ArticlePDF AvailableLiterature Review

Nwaka, S. & Hudson, A. Innovative lead discovery strategies for tropical diseases. Nat. Rev. Drug Discov. 5, 941-955

Authors:
  • ANDI/UNOPS - African Network for Drugs and Diagnostics Innovation at UNOPS

Abstract and Figures

Lead discovery is currently a key bottleneck in the pipeline for much-needed novel drugs for tropical diseases such as malaria, tuberculosis, African sleeping sickness, leishmaniasis and Chagas disease. Here, we discuss the different approaches to lead discovery for tropical diseases and emphasize a coordination strategy that involves highly integrated partnerships and networks between scientists in academic institutions and industry in both wealthy industrialized countries and disease-endemic countries. This strategy offers the promise of reducing the inherently high attrition rate of the early stages of discovery research, thereby increasing the chances of success and enhancing cost-effectiveness.
An innovative lead discovery strategy for tropical diseases.This strategy involves networks and partnerships with industry and academic institutions worldwide to deliver specific drug discovery objectives. The portfolio of prioritized and validated molecular targets developed by the target portfolio network will be used in high-throughput screening (HTS) efforts at various institutions (academia and industry). Hits* emerging from the screening are assessed in whole parasites (in vitro) through the compound-evaluation network. In addition, compounds with an established biological/biochemical rationale or diverse structures, as well as natural products, are sourced and channelled into the compound-evaluation network for whole-parasite screening, with actives subsequently being tested in animal disease models. Through iterative medicinal chemistry and pharmacological profiling using the appropriate network, structure–activity relationships are developed and used to guide synthesis of analogues with enhanced activity. The resulting drug-like lead compounds will then be progressed into focused optimization programmes in collaboration with other partners. The integrated lead discovery strategy of the Special Programme for Research and Training in Tropical Diseases at the World Health Organization (WHO/TDR) involves experienced consultants or mentors who support and provide guidance on various aspects of the preclinical process and to the postdoctoral fellows from disease-endemic countries who are working and being trained on the programme. The involvement of institutions worldwide in the various network activities calls for increased management and fruitful capacity building, especially in the disease-endemic countries. Interactions between the different networks and quality control are managed by WHO/TDR. In this respect, a central database housed at WHO/TDR is a crucial resource for managing individual projects and processing data and compounds. *A 'hit' is compound with selective in vitro activity (usually IC50 <1M) against the target whole organism and/or protein; a 'lead' is a compound with druggable characteristics, that is efficacious in disease animal models with no overt toxicity; a 'drug candidate' is an optimized lead compound with in vitro and in vivo activity equivalent or better than drug standards, acceptable pharmacokinetic and toxicity profile, with a synthesis that is amenable to cost-effective scale-up. Activity criteria for 'hit' and 'lead' compounds are presented in Box 1.
… 
Content may be subject to copyright.
There is a continuing and compelling need for new and
improved treatments for developing-world diseases.
These include bacterial, protozoan and helminth infec-
tious diseases such as tuberculosis, malaria, African
sleeping sickness, leishmaniasis, Chagas disease,
onchocerciasis, lymphatic filariasis and schistosomiasis
1–3
.
A number of factors limit the utility of existing drugs
in resource-poor settings, such as high cost, poor com-
pliance, drug resistance, low efficacy and poor safety
2
.
Because the evolution of drug resistance is likely to com-
promise every drug in time, the demand for new thera-
pies is continuous. Accordingly, a vibrant drug discovery
pipeline is needed to help to ensure the availability of
new products that will reduce mortality and morbidity
resulting from these infections
4,5
.
Discovering lead compounds with the potential
to become usable drugs is a crucial step to ensuring a
sustainable global pipeline for innovative products
4,6
. In
recognition of this need a number of agencies, includ-
ing the Special Programme for Research and Training
in Tropical Diseases at the World Health Organization
(WHO/TDR), various international/national bodies
and philanthropic foundations, have been supporting
the discovery of such agents for tropical diseases (see
Further information for organizations likely to support
this type of research). Some of the fruits of these programmes
have already been taken forward by public–private
partnerships (PPPs)
5,7,8
.
Drug development PPPs that focus on product
identification for specific tropical diseases often require
quality lead compounds to feed into their preclinical pipe-
lines. Some of these organizations are making significant
progress in trying to bring products to the market
through enhanced development programmes, but place
less emphasis on the risky early phases of the discovery
process
2,9,10
. However, owing to the paucity of robust
lead series, these organizations are now trying to invest
more in the early stages of drug discovery. For example,
the Medicines for Malaria Venture (MMV) has recently
initiated a focused call for drug discovery projects to
boost its antimalarial pipeline. In addition, a number
of the diseases mentioned above (lymphatic filariasis,
onchocerciasis and schistosomiasis) lack dedicated
PPPs for innovative product discovery and development.
Recent reports have highlighted the gaps, needs and
opportunities for increased investment and activity in
translational research for new product leads
10–12
. It should
be noted that lead discovery tends not to receive much
funding from the normal scientific granting bodies,
and so there is less incentive for academia to work in
this area.
The majority of international R&D funding and aid
for infectious diseases affecting the developing world is
focused on the ‘big three’ healthcare problems — HIV,
tuberculosis and malaria
9,13
. This is understandable
given the high burden of these diseases. However, there
is a compelling need to invest in innovative strategies to
address the other largely neglected infectious diseases
prevalent in the developing world via enhanced transla-
tional research for new products, as well as capacity build-
ing and utilization in disease-endemic countries
11,14,15
.
We lack a robust pipeline of products in discovery and
development to deliver drugs that meet the desired target
product profiles
(TABLE 1) for these diseases
2,5,16,17
.
Special Programme for
Research and Training in
Tropical Diseases (TDR),
World Health Organization
Geneva, Switzerland.
Correspondence to S.N.
e-mail: nwakas@who.int
doi:10.1038/nrd2144
Published online
13 October 2006
Innovative lead discovery strategies
for tropical diseases
Solomon Nwaka and Alan Hudson
Abstract | Lead discovery is currently a key bottleneck in the pipeline for much-needed
novel drugs for tropical diseases such as malaria, tuberculosis, African sleeping sickness,
leishmaniasis and Chagas disease. Here, we discuss the different approaches to lead
discovery for tropical diseases and emphasize a coordination strategy that involves highly
integrated partnerships and networks between scientists in academic institutions and
industry in both wealthy industrialized countries and disease-endemic countries.
This strategy offers the promise of reducing the inherently high attrition rate of the early
stages of discovery research, thereby increasing the chances of success and enhancing
cost-effectiveness.
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
941
So, there is an urgent requirement for a coordinated
approach involving multi-disciplinary networks of
investigators, as well as partnerships between industry
and the public sector in both developed and developing
countries. One such approach that considers networks,
partnerships and capacity building in an integrated lead
discovery process is illustrated in
FIG. 1. The present paper
discusses strategies to meet the need for lead compounds
for further development for tropical diseases. Specific
examples are drawn from the work of the WHO/TDR
covering a broad range of tropical diseases and from the
approaches taken by other agencies in these areas.
Strategies for drug discovery
The discovery of novel therapeutics for tropical diseases
has largely relied on three strategies: label extension,
piggy-back discovery and de novo drug discovery.
Label extension. Until recently, the primary strategy
for drug discovery for tropical diseases was based
on extending the indications of existing treatments
for other human and animal ailments to tropical
diseases
5,18,19
. This fast-track approach has been suc-
cessful and has resulted in some of the most important
antiparasitic drugs in use today, such as ivermectin
for filariasis/onchcocerciasis, and praziquantel for
schistosomiasis
20–23
(TABLE 2). It continues to have a
major role in the global drug discovery and develop-
ment strategy for tropical diseases. For example, the
veterinary anthelminthic product moxidectin, an
analogue of ivermectin
24
, is being taken into Phase II
clinical trials for the treatment of lymphatic filaria-
sis and onchocerciasis. The main attractions of this
approach are the reductions in cost and time to market
that can be achieved. In addition, over the past three
decades there have been few specific drug discovery
programmes supported by the pharmaceutical industry
that target tropical diseases. However, concern related
to over-reliance on label extensions has arisen in recent
years: many companies have been reluctant to allow
their products to be developed for tropical diseases in
case their economic potential is blighted by unexpected
Table 1a | Limitations of available drugs for parasitic diseases and proposed target profile for new drugs
Some available drugs and their limitations* Proposed target profile
Malaria
Quinine (1930): compliance, safety, resistance
Chloroquine (1945): resistance
Primaquine (1948): safety
Sulfadoxine/pyrimethamine (1961):
resistance
Mefloquine (1984): resistance, safety
Artemisinins (1994): compliance, cost, Good
Manufacturing Practice
Atovaquone/proguanil (1999): cost
For uncomplicated falciparum malaria
Orally active
Low cost of goods (~US$1 per full course treatment)
Effective against drug-resistant parasites; low propensity to generate rapid resistance
Curative within 3 days
Fast acting
Potential for combination with other agents
Paediatric formulation
Stable under tropical conditions (shelf life of >2 years)
Further profiles
Intermittent treatment in pregnancy and early infancy
Plasmodium vivax malaria
Severe malaria
Prophylaxis
Single dose cure
Leishmaniasis
Pentamidine (1939): safety and efficacy/
resistance issues, injectable
Antimonials (1950): safety and efficacy/
resistance issues, injectable
Liposomal Amphotericin B (1990):
cost, injectable
Miltefosine (2002): contraindicated in
pregnancy
Active against all visceral and cutaneous leishmaniasis
Short course of treatment (≤14 days)
Single daily dose, but alternate days or weekly dosing acceptable
Injectable with reduced treatment time acceptable
Oral drug desired
Safer than available treatment
Safe in children and pregnancy desired
Cost less than current treatments (US$200–400)
Stable under standard tropical conditions (shelf life >2 years)
Further profiles
Topical application for cutaneous leishmaniasis desired
Potential for combination with existing agents
Human African trypanosomiasis
Suramin (1920): safety, not effective in late-
stage disease, injectable
Pentamidine (1939): safety and resistance
issues, injectable, not effective in late-
stage disease
Melarsoprol (1949): safety and resistance
issues, injectable
Eflornithine (1991): cost, injectable, only
effective in Trypanosoma gambiense
Active against both major species Trypanosoma rhodesiense and T. gambiense
Active against known resistance strains for example, melarsoprol failures
Treatment for early-stage diseases acceptable but efficacy against both early- and late-stage desired
Parenteral administration for late-stage disease
Oral formulation for early-stage disease desired
Cure in 14 days or less
Cost less than current treatment for early stage disease ($100–140)
Safe in pregnancy
Stable under tropical conditions (shelf life >2 years)
*The dates in parentheses are the approximate dates when the drugs were first used (information adapted and modified from REFS 2,5,22).
REVIEWS
942
|
NOVEMBER 2006
|
VOLUME 5 www.nature.com/reviews/drugdisc
toxicities in these patient classes
5
. This, and other factors,
has driven the quest for novel products for tropical
diseases using additional discovery strategies
5,25
.
‘Piggy-back’ discovery. The ‘piggy-back’ strategy is most
useful when a molecular target present in parasites is
being pursued for other (commercial) indications as it
facilitates the identification of chemical starting points
26
.
Specific examples of this approach include the antimalarial
screening of lead series of histone deacetylase inhibitors
27
that were originally developed for cancer chemotherapy,
and cysteine protease inhibitors that are being developed
for osteoporosis
18
. It should be noted that structure–
activity relationships emerging from the parasite assays
are unlikely to be the same as those observed for the
original indication. It is therefore likely that optimized
clinical candidates emerging from this strategy will be
disease-specific.
