinal plants used in many countries for a wide spectrum of health conditions. Traditionally, those used to cure wounds are Gentiana lutea, G. punctata, G. asclepiadea, G. cruciata, G. oliverii, G. septemphida, and G. gelida. Candidate compounds with skin regeneration and wound-healing potential isolated from gentians are isogentisin, isoorientin, mangiferin, lupeol, pinoresinol, syringaresinol, eustomoside, and sweroside. Based on the rich source of traditional knowledge on the properties of gentians to cure various skin and soft tissue complications; only very few modern pharmacological studies have been performed to test this potential. Our review demonstrates that this field deserves further investigation. Many gentians are declining in number and have high IUCN conservation status, and cultivation and micropropagation methods are the only solution for the development of new drugs based on gentian extracts.
Article
Full-text available
Withania somnifera L. Dunal (Ashwagandha), a key medicinal plant native to India, is used globally to manage various ailments. This review focuses on the traditional uses, botany, phytochemistry, and pharmacological advances of its plant-derived constituents. It has been reported that at least 62 crucial and 48 inferior primary and secondary metabolites are present in the W. somnifera leaves, and 29 among these found in its roots and leaves are chiefly steroidal compounds, steroidal lactones, alkaloids, amino acids, etc. In addition, the whole shrub parts possess various medicinal activities such as anti-leukotriene, antineoplastic, analgesic, anti-oxidant, immunostimulatory, and rejuvenating properties, mainly observed by in vitro demonstration. However, the course of its medical use remains unknown. This review provides a comprehensive understanding of W. somnifera, which will be useful for mechanism studies and potential medical applications of W. somnifera, as well as for the development of a rational quality control system for W. somnifera as a therapeutic material in the future.
Article
Full-text available
Herbal medicines were the main source of therapeutic agents in the ancestral era. Terminalia arjuna (TA) is one such medicinal plant widely known for its several medicinal properties, especially its cardiovascular properties. They have several phytochemicals, such as flavonoids, polyphenols, triterpenoids, tannins, glycosides, and several minerals, proteins, and others that are responsible for the above-mentioned medicinal properties. In this review, we have first elaborated on the various processes and their parameters for the efficient extraction of relevant phytochemicals from TA extracts. Secondly, the mechanisms behind the various medicinal properties of TA extracts are explained. We have also highlighted the role of TA extracts on the green synthesis of metallic nanoparticles, especially silver and gold nanoparticles, with an elucidation on the mechanisms behind the synthesis of nanoparticles. Finally, TA extracts-based polymeric formulations are discussed with limitations and future perspectives. We believe that this review could help researchers understand the importance of a well-known cardioprotective medicinal plant, TA, and its biomedical properties, as well as their role in green nanotechnology and various formulations explored for encapsulating them. This review will help researchers design better and greener nanomedicines as well as better formulations to improve the stability and bioavailability of TA extracts.
Article
Full-text available
Chuanxiong (CX, Ligusticum chuanxiong), Japanese Chuanxiong (JCX, Cnidium officinale), Fuxiong (FX, Ligusticum sinense ‘Fuxiong’), and Jinxiong (JX, Ligusticum sinense ‘Jinxiong’) are aromatic herbs used in China, Japan, and other regions. Their morphology and aromatic odor are similar, resulting in confused and mixed uses. This study compares the volatile compositions of these herbs for defining their medical uses. Headspace solid-phase microextraction–gas chromatography–triple quadrupole–mass spectrometry was employed to separate, identify, and quantify the compounds in the volatile gas of the four herbs. A total of 128 volatile compounds were identified and quantified in 23 these herbal samples. The sums of 106, 115, 116, and 120 compounds were detected in the volatile gas of CX, JCX, FX, and JX, with the mean contents of 4.80, 7.12, 7.67, and 12.0 μg/g, respectively. Types and contents of the main compounds were found to be different in the volatile gas of these herbs. The orthogonal partial least squares discriminant analysis and hierarchical clustering analysis showed the four herbs located in different confined areas or clusters. It is concluded that the volatile compositions in the four herbs are generally similar, but the contents of main volatile compounds are different. These herbs should be clearly differentiated in medical use.