De novo drug discovery. This strategy focuses on the iden-
tification of new chemical entities, both synthetic com-
pounds and natural products, as novel antiparasitic drugs.
It is more long-term than the approach discussed above
and integrates discovery research based on target-based
high-throughput screening (HTS) and medium through-
put screening (MTS) in whole-parasite assays against
specific proteins and whole parasites.
Target-based HTS campaigns have been emphasized
in recent years as a way of harvesting the significant
investment made in parasite genomics programmes by
the international community
28–31
. However, difficul-
ties encountered in moving resultant hits through the
pipeline — for example, in demonstrating a correlation
between enzyme inhibition and activity against whole
parasites — has generated interest in developing HTS
techniques for whole-organism screening
25,32–35
. Recent
whole-cell-based HTS campaigns using compound
libraries containing registered drugs have yielded encour-
aging results
34,35
. In addition, chemoinformatic meth-
odologies linked to genomics, in silico screening
36–38
,
as well as the structural determination of proteins and
their co-crystallization with small molecules, are now
being applied for antibacterial and antiparasitic drug
discovery
39–42
.
MTS in whole-parasite assays using compounds cho-
sen on the basis of a biological, biochemical or structural
rationale remains the most pursued screening option
for parasitic diseases
5,25,43
. The main disadvantage of
Table 1b | Limitations of available drugs for parasitic diseases and proposed target profile for new drugs
Some available drugs and their limitations* Proposed target profile
Chagas disease (American trypanosomiasis)
Nifurtimox (1970): safety, long treatment
compliance, activity limited to acute stage of
disease
Benznidazole (1974): safety, activity limited to
acute stage of disease
Active against blood and tissue forms of disease
Active in chronic forms of the disease
Parental administration with reduced treatment time acceptable
Oral drug desired
Improved safety over current products (free of cardiac effects)
Paediatric formulation
Safe for use in children and pregnancy
Inexpensive
Stable under tropical conditions (shelf life >2 years)
Schistosomiasis
Oxamniquine (1967): only effective against
Schistosoma mansoni, multiple dosing, cost
Praziquantel (1975): does not kill young
worms and eggs; possible resistance reported
New chemical class; alternative to praziquantel is important in the context of resistance
development
New mechanism of action: drug active against mature and immature forms of parasites
including eggs
Active against all major types of schistosome infections
Safety equal or better than praziquantel
Oral use
Inexpensive
Short treatment courses (ideally single oral dose)
Safety profile compatible with use without diagnosis
Safe in children, pregnant women desired
Stable under tropical conditions (shelf life >2 years)
Lymphatic filariasis and onchocerciasis
Diethylcarbamazine (1949): safety, not a
macrofilaricide, not used in Onchocerca
volvulus endemic areas
Albendazole: only used in combination
therapy, little acute microfilaricidal effect
Ivermectin (1989): not a macrofilaricide,
regular administration needed to kill young
worms
New chemical class: alternative to ivermectin and albendazole, important in the context of
resistance development
Macrofilaricidal or permanent sterilization of adult worms (in addition to being microfilaricidal)
Slow action (avoid rapid death of worms to prevent side effects due to immune responses)
Oral use
Inexpensive
Safety equal or better than ivermectin or combinations for LF
Short treatment courses (ideally single oral dose)
Safety profile compatible with use without diagnosis
Safe in children, pregnant women
Stable under tropical conditions (shelf life >2 years)
*The dates in parentheses are the approximate dates when the drugs were first used (information adapted and modified from REFS 2,5,22).
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
943
M
e
n
t
o
r
s
h
i
p
I
n
d
u
s
t
r
y
In vitro/vivo
screening
network
Capacity
buiding/
fellowships
Quality
leads
Optimization
Interface with other players
Drug
candidates
Pharmacokinetics/
metabolism network
Medicinal
chemistry
network
Validated
drug targets
HTS
Target
Portfolio
Network
Compounds
(known rationale,
diverse, natural
products)
Hits
Hits
Leads
A
c
a
d
e
m
i
a
this approach is the low throughput of available assays
(especially those using filariae and schistosomes), and
the limited investment in the development of new robust
assays. A recurring difficulty for all screens (HTS or
MTS) for neglected diseases is the availability of high-
quality compound libraries. Efforts are now being made
to establish compound libraries and HTS screening
capacity at public institutions
(TABLE 3).
It should be mentioned that so far in the field of
anti-infective (including antiparasitic) drug discovery,
the target-based HTS approach has yielded few success
stories
6,44–46
. In part this reflects the high rate of attrition
in the process of progression from early-stage biochemi-
cal hits to robust lead compounds. Many compounds
active in protein-based assays are inactive in whole cells.
This can be due to failure to enter intact cells but can
also occur because the chosen molecular targets are not
in fact essential to the microbes. The latter issue sug-
gests that more work on target validation is needed to
increase confidence levels in the selection of protein
candidates for HTS campaigns. The initial challenge of
identifying molecular targets that are crucial to parasite
survival, coupled with the identification of whole-cell
active compounds, is formidable — and this challenge
is made harder by the need to achieve efficacy in small
animal disease models combining an appropriate level
of potency with suitable pharmacokinetics. With the
possible exception of the cysteine protease inhibitor
K777, which is in development for Chagas disease
47
, the
authors are not aware of any compound in late discovery
phase or development for a human protozoan or
helminth disease that has resulted from a target-based
HTS campaign. The strategy is still valid but needs to be
augmented by increased efforts to select and focus on
validated molecular targets and to improve the quality of
compound libraries selected for the initial screening exer-
cise. It should be seen as complementary to whole-cell
screening and not as a substitute for it.
Figure 1 | An innovative lead discovery strategy for tropical diseases. This strategy involves networks and
partnerships with industry and academic institutions worldwide to deliver specific drug discovery objectives.
The portfolio of prioritized and validated molecular targets developed by the target portfolio network will be used
in high-throughput screening (HTS) efforts at various institutions (academia and industry). Hits* emerging from the
screening are assessed in whole parasites (in vitro) through the compound-evaluation network. In addition, compounds
with an established biological/biochemical rationale or diverse structures, as well as natural products, are sourced and
channelled into the compound-evaluation network for whole-parasite screening, with actives subsequently being tested
in animal disease models. Through iterative medicinal chemistry and pharmacological profiling using the appropriate
network, structure–activity relationships are developed and used to guide synthesis of analogues with enhanced activity.
The resulting drug-like lead compounds will then be progressed into focused optimization programmes in collaboration
with other partners. The integrated lead discovery strategy of the Special Programme for Research and Training in Tropical
Diseases at the World Health Organization (WHO/TDR) involves experienced consultants or mentors who support and
provide guidance on various aspects of the preclinical process and to the postdoctoral fellows from disease-endemic
countries who are working and being trained on the programme. The involvement of institutions worldwide in the various
network activities calls for increased management and fruitful capacity building, especially in the disease-endemic
countries. Interactions between the different networks and quality control are managed by WHO/TDR. In this respect,
a central database housed at WHO/TDR is a crucial resource for managing individual projects and processing data and
compounds. *A ‘hit’ is compound with selective in vitro activity (usually IC
50
<1μM) against the target whole organism
and/or protein; a ‘lead’ is a compound with druggable characteristics, that is efficacious in disease animal models with no
overt toxicity; a ‘drug candidate’ is an optimized lead compound with in vitro and in vivo activity equivalent or better than
drug standards, acceptable pharmacokinetic and toxicity profile, with a synthesis that is amenable to cost-effective
scale-up. Activity criteria for ‘hit’ and ‘lead’ compounds are presented in
BOX 1.
REVIEWS
944
|
NOVEMBER 2006
|
VOLUME 5 www.nature.com/reviews/drugdisc
The tripartite strategy pursuing ‘label extension, ‘piggy-
back’ and ‘de novo drug discovery’ is integrated into the
network and partnership model described below.
Network/partnership models for lead discovery
In recent years, a number of networks, partnerships and
consortia have been established specifically to pursue
tropical disease research. Important examples include
the following:
A collaborative network of institutions supported
under the Grand Challenges in Global Health initia-
tive (funded by the Gates Foundation and Wellcome
Trust; see Further information) to address infectious
disease problems — for example, those with the goal
to discover drugs and delivery systems that limit drug
resistance for infectious diseases (see Further infor-
mation, Grand Challenges in Global Health initiative
— Limit drug resistance)
48
.
The various European Union drug discovery initiatives,
such as the FP6, support consortia (for example, the
Antimal Drug Discovery network for malaria, which
consists of scientists from over 20 institutions, including
academia and industry; see Further information, EU
Commission — Poverty-Related Diseases) as well
as the network focusing on new drugs for persistent
tuberculosis (see Further information, EU Commission
— New TB Drugs).
Several drug discovery projects present within the
portfolio of PPPs such as MMV, Drugs for Neglected
Diseases Inititative (DNDi) and Global Alliance for TB
Drug Development (GATB). For example, the anti -
malarial synthetic peroxide project is based on the network
paradigm
7
, as is the 8-aminoquinoline project at the
University of Mississippi, which is supported by both
MMV and DNDi for malaria and leishmaniasis.
A consortium supported by the Gates Foundation for
the discovery of new drugs for African sleeping sick-
ness is focused on the dicationic structure scaffold
49,50
.
The team consists of investigators from the University
of North Carolina, the Swiss Tropical Institute (STI),
Ohio State University, Kenya Trypanosomiasis Research
Institute, London School of Hygiene and Tropical
Medicine (LSHTM) with Immtech International as
the industrial partner.
Another fairly recent development is the emergence of
dedicated academic and public initiatives that focus on
various aspects of drug discovery for tropical and non-
tropical diseases
2,10,51–53
. Such initiatives are largely sup-
ported with external funding and aim to approach the
level of drug discovery resources and expertise present
in small-to-medium-size biopharmaceutical companies.
Efforts of these centres include HTS and MTS using
synthetic small-molecule and natural-product libraries
(TABLE 3). Examples include the University of Dundees
drug discovery initiative for trypanosomiasis and
leishmaniasis, funded by the Wellcome Trust and other
agencies, which encompasses HTS screening capability,
molecular and parasite biology, and medicinal chemistry
supported by ADME (absorption, distribution, metabo-
lism and excretion) assays. Another example is the
University of California San Francisco Sandler Center
and Tropical Diseases Research Unit (supported by the
Sandler Family Foundation and the National Institute
of Allergy and Infectious Diseases (NIAID)), which is
a consortium of laboratories dedicated to the discovery
and development of new drugs for tropical diseases with
core competences in genomics and proteomics, struc-
tural biology, chemistry, cell-based screens as well as
pharmacokinetics. Other relevant organizations main-
taining antiparasite screening operations include the
Harvard/Broad initiative, the Walter Eliza Hall Institute
for Medical Research and the St Jude Childrens Research
Hospital Memphis
(TABLE 3).
The above organizations are all making valuable
contributions in one way or another in the search for
new therapies for specific tropical diseases. However,
as the goal of these initiatives is to discover new leads
or drug candidates for tropical diseases, we now need
a strategy to track and monitor the progress of these
activities and to avoid unnecessary duplication of effort.
A coordination strategy that enhances networking and
exchange of information between these entities would
help to maximize the return on the investment made by
the various stakeholders. One possible way of enhancing
information flow is to set up a website on which inves-
tigators are encouraged to record what HTS campaigns
they have conducted or which are ongoing.
The integrated and centrally coordinated strategy
discussed below represents a focused attempt by WHO/
TDR to address a specific gap in the earlier phases of
the discovery pipeline: the identification of robust lead
compounds for tropical diseases. The idea is to share
some lessons that might be helpful for institutions
Table 2 | Some available drugs for tropical diseases
Diseases Drug Origin
Chagas’ disease Benznidazole Veterinary R&D (Roche)
Nifurtimox Veterinary R&D (Bayer)
Human African
trypanosomiasis
Eflornithine (DFMO) Anticancer R&D (MMD/TDR)
Leishmaniasis Lipo. Ampho. B (NexStar/WHO)
Miltefosine Anticancer R&D (Zentaris/TDR)
Schistosomiasis Praziquantel Veterinary R&D (Pfizer/TDR)
Oxamnaquine Veterinary R&D (Pfizer)
Helminth infections Albendazole Veterinary R&D (SKB)
Onchocerciasis Ivermectin Veterinary R&D (Merck/TDR)
Malaria Mefloquine (WRAIR/H-LaRoche/TDR)
Halofantrine (WRAIR/SKB/TDR)
Artemether (China/RPR/TDR)
Atovaquone/prog. (Wellcome (now GSK))
Arteether (Artecef/TDR)
Lapdap (GSK/TDR)
Lumefantrin/Artemeter (Novartis)
Companies and partners involved in their development are indicated. The original indications
for some of the drugs are highlighted. MMD, Marion Merrell Dow; SKB, SmithKline Beecham
(now GSK, Glaxo SmithKline); RPR, Rhône-Poulenc Rorer.
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
945
working on drug discovery for tropical diseases. For
many years, WHO/TDR has focused its drug discovery
resources on funding a network of compound assess-
ment centres
(FIG. 2). The need to follow up actives
emerging from these test centres with dedicated medic-
inal chemistry backed up by pharmacokinetic investi-
gations has now been recognized and supported with
the establishment of two further specialist networks.
Furthermore, in order to harness the output from the
various parasite genome programmes
28–30
, an additional
network has been created to triage bioinformatic data
and to identify a pipeline of molecular targets for
various disease-causing parasites. These networks are
described below.
The drug target portfolio network. In order to interpret
and capitalize on the data emerging from parasite
genome programmes, a network has been created to
develop a globally accessible database populated with a
prioritized list of potential drug targets. A comprehensive
listing of putative drug targets from human protozoan
and helminth parasites has not been carried out sys-
tematically to date. The project will help address this
issue and provide published recommendations for
selected molecular targets suitable for progression to
HTS campaigns. This should help promote the strat-
egy in both industry and academia. The TDR drug
target portfolio network consists of groups based at the
University of Washington, Seattle, USA; the University
of Pennsylvania, USA; the Sanger Centre, Cambridge,
UK; the Walter Eliza and Hall Institute for Medical
Research (WEHI), Melbourne, Australia; and the Institute
for Research in Biotechnology (UNSAM), Argentina.
The participation of an institution from a disease-
endemic country in this global consortium adds a novel
capacity-building dimension.
In addition to the above, a recent drug discovery
collaboration between Pfizer and WHO/TDR is being
extended to include support for the target portfolio
project. Pfizer is already supporting this network by
bringing its own genome triaging expertise and tech-
niques
36
to bear on the selection and prioritization of
molecular targets. A recent publication on target priori-
tization for Mycobacterium tuberculosis
55
demonstrates
the utility of this exercise across tropical diseases. Pfizer
is working with the University of Pennsylvania, the
University of California San Francisco Sandler Center,
and Inpharmatica to identify parasite homologues of
their own commercial targets for other indications. The
druggability’ of such parasite targets will be assessed and
ranked in order to facilitate prioritization by the drug
target network. The triaged information will be made
available through a database that is being developed by
the network. The synergistic, overlapping and coordi-
nated activities of the different groups present an oppor-
tunity for building a chemoinformatics and in silico drug
discovery platform for tropical diseases.
Compound screening and evaluation network. The
biological assessment network has been the engine of
TDRs drug discovery strategy for many years. It is a
unique integrated global collection of compound assess-
ment centres that allows scientists from academia and
industry to submit compounds for test free of charge.
This has given the network unrivalled access to many
thousands of diverse compounds in the search for new
antiparasitic leads. However, in the process of assessing
these diverse collections, various recurring problems
have been noted. For example, the turn-around time has
been a contentious issue, particularly with the assess-
ment of individual compounds or small collections.
This arises because of the need of the screening centres
to amass sufficient samples to make it time-effective
to run multiple parasite assays at once. However, the
screening centres are now focused on evaluating agreed
numbers of compounds based on available budgets
to ensure an efficient turn-around of data. Another
problem encountered is that many samples supplied
for test and subsequently found active enough to jus-
tify progression have not been available in sufficient
quantity. Consequently, only preliminary test data have
Box 1 | Definitions and activity criteria for hits and leads:
Hit activity criteria for protozoa
Plasmodium falciparum (K1) IC
50
<0.2 μg per ml, SI*>100
Trypanosoma brucei rhodesiense (STIB 900) IC
50
<0.2 μg per ml, SI* >100
Trypanosoma cruzi (Tulahuen) IC
50
<1.0 μg per ml, SI* >50
Leishmania donovani (L82)
Axenic amastigotes IC
50
<0.5 μg per ml, SI* >20
Amastigotes in macrophage IC
50
<1 μg per ml, SI* >20
SI* = IC
50
L-6/IC
50
parasite
Lead activity criteria for protozoa
Active in vivo (mice) in 10% dimethyl sulphoxide (DMSO) formulation at n × ≤100 mg
per kg as measured by >90% reduction in parasitaemia* and/or increase in life span**;
n = number of doses given intraperitoneally, subcutaneously or per orally daily, and
varies usually from 1–5
Malaria: Plasmodium berghei (ANKA strain), usually at 4 × 50 mg per kg*
,
**
African trypanosomiasis: T. b. brucei (STIB 795 strain), usually at 4 × 50 mg per kg*
,
**
American trypanosomiasis: T. cruzi (Tulahuen)**
Leishmaniasis: L. donovani (HU3)*
Not overtly toxic in animals at efficacious dose
Active in vitro against relevant parasite strains (for example, drug-resistant)
Hit activity criteria for helminths
Schistosomiasis: Schistosoma mansoni adults 100% inhibition of motility at 5 μg per ml
Onchocerciasis: Onchocerca lienalis mf 100% inhibition of motility at 1.25 × 10
–5
M
Onchocerca gutturosa adults 100% inhibition of motility or formazan formation at
1.25 × 10
–5
M with no obvious sign of toxicity to the monkey kidney feeder cell layer
Lead activity criteria for helminths
Active in vivo (mice) when given intraperitoneally or subcutaneously in 10% DMSO
formulation at 5 × 100 mg per kg as measured by a statistically significant reduction in
worms (>80% is highly active)
Schistosomiasis: S. mansoni adults
Onchocerciasis: O. lienalis mf
Not overtly toxic in animals at efficacious dose
Values are illustrative and are based on experience with compounds that have moved through
the evaluation network
5
.
REVIEWS
946
|
NOVEMBER 2006
|
VOLUME 5 www.nature.com/reviews/drugdisc
been obtained, which, once provided to the supplier,
have often been published without any further action
being planned. There is now increasing due diligence
being performed to assess the quality of the compounds
supplied for test and to ascertain that a commitment to
follow them up will be made by the supplier.
The investigators responsible for the individual test
centres funded by TDR are world-renowned parasitolo-
gists. Collectively, they offer a comprehensive range of
in vitro (whole parasite) screens and animal disease
models, permitting in-depth profiling against a range
of parasites and detailed assessment in various in vivo
systems to guide lead identification/optimization
(FIG. 2).
The network consists of five centres: the Swiss Tropical
Institute (STI), Basel, which provides in vitro and
in vivo screens for malaria, African trypanosomiasis,
leishmaniasis and Chagas disease; the London School of
Hygiene and Tropical Medicine (LSHTM), which pro-
vides in vitro and in vivo screens for schistosomiasis, as
well as in vivo assays for leishmaniasis and Chagas dis-
ease; the Northwick Park Institute for Medical Research
(NPIMR), London, which provides in vitro and in vivo
screens for filariasis and onchocerciasis; the Theodor
Bilharz Research Institute (TBRI), Cairo, which provides
Table 3 | Academic and public institutes offering drug screening opportunities for tropical and other diseases
Region HTS screens Whole-parasite screening and disease type
North
America
NIH USA: Chemical Genomics Initiative and Molecular Libraries
Screening Center Network (http://nihroadmap.nih.gov)
University of California San Francisco Sandler Center and
Tropical Diseases Research (http://www.ucsf.edu/mckerrow/
protocol.html)
Harvard and Broad Institute Initiative (www.broad.harvard.edu/
chembio/plaform/screening/index.htm)
Stanford University: High Throughput-Bioscience Center
(http://www.htbc.stanford.edu)
Purdue Center for Combinatorial Chemical Biology
(http://www.chem.purdue.edu/CCCB/index/shtml)
Yale University: Chemical Genomics (http://www.yale.edu)
St. Jude Children’s Research Hospital HTS efforts
(http://www.stjude.org).
McGill University: HTS facility (www.medicine.mcgill.
ca.biochem/htsfacility/index.htm)
McMaster: HTS lab (www.hts.mcmaster.ca)
Canadian Institute for Health Research, network for chemical
biology (http://www.cihr.irsc.gc.ca/e/28269.html)
Walter Reed Army Institute for Research WRAIR — malaria,
leishmaniasis and others (http://wrair-www.army.mil/)
University of Washington Seattle — malaria, trypanosomiasis
(http://depts.washington.edu/daid/)
University of California San Francisco — malaria,
trypanosomiasis (http://www.ucsf.edu/)
University of Mississippi — malaria, leishmaniasis
(http://www.pharmacy.olemiss.edu/ncnpr/)
University of Southern Florida — malaria
Europe University of Dundee Drug Discovery Initiative
(http://www.welcome.ac.uk/doc_wtx027342.html)
European Molecular biology Laboratory: Chemical Genomics
Core Facility (http://www.embl.org)
HT-Technology Development Studio, Max Planck Institute
(http://tds.mpi-cbg.de/webtds/4.html)
Medical Research Council London — malaria
(http://www.mrctechnology.org)
WISDOM: Initiative for grid-enabled drug discovery against
neglected diseases (http://wisdom.healthgrid.org/)
Swiss Tropical Institute — malaria, leishmaniasis, trypanosomiasis,
helminths (www.sti.ch)
London School of Hygiene and Tropical Medicine — malaria,
leishmaniasis, trypanosomiasis, schistosomiais (www.lshtm.ac.uk)
Northwick Park Institute for Medical Research — filariasis,
onchocerchiasis
Institute Pasteur — malaria, trypanosomiasis (http://www.pasteur.fr)
University of Anwerp, Laboratory of Microbiology, Parasitology and
Hygiene — malaria, trypanosomiasis, leishmaniasis
(http://www.ua.ac.be/lmph)
University of Liverpool — malaria (www.liv.ac.uk)
Asia/
Australia
Walter Eliza Hall Institute for Medical Research
(http://www.wehi.edu.au)
Griffith University, Eskitis Institute (www.griffith.edu.au/
centers/eskitits/
Kitasato Institute Japan — malaria, leishmaniasis, schistosomiasis
(www.kitasato.or.jp)
Indian Central Drug Research Institute Lucknow — expertise on
malaria, leishmaniasis, filariasis (www.cdriindia.org)
Institute of Parasitic Diseases Shanghai China — malaria,
schistosomiasis
National Center for Genetic Engineering and Biotechnolgy,
Thailand — malaria, tuberculosis (http://www.biotec.or.th)
Africa
and
Middle
East
Tel Aviv University: National center for HTS of Novel Bioactive
Compounds (http://www.tau.ac.il/~nchts/main.htm)
Theodor Bilharz Research Institute Cairo — schistosomiais
(http://www.tbri.sci.eg/main.htm)
Kenya Medical Research Institute — malaria, leishmaniasis
(http://www.kemri.org/kemri_centres.asp)
University of Ibadan Nigeria — malaria (http://www.ui.edu.ng/)
South
America
Instuto Oswaldo Cruz — malaria, trypanosomiasis
(www.ioc.fiocruz.br)
Instituto Venezolono de Investigaciones Cientificas, Venezuela
— trypanosomiasis
Institute for Advanced Scientific Investigation and High
Technology — malaria, leishmaniasis
Some of the institutions involved in high-throughput screening (HTS) are not primarily focused on tropical diseases (see also REF. 51). The list might not be exhaustive.