Article
Full-text available
Ulcerative colitis (UC) is an inflammatory bowel disease that affects the patients’ colorectal area culminating in an inflamed ‘leaky gut.’ The majority of UC treatments only provide temporary respite leading to its relapse. Therefore, this study investigated the efficacy of the enteric-coated ‘Cologrit’ (EC) tablet in alleviating UC-like inflammation. Cologrit is formulated using polyherbal extracts that have anti-inflammatory qualities according to ancient Ayurveda scriptures. Phytochemical profiling revealed the presence of gallic acid, rutin, ellagic acid, and imperatorin in Cologrit formulation. Cologrit treatment decreased inflammation in LPS-induced transformed THP-1 macrophages, and TNF-α-stimulated human colorectal (HT-29) cells through the modulation of NFκB activity, IL-6 production, and NFκB, IL-1β, IL-8, and CXCL5 mRNA expression levels. Cologrit also lessened human monocytic (U937) cell adhesion to HT29 cells. Methacrylic acid-ethylacrylate copolymer-coating of the enteric Cologrit tablets (EC) supported their dissolution, and the release of phytochemicals in the small intestine pH 7.0 environment in a simulated gastrointestinal digestion model. Small intestine EC digestae effectively abridged dextran sodium sulfate (2.5% w/v)-induced cell viability loss and oxidative stress in human colon epithelial Caco-2 cells. In conclusion, the enteric-coated Cologrit tablets demonstrated good small intestine-specific phytochemical delivery capability, and decreased UC-like inflammation, and oxidative stress through the regulation of TNF-α/NFκB/IL6 signaling axis.
Article
Full-text available
Even in modern times, the popularity level of medicinal plants and herbal medicines in therapy is still high. The World Health Organization estimates that 80% of the population in developing countries uses these types of remedies. Even though herbal medicine products are usually perceived as low risk, their potential health risks should be carefully assessed. Several factors can cause the toxicity of herbal medicine products: plant components or metabolites with a toxic potential, adulteration, environmental pollutants (heavy metals, pesticides), or contamination of microorganisms (toxigenic fungi). Their correct evaluation is essential for the patient’s safety. The toxicity assessment of herbal medicine combines in vitro and in vivo methods, but in the past decades, several new techniques emerged besides conventional methods. The use of omics has become a valuable research tool for prediction and toxicity evaluation, while DNA sequencing can be used successfully to detect contaminants and adulteration. The use of invertebrate models (Danio renio or Galleria mellonella) became popular due to the ethical issues associated with vertebrate models. The aim of the present article is to provide an overview of the current trends and methods used to investigate the toxic potential of herbal medicinal products and the challenges in this research field.
Article
Full-text available
Public research and innovation initiatives in animal health aim to deliver key knowledge, services and products that improve the control of animal infectious diseases and animal welfare to deliver on global challenges including public health threats, environmental concerns and food security. The Technology Readiness Level (TRL) is a popular innovation policy instrument to monitor the maturity of upcoming new technologies in publicly funded research projects. However, while general definition of the 9 levels on the TRL-scale enable uniform discussions of technical maturity across different types of technology, these definitions are very generic which hampers concrete interpretation and application. Here, we aligned innovation pipeline stages as used in the animal health industry for the development of new vaccines or drugs with the TRL scale, resulting in TRL for animal health (TRLAH). This more bespoke scale can help to rationally allocate funding for animal health research from basic to applied research, map innovation processes, monitor progress and develop realistic progress expectations across the time span of a research and innovation project. The TRLAH thus become an interesting instrument to enhance the translation of public research results into industrial and societal innovation and foster public-private partnerships in animal health.
Article
Medicinal plants and their bioactive molecules are integral components of nature and have supported the health of human societies for millennia. However, the prevailing view of medicinal biodiversity solely as an ecosystem-decoupled natural resource of commercial value prevents people from fully benefiting from the capacity of nature to provide medicines and from assessing the vulnerability of this capacity to the global environmental crisis. Emerging scientific and technological developments and traditional knowledge allow for appreciating medicinal plant resources from a planetary health perspective. In this Personal View, we highlight and integrate current knowledge that includes medicinal, biodiversity, and environmental change research in a transdisciplinary framework to evaluate natural medicinal resources and their vulnerability in the anthropocene. With Europe as an application case, we propose proxy spatial indicators for establishing the capacity, potential societal benefits, and economic values of native medicinal plant resources and the exposure of these resources to global environmental change. The proposed framework and indicators aim to be a basis for transdisciplinary research on medicinal biodiversity and could guide decisions in addressing crucial multiple Sustainable Development Goals, from accessible global health care to natural habitat protection and restoration.