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
947
in vitro and in vivo screens for schistosomiasis; and the
Laboratory for Microbiology, Parasitology and Hygiene
at the University of Antwerp (LMPH), which provides
in vitro screens for malaria, leishmaniasis and trypano-
somiasis. Overall, the network is capable of process-
ing about 20,000 compounds per annum through the
in vitro screens and evaluating approximately 1,000
compounds per annum in vivo, based on the funding
currently provided by WHO/TDR. The turn-around
time for generating data varies for the various in vitro
and in vivo models used. On average, most in vitro test
data is available within 4–8 weeks of receiving the com-
pound whilst the turn around time for in vivo assessment
is about 8 weeks.
The compounds fed into these centres are sourced
from both industrial and academic partners and in
most cases the supply is under contractual agreement.
In recent years, hits emerging from these screens
have largely been pursued through the acquisition of
analogues, either from the original suppliers or from
commercial purveyors of compound libraries. Although
this has allowed development of preliminary structure–
activity relationships, often it has not allowed the work
to progress sufficiently to allow identification of lead
compounds that are orally efficacious in animal disease
models. The lack of a coordinated strategy encompass-
ing medicinal chemistry, pharmacology and toxicology
is now being addressed — active compounds are further
supported through the TDR medicinal chemistry and
pharmacokinetic networks in order to optimize activity
and generate robust leads. The activity criteria used for
some of these screens are presented in
BOX 1.
There are other well-established screening centres
for various tropical diseases
(TABLE 3), although these
tend not to cover the range of parasite assays embraced
by the TDR network. These include the Walter Reed
Army Institute for Research (WRAIR), which has
worked closely with TDR for many years in seeking
antimalarial leads; the Indian Central Drug Research
Institute, Lucknow; the Kitasato Institute, Japan; the
Institute for Parasitic Diseases, China; the University
of Washington, the University of North Carolina, the
University of Mississippi and the University of California
San Francisco, USA.
Database and compound storage resource in project
management. Another important element in the man-
agement of network and partnership activities is the
use of a secure interactive database for data, project and
communication management. The database managed
centrally by TDR enables the organization, collation
and management of all compounds sourced as well as
the resultant data subsequently generated from the test
centres. Compounds are organized with clear identifi-
cation numbers. A planned update of the TDR database
will enable relevant partners to enter data directly onto
the database from a remote location using a pass-
word-protected mechanism. The database promotes
enhanced communication, as recent results or presen-
tations can be shared as needed with relevant partners
at different locations in real time — for example, during
tele- or video conferencing. The integrated database
is secure and respects all confidential structures from
collaborators. Another equally important type of data-
base is the open-source database that contains various
research reagents
67,68
.
Linked to the function of the database is a central
compound storage facility where all the samples sourced
by TDR are collated and distributed to screeners in an
appropriate format. For the past few years, TDR has
retained RCC, Basel
(TABLE 4), as its compound storage
facility. The need for various types of databases
(whether secure or open-source) as well as compound
management
69,70
and storage exemplifies additional
elements of managing virtual drug discovery that are
not often discussed.
STI – Basel:
Malaria, African
trypanosomiasis,
Chagas disease,
Leishmaniasis
NPIMR-London:
Filariasis
TBRI-Cairo and LSHTM:
Schistosomiasis.
LMPH-Antwerp:
Malaria, African
trypanosomiasis,
Leishmaniasis,
Chagas disease
STI – Basel:
Malaria, African
trypanosomiasis,
Chagas disease,
Leishmaniasis
NPIMR-London:
Filariasis
Medicinal chemistry and pharmacokinetics/metabolism
networks aligned with screening centres
TBRI-Cairo and LSHTM:
Schistosomiasis
LSHTM:
Leishmaniasis,
Chagas disease
Lead identification
Secondary in vivo assays
Hit identification
Primary and repeat
in vitro assays
Lead optimization
Follow-on studies
Figure 2 | WHO/TDR-funded compound evaluation network. The compound evaluation network performs primary
in vitro screens against the various parasites. Compounds that meet the in vitro activity and cytotoxicity criteria (‘hits’) are
progressed to in vivo analysis, and subsequently medicinal chemistry and pharmacokinetic analysis to identify ‘leads’.
LMPH, Laboratory of Microbiology, Parasitology and Hygiene (Antwerp); LSHTM, London School of Hygiene and Tropical
Medicine; NPIMR, Northwick Park Institute for Medical Research (London); STI, Swiss Tropical Institute (Basel);
TBRI, Theodor Bilharz Research Institute (Cairo); WHO/TDR, Special Programme for Research and Training in Tropical
Diseases at the World Health Organization. Activity criteria for ‘hits’ and ‘leads’ are presented in
BOX 1.
REVIEWS
948
|
NOVEMBER 2006
|
VOLUME 5 www.nature.com/reviews/drugdisc
The medicinal chemistry and pharmacokinetics/
metabolism networks. The medicinal chemistry effort
currently involves one large pharmaceutical company
(Pfizer) partnering with TDR, and two biopharmaceu-
tical companies, Serono and Pharmacopeia
(TABLE 5).
Several academic institutions are also part of this net-
work: the University of Nebraska, USA; University of
Dundee, UK; University of Cape Town, South Africa;
Ohio State University, USA; and St. Jude Childrens
Research Hospital, Memphis, USA. These centres work
on the active compounds emerging from either the
screening network (whole cells) or from the HTS cam-
paigns (protein-based assays directed towards specific
molecular targets). Some of the recent active compounds
(validated hits) emerging from TDRs screening of com-
mercially sourced compounds are part of the series being
progressed through the medicinal chemistry network.
These include seven leads for various tropical diseases:
four for malaria, one for leishmaniasis, one for African
sleeping sickness and two for helminths.
The network seeks to carry out medicinal chemistry
using postdoctoral fellows based at those centres. These
pursue ‘hit to lead’ or early stage ‘lead optimization’ in
the normal iterative cycle of synthesis and biological
assessment, feeding compounds back into the screen-
ing centres
(FIG. 1). A number of fellows are linked to
institutions in developing countries. The participation
of a first-rate medicinal chemistry laboratory in a dis-
ease-endemic country provides an opportunity for the
establishment in Africa of a centre of excellence to help
promote innovation in this core area of lead discovery.
The pharmacokinetics/metabolism network, which
provides essential data for the chemists, presently con-
sists of the Monash University, Australia, and various
TDR collaborating companies such as Pfizer, Serono and
Pharmacopeia that are providing this service in-kind as
part of the ongoing collaboration. Additional academic
centres are being sought to augment this network.
Another approach to providing chemical support for
tropical disease research (one widely used by large pharma
for other therapeutic areas) is to draw on the services of
contract research organizations and institutions located in
advanced developing countries such as India and China
54
.
These are probably most productively deployed to syn-
thesize focused chemical libraries rather than engaging
in the more specific process of lead optimization. In
general, it will be beneficial to establish coordination
mechanisms similar to the integrated approach discussed
in this paper, to help harness the huge resources available
in these technologically advanced developing countries.
The risk of poor commercial return might explain why
some companies in these countries are not investing in
product discovery for tropical diseases endemic to these
geographic regions. However, an increasing number
of companies in China, India, Korea, South Africa and
Singapore are participating in PPPs to develop products
for various developing world diseases
2,9
.
Partnership characteristics and opportunities
The past 6 years has witnessed a dramatic increase in
interest in R&D directed towards producing new drugs
for tropical diseases. This has been fuelled by the creation
of various partnerships involving academia, industry and
PPPs, and the arrival of new funding from both govern-
ments and philanthropic foundations, in particular the
Gates, Wellcome Trust and Rockefeller Foundations
2,56
(
BOX 2; TABLE 6). Industry is increasing its participation
2,9
:
GSK has dedicated its Tres Cantos facility in Spain to
developing world diseases (mainly malaria and tuber-
culosis) and continues to collaborate with MMV and
GATB; the Novartis Institute in Singapore is focusing
on tuberculosis (in partnership with GATB) and den-
gue and has recently extended to malaria in partnership
with MMV and the Wellcome Trust; AstraZeneca India
is investing in tuberculosis drug R&D, as is Johnson &
Johnson; and Sanofi-Aventis has established an Impact
Malaria programme and continues to collaborate with
TDR and DNDi.
These efforts are required to sustain the drug devel-
opment pipeline for tropical diseases in the medium and
long term. In addition, drug R&D for certain diseases
Table 4 | TDR drug discovery collaborations
Name of company Type of business Type of collaborations with TDR
Pfizer Pharma/animal health Compound supply for testing
Medicinal chemistry/
pharmacokinetics
Potential HTS campaigns
Cheminformatics
Training
Serono Pharma
HTS campaigns
Medicinal chemistry/
pharmacokinetics
Training
Bayer Pharma/Animal Health
Compound supply for testing
Sanofi-Aventis Pharma
Compound supply for testing
Other collaboration
Pharmacopeia Pharma
Medicinal chemistry/
pharmacokinetics
Training
TopoTarget Pharma
Compound supply for testing
Lead optimization for malaria
Paratek Pharma
Compound supply for testing
Meiji Pharma/Animal health
Compound supply for testing
Chemtura Crop protection/vector
control
Compound supply for testing
Syngenta Agrochemicals Compound supply for testing
Dow AgroSciences Agrochemicals
Compound supply for testing
ChemDiv Chemical libraries and
contract synthesis
Compound supply for testing
Princeton
BioMolecular
Research
Chemical libraries and
contract synthesis
Compound supply for testing
Specs Chemical libraries and
contract synthesis
Compound supply for testing
ChemRoutes Chemical libraries and
contract synthesis
Compound supply for testing
RCC Contract Research Compound storage and handling
HTS, high-throughput screening.
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
949
continues to be neglected, such as those encompassed
by kinetoplastids (which DNDi is investing in, as well
as helminth infections, for which a new initiative is
being created
(BOX 3)). In a new development, Pfizer has
recently signed an agreement with the WHO/TDR to
provide compounds and other drug discovery support to
help identify leads for a wide range of tropical diseases:
malaria, African sleeping sickness, Chagas, leishma-
niasis, lymphatic filariasis, onchocerciasis and schisto-
somiasis. A similar collaboration directed at progressing
various HTS campaigns has been secured with Serono.
These new partnerships present a unique opportunity
for tropical diseases and will probably help draw other
companies into this field of research.
The recent re-emergence of interest in tropical dis-
ease research from the pharmaceutical sector is also
supported by the involvement of the animal health and
agrochemical industries, as well as specialty chemical
companies, who are all contributing compounds for
evaluation for human infectious diseases. This is exem-
plified by the collaboration of WHO/TDR with such
companies as Syngenta, Chemtura and Bayer Animal
Health
(TABLE 4). Forte Dodge is partnering with TDR in
clinically progressing the animal-health product moxid-
ectin for the treatment of onchocerciasis
24
.
Although TDR maintains a network of screening
centres with a substantial overall capacity for testing com-
pounds in whole-cell screens, it can be difficult to source
samples with an appropriate rationale for testing and in
sufficient quantity to facilitate follow-up assessment.
Increasing efforts are being put into sourcing high-quality
compounds with a defined testing rationale. Companies
are sought that might have biologically, biochemically or
pharmacophore-relevant compounds that they would be
willing to provide for assessment for their potential to
treat tropical diseases. Experience shows that although
many scientists in industry are extremely willing to
provide compounds for testing, higher management is
often more reticent due to perceived problems in expos-
ing their intellectual property to competitors or putting
at risk drugs in commercial development. Such issues,
although often requiring protracted discussion, can
usually be addressed satisfactorily, and TDR plus other
PPPs have been successful in establishing agreements
with industry to enable them to partner and contribute
compounds for evaluation against tropical diseases.
Partnerships for drug discovery with both industry
and academia typically involve the following: the supply
of biologically or biochemically relevant compounds,
or natural products, for screening against parasites;
supporting investigators to validate and obtain pro-
teins as molecular targets for HTS campaigns; access-
ing or supporting centres to conduct HTS campaigns
using diverse or focused compound libraries; funding
medicinal chemistry, pharmacokinetics/metabolism and
toxicological assessment for lead identification and opti-
mization, as well as supporting the establishment and
maintenance of databases to facilitate drug discovery for
tropical diseases.
These collaborations are normally covered by ‘materi-
als transfer’ or ‘collaborative’ contracts as exemplified by
the recent agreements between WHO/TDR and Pfizer,
Serono and Chemtura. The Pfizer collaboration focuses
initially on lead discovery, with the company supply-
ing thousands of compounds (including those with a
known biological/biochemical rationale) to be tested
against target parasites in TDRs screening network. Hits
emerging from this programme will be expanded using
TDR-funded medicinal chemists based at Pfizer. The col-
laboration is being extended to pursue HTS campaigns
against molecular targets, and to use cheminformatics to
identify new targets and compounds
36,37,57
.
The Serono–WHO/TDR collaboration centres on
drug discovery through HTS campaigns and allows for
hit expansion with medicinal chemistry support. Serono
is using its compound libraries for HTS against putative
new drug targets selected by TDR in association with
collaborators based in academia. Hits identified in these
protein-based assays are assessed in the TDR parasite
screens. Whole-cell actives are then further elaborated
to develop SAR using TDR-funded medicinal chemistry
resources based in Serono. The Chemtura–WHO/TDR
agreement focuses initially on supply of test compounds.
These and other companies helping TDR in the discov-
ery of new leads are highlighted in
TABLE 4. Collectively,
these represent a significant coordinated level of early
discovery activity for multiple tropical diseases.
The globally integrated and focused strategy depicted
in
FIG. 1 is helping to stimulate industry worldwide to
participate in lead discovery for tropical diseases. It is
also attracting more academic investigators to work in
the field of ‘neglected diseases’ drug discovery. It com-
plements and synergizes with the activities of PPPs such
as MMV, DNDi and GATB by facilitating the progres-
sion of new leads into the development pipeline of these
organizations or other institutions. It is noteworthy that a
number of lead series currently being developed by PPPs
benefited from initial involvement with WHO/TDR,
either by direct support for synthesis programmes or by
access to the compound-assessment network. Examples
include the antimalarial ozonides
7
, bis-amidines
49
and
dihydrofolate reductase inhibitors
39
being developed by
Table 5 | The medicinal chemistry network
Workstations (pharma/
academia)
Number of post-
doctoral fellows
Pfizer Two fellows
Serono Two fellows
Pharmacopeia One fellow
University Cape Town Two fellows
University of Nebraska One fellow
University of Dundee One fellow
St Jude Children Hospital One fellow
Ohio State University One fellow
Some of the postdoctoral fellows are from disease-endemic
countries. They receive on-the-job training in order to
contribute to specific projects in the network. The interface
between medicinal chemistry and the networks engaged in
compounds assessment and pharmacokinetic profiling is
managed jointly by WHO/TDR and partners.
REVIEWS
950
|
NOVEMBER 2006
|
VOLUME 5 www.nature.com/reviews/drugdisc
MMV, and trypanothione reductase
16
and farnesyltrans-
ferase inhibitors
58
previously supported by DNDi. This
has provided these organizations with sufficient data to
allow critical assessment of the work and to commence
funding at the lead optimization stage.
Arguably, one of the more novel and attractive ele-
ments of this innovative lead discovery approach is the
integration of capacity building through clear project
deliverables. Such capacity building in lead discovery, as
shown in
FIG. 1, is not restricted to the developing world
— through network activities it can also be pursued in
institutions in developed countries. In the context of
disease-endemic countries, this approach will encourage
local technology development — for example, through
exploration of natural products as potential leads, and
technology transfer through the various network activi-
ties. TDR is supporting two natural-product screening
centres in Kenya and Nigeria. Sustainable capacity can
be developed in disease-endemic countries if the most
talented scientists and institutions are engaged in these
partnerships for lead discovery with the same level of
rigour as their colleagues in developed countries. In
Box 2 | Synergies of partners in lead discovery for tropical diseases
Academia. The role of academic laboratories is indispensable in lead discovery for tropical diseases. Tropical disease
expertise in in vitro and in vivo screening
(TABLE 3), development of new screening tools, improved animal models,
genomics, and target identification/validation resides within academia. In addition, academic laboratories are making
great strides in high-throughput screening (HTS), medicinal chemistry and pharmacokinetic analysis to aid the
discovery and progression of leads into development candidates for tropical diseases, and this should be encouraged.
For example, TDR has collaborated with the Walter Eliza and Hall Institute for Medical Research in Melbourne to run
HTS against three parasite targets: trypanothione reductase (from Trypanosoma cruzi), farnesyl pyrophosphate
synthetase (Trypanosoma brucei) and pyrophosphokinase (Plasmodium falciparum). The initial hits emerging from the
three campaigns have been evaluated in whole parasites at the WHO/TDR compound screening centres, and follow-
up medicinal chemistry/pharmacokinetic studies are now under consideration. Similar HTS campaigns have been
commissioned by Drugs for Neglected Diseases Inititative (DNDi) at other academic institutions. Such laboratories
(TABLE 3), as discussed previously, are well established for HTS campaigns. Further participation from academia will
probably be encouraged by such success stories as the selection of a synthetic peroxide OZ277 by the Medicines for
Malaria Venture (MMV) for development for malaria (now in Phase II clinical trials)
7
. This involved participation from
both industry and academia, as well as input from experts with pharma experience. Although the increasing role of
academic centres in drug discovery for tropical diseases is a welcome development, partnership with industry and
other centres will help to maximize output.
Industry. TDR and the new private–public partnerships (PPPs) have a track record in collaborating with industry to identify
new drug candidates for human health
64
. Indeed, a focused lead discovery effort that incorporates a clear milestone-driven
approach and a ‘win–win’ intellectual property strategy promises to attract more industry involvement. Industry typically
provides compound libraries, infrastructure and know-how for HTS, medicinal chemistry and ADMET (absorption,
distribution, metabolism, excretion and toxicity) profiling. However, academia is increasingly starting to have a significant
role in the provision of chemistry and pharmacokinetic expertise and in developing HTS capabilities
7,34,35,53,64,65
.
TDR or other public-sector participants typically bring knowledge of the disease and target product profile to guide
discovery, molecular targets for screening from academic collaborators and funding from its stakeholders to leverage the
investment of industry (for example, by funding personnel, including fellows from developing countries, to work on
projects as well as portfolio management).
PPPs. Some PPPs involved in tropical disease R&D
(TABLE 6) are focusing on product development with less investment in
the early stages of lead discovery (for example, HTS against molecular targets and medium-throughput screening against
parasites in vitro). Investment in early drug discovery is essential in order to ensure a sustainable portfolio of lead
compounds for further optimization and development. The focused lead discovery strategy and the work of academic
centres discussed here will complement and synergize with PPPs such as MMV, Global Alliance for TB Drug Development
(GATB)
and DNDi.
Philanthropic foundations. Organizations such as the Bill and Melinda Gates Foundation, Rockefeller Foundation and the
Wellcome Trust are investing in discovery
(TABLE 6). They make funding available for such work as long as a clear
rationale exists supported by the appropriate technical and management expertise to achieve agreed objectives. Some of
the projects supported through the Gates Grand Challenges initiative should help to stimulate lead discovery in the
medium to long term. In response to the gap in the availability of new tuberculosis leads, the Gates Foundation has
developed a new strategy for tuberculosis drug discovery, which is much needed (See Further information, Bill & Melinda
Gates Foundation: Call For Proposals — Tuberculosis). It would seem that the strategy for this programme is similar in many
respects to the network and partnership approach discussed in this paper. It is anticipated that the target prioritization
exercise discussed above, which encompasses tuberculosis (see also
REF. 55), will complement this new effort.
In addition to supporting the University of Dundee’s drug discovery efforts, the Wellcome Trust is also funding a more
general initiative focused on early stages of lead discovery/optimization. However, this is not directed specifically towards
tropical diseases and can encompass any human disease.
National and international research agencies. Some national and international agencies
(TABLE 6) might have
opportunities for funding and capacity building in various areas of tropical diseases.
These various initiatives, some with varied scope, funding and strategy are an excellent demonstration of the progress
being made in tropical disease R&D. It also shows the need to increase coordination in order to better manage interfaces,
gaps and data flow from these efforts.
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
951
contrast to past capacity-building activities in developing
countries, this approach is linked to clear project delivera-
bles, management and accountability. There have already
been some successes through the fellowship component
of this approach, with scientists involved with HTS cam-
paigns at Serono going back to their countries to put
their new-found experience into practice
59
.
Currently, global interest in the promotion of innova-
tion for new treatments for developing-world diseases
is high. The G8 meeting in 2005
(REF. 60), as well as the
Millennium Development Goals
61
, emphasize the role of
partnerships in providing worldwide healthcare solutions.
The WHO Commission on Intellectual Property Rights,
Innovation and Public Health also highlighted the need
for innovative product discovery for diseases affecting
developing countries
11
. Indeed, increased emphasis on
innovative lead discovery will help ensure sustainability
in the availability of new products for the control of tropi-
cal diseases both in the medium and long term. The hope
is that public donor agencies and foundations will invest
more resources in this area. Some recent indication of
such improved funding comes from a call from the Gates
Table 6 | Organizations funding (or likely to fund) research into tropical diseases
Organization Website
Philanthropic foundations
Bill & Melinda Gates Foundations http://www.gatesfoundation.org
Burroughs Wellcome http://www.bwfund.org
Grand Challenges http://www.grandchallengesgh.org
Rockefeller Foundation http://www.rockfound.org
Wellcome Trust http://www.wellcome.ac.uk
Public organizations/institutions*
European Union http://ec.europa.eu/research/health/poverty-diseases
UK DFID http://www.dfid.gov.uk/research
US National Institute of Health http://www.nih.gov; http://www.niaid.nih.gov
International Development Research Council Canada http://www.idrc.ca
Canadian Institute for Health Research http://www.cihr-irsc.gc.ca
Public–private partnerships
Drugs for Neglected Diseases Initiative http://www.dndi.org
Global Alliance for TB Drug development http://www.tballiance.org
Medicines for Malaria Venture http://www.mmv.org
Institute for One World Health http://www.iowh.org
International organizations and development banks
Special Programme for Research and Training (TDR) at WHO http://www.who.int/tdr
The World Bank http://www.worldbank.org
Regional Development Bank http://www.iadb.org; http://www.afdb.org;
http://www.adb.org
United Nations Education, Scientific and Cultural
Organization (UNESCO)
http://www.unesco.org
This list is not exhaustive. *Certain national agencies concerned with international development and research such as United
States Agency for International Development USAID (www.usaid.gov), Japan International Cooperation Agency JICA
(http://www.jica.go.jp), Swedish International Development Agency (http://www.sida.se), German Agency for Technical
cooperation (http://www.gtz.de/home/english), Swiss Agency for Development and Cooperation (www.sdc.admin.ch),
Danish International Development Agency DANIDA, Canadian International Development Agency (www.cida.gc.ca), Netherlands
Ministry of Development Cooperation, and other national as well as research councils and ministers.
Foundation for proposals for discovery research directed
towards TB (see Further information, Bill & Melinda
Gates Foundation: Call For Proposals — Tuberculosis).
In view of the changing nature of drug research for
tropical diseases, more robust coordination mechanisms
are needed for lead discovery. A promising vehicle for
delivering and coordinating the discovery of new lead
compounds is exemplified by the integrated networks
embracing compound screening, medicinal chemistry,
pharmacokinetics/metabolism, and the development of
a prioritized drug target portfolio
(FIG. 1). This strategy
offers a cost-effective solution to filling the demand for
robust lead compounds suitable for further develop-
ment
62,63
. Initial cost assessment based on TDRs lead
discovery experience using the network and partnership
method described here suggests that two high-quality
lead compounds can be discovered every year with an
annual budget of about US$7 million. This includes
investment in the development of new technologies
— such as new drug screening tools and prioritization
of drug targets to facilitate HTS campaigns, as well as
capacity building — that will help sustain lead discovery
REVIEWS
952
|
NOVEMBER 2006
|
VOLUME 5 www.nature.com/reviews/drugdisc
for tropical diseases in the medium to long term. This is
good value for money considering the high attrition rate
in early drug discovery. Obviously this estimate does not
include in-kind contributions from the industry.
Challenges of lead discovery
Managing multiple partners (FIG. 1; BOX 2) from diverse
cultures and with a wide range of expertise in a network
charged with a unique drug discovery objective is not an
easy undertaking
2,21
. The success of these networks largely
depends on the people doing the work, the available
facilities, the mechanism(s) put in place for programme
management by the coordinating body and the incentives
provided for reaching defined goals. Perhaps the single
most important stimulus is the appreciation that the work
being performed is for the public good, not for profit.
Many scientists in industry are willing to contribute their
expertise to help advance projects focused on developing-
world diseases. This is also true of academic scientists
who are increasingly pursuing product-driven research
for tropical diseases even though it is often perceived to
be less rewarding in terms of career prospects than some
of the more fashionable healthcare issues. Attracting
younger academic investigators into this field will require
an increase in targeted funding. However, good science
alone is not sufficient to ensure success — in all the
programmes discussed the science has to be supported
by strong management. In this context, the importance
of a project champion cannot be overemphasized, and
without such a leader many promising avenues of drug
discovery will fail to make progress. The overall balance
of good science, appropriate funding, enthusiasm and
clear management will determine the outcome of all
science-based programmes.
It should also be noted that a key element in success-
fully managing virtual drug discovery is flexibility in
decision making
2
. This includes the ability to prioritize
projects, re-allocate resources and terminate projects that
are not going well. However, the industry mantra ‘kill
quick, kill cheap’ often does not find ready acceptance
in an academic setting, and the different management
styles resulting from the varying cultural experience of
all the partners (academia, industry and donors) in a net-
work/partnership setting can sometimes cause difficulty
in decision making.
Mitigation of risks in lead discovery
Lead discovery is an inherently high-risk activity, as
demonstrated by the corresponding attrition rates. Some
of the challenges highlighted above can be overcome
through the establishment of clear processes for project
and portfolio management in order to deliver lead dis-
covery objectives. The need for competitive project selec-
tion and review procedure by external experts promises
to reduce attrition and the cost of lead discovery. Expert
scientists from academia and industry are readily avail-
able to support and invest their time at no or limited
cost for the purposes of reviewing and recommending
promising projects for such endeavours.
A clear understanding of tropical diseases, desired
product profiles for new drugs to guide R&D, and the
needs of disease-endemic countries, are key to the dis-
covery of relevant molecules for further development
2
.
The use of focused target product profiles
(TABLE 1)
to guide lead discovery and development candidate
selection increases the chances of successful control pro-
grammes if and when such products reach the market.
The focus on innovative lead discovery fills a crucial
gap in the tropical diseases drug development pipeline.
The plan recognizes the important role of partnerships,
as well as the participation of developing country scien-
tists and institutions, in order to achieve the Millennium
Development Goals and to provide a lasting solution to
the product-access crises.
Box 3 | A new initiative focusing on helminth drug discovery
In recognition of the urgent need for new anthelminthics, WHO/TDR is currently
coordinating and facilitating drug discovery for helminth infections. An informal
consultation meeting convened by TDR in February 2005 identified the need for an
initiative to facilitate the discovery and development of new products for diseases
resulting from infections with schistosoma and the filariae. The Genomics and Discovery
Research committee of TDR endorsed proceeding with such an initiative but
recommended that a focused meeting of world experts from industry, academia and the
donor community be convened to provide further guidance. Subsequently, the meeting
was held in Tokyo in March 2006 and the concept of the helminth initiative fully
endorsed. TDR was tasked with the establishment and incubation of the Helminth
Initiative, focusing on antihelminthic drug discovery to identify new candidates that can
be advanced to development. It is expected that success in the next 2–3 years might help
to build a case for an independent public–private partnership for anthelminthic R&D.
Genomics HTS
Lead ID
(in vitro/in vivo
test, medicinal
chemistry, PK)
Lead
optimization
Preclinical
development
Malaria:
four lead series
WEHI:
three targets
Pfizer collaboration
Helminth initiative
Serono collaboration
Portfolio
of drug
targets and
database
being
developed
African
trypanosomiasis:
one lead series
Leishmaniasis:
one lead series
Helminths:
one lead series
HDAC
inhibition for
malaria
Figure 3 | The growing WHO/TDR drug discovery portfolio. Molecular targets being
pursued as part of the Serono/TDR collaboration include: a Plasmodium falciparum serine
protease, PfSub-1, provided by M. Blackman (MRC, London), a P. falciparum Ca
2+
-dependent
protein kinase, PfCDK-1, supplied by B. Kappes (U. Heidelberg) and a cysteine peptidase
from Leishmania mexicana, LmCPB, provided by J. Mottram (University of Glasgow). The
targets pursued at WEHI include Trypanosoma cruzi trypanothione reductase from A.
Fairlamb (University of Dundee), Trypanosoma brucei farnesyl pyrophosphate synthase
provided by E. Oldfield (Univeristy of Illinois), and P. falciparum pyrophosphokinase from
Sirawiraporn (Mahidol University). HDAC, histone deacetylase; WEHI, Walter Eliza and
Hall Institute for Medical Research; WHO/TDR, Special Programme for Research and
Training in Tropical Diseases at the World Health Organization.
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
953
1. Trouiller, P. et al. Drug development for neglected
diseases: a deficient market and a public-health policy
failure. Lancet 359, 2188–2195 (2002).
A review article discussing drug development
for neglected diseases and non-market-
driven mechanisms to stimulate drug
development.
2. Nwaka, S. & Ridley, R. G. Virtual drug discovery and
development for neglected diseases through public-
private partnerships. Nature Rev. Drug Discov. 2,
919–928 (2003).
Presents an overview of processes for drug
discovery and development for tropical diseases
through public–private partnerships.
3. World Health Organization. World Health Report
(2004).
4. Nwaka, S. Drug discovery and beyond: the role of
public–private partnerships in improving access to
new malaria medicines. Trans. Royal Soc. Trop. Med.
Hygiene 99, S20–S29 (2005).
5. Pink, R. et al. Opportunities and challenges in
antiparasitic drug discovery. Nature Rev. Drug Discov.
4, 727–740 (2005).
6. Bleicher, K. H. et al. Hit and lead generation: beyond
high-throughput screening. Nature Rev. Drug Discov.
2, 369–378 (2003).
7. Vennerstrom, J. L. et al. Identification of an
antimalarial synthetic trioxolane drug
development candidate. Nature 430, 900–904
(2004).
Elegant work on the identification of OZ 277 as
development candidate for malaria chemotherapy.
The work exemplifies what can be achieved through
public–private partnerships and investment in
early discovery research.
8. TDR News Number 73 (2004).
9. Moran, M. et al. The New Landscape for Neglected
Diseases Drug Development (Wellcome Trust,
London, 2005).
10. Nwaka, S. & Widdus, R. in Combating Diseases
Associated with Poverty: Financing Strategies for
Product Development and the Potential Role of
Public–Private Partnerships (eds Widdus, R. &
White, K.) 164–168 (Initiative on Public-Private
Partnerships for Health, Geneva. (2004).
11. Report of the Commission on Intellectual Property
Rights, Innovation and Public Health. Public Health,
Innovation and Intellectual Property Rights (CIPIH)
(World Health Organization, 2006).
12. International Federation of Pharmaceutical
Manufacturers Association. Research and
Development for Neglected Diseases: Lessons
Learned and Remaining Challenges (IFPMA, 2005).
13. Malaria research and development: an assessment
of global investment. Malaria R&D Alliance Report
(2005).
14.
Hotez, P. et al. Incorporating a rapid-impact package
for neglected tropical diseases with programs for
HIV/AIDS, tuberculosis, and malaria. PLoS Med. 3
(2006).
15. Report of the Commission for Africa. Our Common
Interest [online], <http://www.commissionforafrica.
org/english/report/thereport/english/11-03-05_cr_
report.pdf
> (2005).
16. Fairlamb, A. H. Target discovery and validation with
special reference to trypanothione. In Drugs against
parasitic diseases: R&D methodologies and issues
(eds Fairlamb, A. H., Ridley, R. G. & Vial, H. J.)
107–118 (WHO/TDR, Geneva 2003).
17. Behm, C. A. et al. RNAi-based discovery and
validation of new drug targets in filarial nematodes.
Trends Parasitol. 21, 97–100 (2005).
18. Rosenthal, P. J. Antimalarial drug discovery: old and
new approaches. J. Exp. Biol. 206, 3735–3744
(2003).
19. Witty, M. Current strategies in the search for novel
antiparasitic agents. Int. J. Parasitol. 29, 95–103
(1999).
20. Remme, J. H. F. et al. Strategic emphasis for tropical
diseases research: a TDR perspective. Trends
Parasitol. 18, 421–425 (2002).
21. Ridley, R. G. Research on infectious diseases requires
better coordination. Nature Med. 10, S137–140
(2004).
22. Horton, J. Drug development for tropical diseases —
present situation, future perspectives. Trends Parasitol.
19, P06 (2003).
23. Omura, S. and Crump, A. The life and times of
ivermectin — a success story. Nature Rev. Microbiol.
2, 984–989 (2004).
24. Cotreau, M. M. et al. The antiparasitic moxidectin:
safety, tolerability, and pharmacokinetics in humans.
J. Clin. Pharmacol. 43, 1108–1115 (2003).
25. Fidock, D. A. et al. Antimalarial drug discovery:
efficacy models for compound screening. Nature Rev.
Drug Discov. 3, 509–520 (2004).
26. Gelb, M. H. et al. Protein farnesyl and N-myristoyl
transferases: piggy-back medicinal chemistry targets
for the development of antitrypanosomatid and
antimalarial therapeutics. Mol. Biochem. Parasitol.
126, 155–163 (2003).
27. Andrews, K. T. et al. Anti-malarial effect of histone deace-
tylation inhibitors and mammalian tumour cytodiffer-
entiating agents. Intl. J. Parasitol. 30, 761–768 (2000).
28. Gardner, M. J. Genome sequence of the human
malaria parasite Plasmodium falciparum. Nature.
419, 498–511 (2002).
29. El-Sayed, N. M. et al. Comparative genomics of
trypanosomatid parasitic protozoo. Science 309,
404–409 (2005).
A reference article on the comparative genomics of
trypanosomatid parasites.
30. Mitreva, M. et al. Comparative genomics of
nematodes. Trends Genet. 21, 573–281 (2005).
31. Joachimiak, M. P. et al. The impact of whole genome
sequence data on drug discovery- a malaria case
study. Mol. Med. 7, 698–710 (2001).
Future perspectives
The concept of integrated drug discovery for tropical
diseases through networks/partnerships
(FIG. 1) using
virtual and portfolio methodology
2
promises to revolu-
tionize translational research not only for tropical dis-
eases but for other diseases too. In the past, it was often
thought that pharmaceutical companies had drug can-
didates ‘sitting on the shelf ’ for tropical diseases and that
these could be liberated given the appropriate financial
inducement. This thinking is somewhat naive given that
pharmaceutical companies are now involved in nearly
half of new neglected-disease drug development activity
on a non-commercial basis
2,9,10,19
. The fact is that industry
is also trying to identify efficient and cost-effective ways
to increase their productivity in drug discovery
19,44,45
.
Some companies now see value in supporting tropical
disease drug discovery as a way of boosting their lead
discovery efforts for profitable diseases. For example,
a ‘hit to lead’ programme targeting a parasite enzyme
might help a company build a chemical library around
the lead for testing against a human isoenzyme germane
to a commercial market. This concept is increasingly
gaining acceptance. Furthermore, the animal health
industry is in need of new chemical entities for the vet-
erinary market. Investment in lead discovery for human
parasitic diseases might be seen as an avenue to identify
new potential animal-health products.
The implications of this approach for the public health
sector and pharma, animal health and agrochemical
industries are considerable. Overall, the approach will
help to reduce the high risk and cost associated with
lead discovery
62,63
as well as stimulating the pipelines
of the various sectors. For public health, this approach
promises to create value through managed lead discov-
ery portfolio efforts such as the ones being pursued by
TDR
(FIG. 3) and other organizations. The fruits of these
endeavours can then be transferred to development
partners or leveraged for additional resources to support
future lead discovery efforts for diseases that have little
or no potential of commercial return.
The networks are also a strong instrument for
facilitating capacity-building in drug discovery, insti-
tutional strengthening and technology transfer to
disease-endemic countries. They might also present a
good platform for harnessing available drug discovery
expertise in advanced developing countries. Clearly,
networking, partnerships and capacity-building are
useful and have yielded good results and should be seen
as part of the solution. Strong political will and local
commitment to research and economic development
are also needed. In the words of the WHO Commission
on Intellectual Property Rights, Innovation and Public
Health
11
, “In the longer term, the development of
innovative capacity for health research in developing
countries will be the most important determinant of
their ability to address their own need for appropri-
ate health-care technologies.” As the report goes on to
say, “The formation of effective networks, nationally
and internationally, between institutions in develop-
ing countries and developed countries, both formal
and informal, are an important element to building
innovative capacity. ” This reflects the views expressed
in this paper on the need for increased investment
in upstream drug discovery for tropical diseases.
REVIEWS
954
|
NOVEMBER 2006
|
VOLUME 5 www.nature.com/reviews/drugdisc
32. Jones, A. K. et al. Chemistry-to-gene screens in
Caenorhabditis elegans. Nature Rev. Drug Discov. 4,
321–330 (2005).
Exemplifies use of C. elegans in discovery of novel
anthelminthics.
33. Bajorath, J. Integration of virtual and high-throughput
screening. Nature Rev. Drug Discov. 1, 882–893
(2002).
34. Mackey, Z. B. et al. Discovery of Trypanocidal
compounds by whole cell HTS of Trypanosoma brucei.
Chem. Biol. Drug Des. 67, 355–363 (2006).
35. Chong, C. R. et al. A clinical drug library screen
identifies astemizole as an antimalarial agent. Nature
Chem. Biol. 2, 415–416 (2006).
Identification of an antimalarial lead through
screening a drug-based library against whole cells.
36. Lipinski, C. & Hopkins, A. Navigating chemical space for
biology and medicine. Nature 432, 855–861 (2004).
37. Paolini G. V. et al. Global mapping of pharmacological
space. Nature Biotechnol. 24, 805– 815 (2006).
A discussion of new cheminformatic tools that will
be applicable to parasitic drug discovery.
38. Clark, D. E. and Pickett, S. D. Computational methods
for the prediction of ‘drug-likeness’. Drug Discov Today
5, 49–58 (2000).
39. Yuvaniyama, J. et al. Insights into antifolate resistance
from malarial DHFR-TS structures. Nature Struct. Biol.
10, 357–367 (2003).
40. Kuo, M. R. et al. Targeting tuberculosis and malaria
through inhibition of enoyl reductase: compound
activity and structural data. J. Biol. Chem. 278,
20851–20859 (2003).
41. Blundell, T. L. et al. High-throughput crystallography
for lead discovery in drug design. Nature Rev. Drug
Discov. 1, 45–54 (2002).
42. Anderson, A. C. The process of structure-based drug
design. Chem. Biol. 10, 787–797 (2003).
43. Cos, P. et al. Anti-infective potential of natural
products: How to develop a stronger in vitro ‘proof-of-
concept’. J. Ethnopharmacol. 106, 290–302 (2006).
44. Bush, K., Macielag, M. & Weidner-Wells, M. Taking
inventory: antibacterial agents currently at or beyond
phase I. Curr. Opin. Microbiol. 7, 466–476 (2004).
45. Overbye, K. M. & Barrett, J. F. Antibiotics: where did
we go wrong? Drug Discov. Today 10, 45–52 (2005).
46. Gribbon, P. & Sewing, A. High throughput drug
discovery: what can we expect from HTS? Drug Discov.
Today 10, 17–22 (2005).
47. Barr, S. C. et al. A cysteine protease inhibitor protects
dogs from cardiac damage during infection by
Trypanosoma cruzi. Antimicrob. Agents Chemother.
49, 5160–5160 (2005).
48. Cohen, J. Public health. Gates Foundation picks
winners in Grand Challenges in Global Health. Science
309, 33–35 (2005).
49. Sturk, L. M. et al. Distribution and quantitation of the
anti-trypanosomal diamidine 2, 5-bis(4-
amidinophenyl)furan (DB75) and its N-methoxy
prodrug DB289 in murine brain tissue. Acta Trop. 91,
131–43 (2004).
50. McKerrow, J. H. Designing drugs for parasitic diseases
of the developing world. PLoS Med. 2 (2005).
Presents a summary of HTS capability in academia.
51. Dalrymple, M. et al. Academia-industry partnerships
in drug discovery. Expert Opin. Drug Discov. 1, 1–5
(2006).
52. Brown, E. Screening in academe: a perspective on
implementation of university-based small molecule
screening. J. Biomol. Screen. 8, 377–379 (2003).
53. Stein, R. High-throughput in academia: the Harvard
experience. J. Biomol. Screen. 8, 615–619 (2003).
54. Morel, C. et al. Health innovation networks to help
developing countries address neglected diseases.
Science 309, 401–404 (2005).
A good example of how target prioritization can aid
drug discovery for tropical diseases.
55. Hasan, S. et al. Prioritizing genomic drug targets in
pathogens: application to Mycobacterium
tuberculosis. PLOS Comp. Biol. 2, e61 (2006).
An analysis of the changing landscape of
neglected-disease research emphasizing increasing
involvement from pharmaceutical companies.
56. Moran M. A breakthrough in R&D for Neglected
diseases: new ways to get the drugs we need. PLoS
Med. 2 (2005).
57. Hopkins, A. L. and Groom, C. R. The druggable
genome. Nature Rev. Drug Discov. 1, 727–730
(2002).
58. Buckner, F. S. et al. Protein farnesyl transferase
inhibitors for the treatment of malaria and African
trypanosomiasis. Curr. Opin. Investig. Drugs. 6,
791–797 (2005).
59. Perrin, D., Scheer, A. & Wells T. Collaborating to find
new approaches to tropical diseases. Eur. Pharma.
Rev. 3, 52–55 (2006).
60. G8 Gleneagles Communique on Africa [online],
<http://www.fco.gov.uk/Files/kfile/PostG8_
Gleneagles_Communique.pdf> (2005).
61. Juma, C. & Yee-Cheong, L. UN Millenium
Project- task force on science, technology and
innovation [online], <http://www.
unmillenniumproject.org/reports/tf_science.htm>;
<http://www.unmillenniumproject.org/reports/tf_
essentialmedecines.htm> (2005).
62. DiMasi, J., Hansen, R., and Grabowski, H. The price of
innovation: new estimates of drug development costs.
J. Health Econ. 22, 325–330 (2003).
63. MMV business plan (2003).
64. Gutteridge, W. E. TDR collaboration with
pharmaceutical industry. Trans. Royal Soc. Trop. Med.
Hyg. 24 May 2006 [epub ahead of print].
65. Baldwin, J. et al. High-throughput screening for potent
and selective inhibitors of Plasmodium falciparum
dihydroorotate dehydrogenase. J. Biol. Chem. 280,
21847–21853 (2005).
66. Gelb, M. H. & Hol, W. G. Parasitology. Drugs to
combat tropical protozoan parasites. Science, 297,
343–344 (2002).
67. Bahl, A. et al. PlasmoDB: the Plasmodium genome
resource. A database integrating experimental and
computational data. Nucleic Acids Res. 31, 212215
(2003).
68. Maurer, S. M., Rai A., and Sali A. Finding cures for
tropical diseases: is open source an answer? PLoS
Med. 3, e56 (2004).
69. Spencer, A. P. The challenges of managing a
compound collection. Eur. Pharma Rev. 51–57
(2004).
70. Yates, I. Compound management comes of age.
Drug Discov. World 4, 35–42 (2003).
Acknowledgements
The authors would like to thank R. Ridley, A. Oduola and
J. Lazdins for their support, A. Fairlamb and T. Wells for
critically reading the manuscript. We also thank F. Fakorede,
M.-A. Mouries, C. Alias and L. Swarb for their help with
the paper.
Competing interests statement
The authors declare no competing financial interests.
FURTHER INFORMATION
Medicines for Malaria Venture: http://www.mmv.org
Grand Challenges in Global Health — Limit Drug Resistance:
http://www.gcgh.org/subcontent.aspx?SecID=396
EU Commission — Poverty-Related Diseases:
http://europe.eu.int/comm/research/health/poverty-
diseases/projects/124_en.htm
EU Commission — New TB Drugs: http://ec.europa.eu/
research/health/poverty-diseases/projects/130_en.htm
Bill & Melinda Gates Foundation: Call For Proposals –
Tuberculosis: http://www.gatesfoundation.org/
GlobalHealth/Grantseekers/RFP/RFP_TB.htm
Access to this links box is available online.
REVIEWS
NATURE REVIEWS
|
DRUG DISCOVERY VOLUME 5
|
NOVEMBER 2006
|
955
... The functional group at position C-3 could therefore regulate lipophilicity [37]. Consistent with the parent drug CPX (1), analogs 2-8 and 11-22 are predicted to also have high gastrointestinal (GI) absorption; since these test compounds have less than ten rotatable bonds (< 10) and a total polar surface area below one hundred and forty (< 140) (Appendix Table A. 3-4) [48]. CPX (1) and 8 are the only test compounds predicted to show blood-brain barrier (BBB) permeation, besides high GI absorption (Appendix A Table A ...
... CPX (1) proved to be a hit compound (IC 50 ≤ 10, SI ≥ 10) against the animal trypanosome T. congolense [47,48], with low cytotoxicity (IC 50 ≥ 100 µM) [47,48]. However, in most countries, fluoroquinolones are prohibited from extra label drug use in all food-producing animal species because withdrawal times are not available [56]. ...
... CPX (1) proved to be a hit compound (IC 50 ≤ 10, SI ≥ 10) against the animal trypanosome T. congolense [47,48], with low cytotoxicity (IC 50 ≥ 100 µM) [47,48]. However, in most countries, fluoroquinolones are prohibited from extra label drug use in all food-producing animal species because withdrawal times are not available [56]. ...
Article
Full-text available
Tropical diseases, such as African trypanosomiasis, by their nature and prevalence lack the necessary urgency regarding drug development, despite the increasing need for novel, structurally diverse antitrypanosomal drugs, using different mechanisms of action that would improve drug efficacy and safety. Traditionally antibacterial agents, the fluoroquinolones, reportedly possess in vitro trypanocidal activities against Trypanosoma brucei organisms. During our research, the fluroquinolone, ciprofloxacin (1), and its analogs (2–24) were tested against bloodstream forms of T. brucei brucei, T. b. gambiense, T. b. rhodesiense, T. evansi, T. equiperdum, and T. congolense and Madin-Darby bovine kidney cells (cytotoxicity). Ciprofloxacin [CPX (1)] demonstrated selective trypanocidal activity against T. congolense (IC50 7.79 µM; SI 39.6), whereas the CPX derivatives (2–10) showed weak selective activity (25 < IC50 < 65 µM; 2 < SI < 4). Selectivity and activity of the CPX and 1,2,3-triazole (TZ) hybrids (11–24) were governed by their chemical functionality at C-3 (carboxylic acid, or 4-methylpiperazinyl amide) and their electronic effect (electron-donating or electron-withdrawing para-benzyl substituent), respectively. Trypanocidal hits in the micromolar range were identified against bloodstream forms of T. congolense [CPX (1); CPX amide derivatives 18: IC50 8.95 µM; SI 16.84; 22: IC50 5.42 µM; SI 25.2] and against T. brucei rhodesiense (CPX acid derivative 13: IC50 4.51 µM; SI 10.2), demonstrating more selectivity toward trypanosomes than mammalian cells. Hence, the trypanocidal hit compound 22 may be optimized by retaining the 4-methylpiperazine amide functional group (C-3) and the TZ moiety at position N-15 and introducing other electron-withdrawing ortho-, meta-, and/or para-substituents on the aryl ring in an effort to improve the pharmacokinetic properties and increase the trypanocidal activity. Graphical abstract Structure–activity relationships of ciprofloxacin-1,2,3-triazole hybrids were governed by the chemical functionality at C-3 and electronic effect.
... The cytotoxic activity of the anacardic acid mixture on mammalian cells was performed in the LLC-MK2 lineage (Rhesus monkey Kidney Epithelial Cells). The cells were cultured in sterile 96-well plates at a concentration of 10 5 cells/ml in DMEM medium at 10% FBS, penicillin (100 IU/ ml), and streptomycin (100 µg/l) at 37 °C in an atmosphere with 5% CO 2 for 24 h (Nwaka and Hudson 2006). ...
... The toxicity of substances on LLC-MK2 served as a basis for defining the concentrations used in vitro assays (Nwaka and Hudson 2006). ...
Article
The work presents an optimization of the extraction and purification processes of the anacardic acids mixture and its evaluation as a potential antichagasic drug. The purified mixture was obtained by chromatography techniques and characterized by 1H and 13C NMR. The cytotoxicity was evaluated in Rhesus monkey Kidney Epithelial Cells (LLC-MK2) after 24 h of exposure in concentrations between 400 and 6.25 μg/ml. The new procedure was efficient for the purification of the anacardic acid mixture and separating them, with a 31.6% yield, considering a 3-step process. The mixture presented cytotoxicity at concentrations of 400 and 200 μg/ml, with CC50/ 24 h of 286 μg/ml and IC50 of 12.9 μg/ml These are promising initial results, since the epimastigotes proliferate alone demonstrating a potential trypanocidal effect on Trypanosoma cruzi strain Y, higher than isolated anacardic acids. Results show of the anacardic acid mixture as a bioproduct with potential antichagasic, a without isolation steps necessary to obtain of the pure compounds and, therefore, more economically viable and sustainable.
... 7 Medicinal plants have been the source of several drugs including antimalarials such as quinine and artemisinin. 7,8 Several medicinal plants have been used for the treatment of malaria in Ethiopia 9 including those that experimentally showed to possess in-vivo antimalarial activity against P. berghei infected mice, such as Echnops kebericho, 10 Adhatoda schimperiana 11 and Vernonia amygdalina Del. 12 Croton dichogamus Pax is a shrub that grows in Ethiopia, Kenya, Somalia, Rwanda, Mozambique, Tanzania, Madagascar and Uganda. It has thornless branches and green upper-and silvery-bottom-colored leaves that become bright orange when dying. ...
Article
Full-text available
Background Malaria is causing high mortality and morbidity due to Plasmodium’s resistance to currently available anti-malarial drugs and mosquito’s resistance to insecticides. Thus, there is a critical need to search for novel anti-malarial drugs from natural sources. Therefore, this study investigated in vivo antimalarial activities of two Ethiopian medicinal plants, Croton dichogamus Pax and Ehretia cymosa Thonn, in Plasmodium berghei infected Swiss albino mice. Methods Soxhlet extraction method using 80% methanol as a solvent was used to prepare crude extracts of the two plants. Acute oral toxicity and 4-day suppressive in vivo antimalarial activity tests were performed on healthy female mice and P. berghei infected male mice, respectively. Antimalarial activity of the crude extracts at doses of 100, 200, and 400 mg/kg and the standard drug, chloroquine were used to assesse in Plasmodium berghei infected Swiss albino mice. Parasitemia level, packed cell volume, body weight, and rectal temperature of the mice were determined before infection (day 0) and after treatment (day 4). Survival time was determined by recording the date on which the mice died, considering the date of infection as day 0. The recorded data were analyzed using ANOVA and SPSS version 24. Results The result of the acute toxicity study revealed that the crude extracts were non-toxic at doses up to 2 g/kg. The extract of E. cymosa suppressed parasitemia level by 66.28, 63.44 and 63.14% at 400, 200, and 100mg/kg, levels while C. dichogamus extract suppressed parasitemia level by 45.29% at a dose of 400mg/kg. The remaining two dose levels of C.dichogamus extract suppressed parasitemia level by < 30%. Conclusion C. dichogamus and E. cymosa showed anti-plasmodial activities. E. cymosa exhibited a more pronounced anti-plasmodial effect than C. dichogamus. The activities of both plants observed in this study support their traditional use as antimalarial drugs. Further studies on these plants using solvent fractions are required to identify their active ingredients.
... In addition, the selectivity index (SI) found for the ethyl acetate fraction indicates greater toxicity for parasites than for macrophages. According to Nwaka and Hudson [41], the SI acceptable for new drug candidates should be close or greater than 20. Therefore, SI results are essential in the search for effective natural products for the treatment of leishmaniasis. ...
Article
Full-text available
Leishmaniases are infectious-parasitic diseases that impact public health around the world. Antileishmanial drugs presented toxicity and increase in parasitic resistance. Studies with natural products show an alternative to this effect, and several metabolites have demonstrated potential in the treatment of various diseases. Terminalia catappa is a plant species with promising pharmaceutical properties. The objective of this work was to evaluate the therapeutic potential of extracts and fractions of T. catappa on Leishmania amazonensis and investigate the immunomodulatory mechanisms associated with its action. In anti-Leishmania assays, the ethyl acetate fraction exhibited activity against promastigotes (IC50 86.07 ± 1.09 μg/mL) and low cytotoxicity (CC50 517.70 ± 1.68 μg/mL). The ethyl acetate fraction also inhibited the intracellular parasite (IC50 25.74 ± 1.08 μg/mL) with a selectivity index of 20.11. Treatment with T. catappa ethyl acetate fraction did not alter nitrite production by peritoneal macrophages stimulated with L. amazonensis, although there was a decrease in unstimulated macrophages treated at 50 μg/mL (p = 0.0048). The T. catappa ethyl acetate fraction at 100 μg/mL increased TNF-α levels (p = 0.0238) and downregulated HO-1 (p = 0.0030) and ferritin (p = 0.0002) gene expression in L. amazonensis-stimulated macrophages. Additionally, the total flavonoid and ellagic acid content for ethyl acetate fraction was 13.41 ± 1.86 mg QE/g and 79.25 mg/g, respectively. In conclusion, the T. catappa ethyl acetate fraction showed leishmanicidal activity against different forms of L. amazonensis and displayed immunomodulatory mechanisms, including TNF-α production and expression of pro and antioxidant genes.
... These include selecting and reviewing the projects, negotiating partnership agreements, managing the interfaces of drug R&D, allocating project portfolios, and integrating upstream and downstream interfaces [49]. According to the previous literature, managers, and coordinators may focus on developing organizational dynamic capabilities in R&D collaborative project portfolio management, which promotes NTD clinical trials and drug availability [22,[50][51][52]. ...
Article
Full-text available
Public-private partnerships (PPPs) for neglected tropical diseases (NTDs) are often studied as an organizational form that facilitates the management and control of the huge costs of drug research and development. Especially the later stages of drug development, including clinical trials, become very expensive. This present study investigates whether and how the type of PPPs influences the initiation and duration of NTD clinical trials. Using the ClinicalTrials.gov database, a dataset of 1175 NTD clinical studies that started between 2000 and 2021 is analyzed based on affiliation information and project duration. For the NTD clinical trials that resulted from PPPs, the collaborating types were determined and analyzed, including the public sector-, private sector-, governmental sector-, and nongovernmental organization-led collaborations. The determinants for the discontinuation of all stopped clinical trials were categorized into scientific-, funding-, political-, and logistic dimensions. The results reveal that public sector-led PPPs were the most common collaborative types, and logistic and scientific issues were the most frequent determinants of stopped clinical trials. Trial registration: ClinicalTrials.gov.
Article
Full-text available
Applications of the Drosophila melanogaster (D.m.) research model have an important means both for genetic investigations and for the study of metal toxicity, because D.m. has physiological mechanisms comparable to those in human organisms. In this research, the toxic effect of lead (Pb2+) and copper (Cu2+) on four D.m. genotypes—the wild genotype (Oregon-R, used as control) and three mutant genotypes (white, brown, and white-vestigial)—was compared. Five replicates were made to observe the development progress of monitorized genotypes exposed to five different concentrations 0, 0.50, 0.75, 1.00, and 2.00 mM of copper (CuSO4) and lead Pb(C2H3O2)2. Proliferation rates of larvae, pupae, and adults depend on genetic factors, metals used (copper or lead), and their concentrations. The white-vestigial mutant genotype showed the greatest sensitivity at IC50 concentration (inhibition of proliferation of more than 50% compared with the control sample) at doses of 1.00 mM for Cu2+ and 2.00 mM for Pb2+. In contrast, the control genotype (Oregon-R) showed only an inhibition IC50 concentration of 2.00 mM for Cu2+. The white-vestigial mutant genotype showed the greatest sensitivity at IC50 concentration (inhibition of proliferation of more than 50% compared with the control sample) at doses of 1.00 mM for Cu2+ and 2.00 mM for Pb2+. In contrast, the control genotype (Oregon-R) showed an inhibition at the IC50 concentration of 2.00 mM for Cu2+. The results conclude that (i) the dose influences the prolificacy rate in a directly proportional way, (ii) the comparative analyses between Cu2+ and Pb2+ revealed a more acute effect of Cu2+, and (iii) differentiated prolificacy values according to genotypes were recorded. Those reflect the importance of using D.m. as a research model in the comparative studies of the interactions between genetic factors and metal toxicity. Also, this study provides significant information on non-toxic maximum doses for organisms.
Article
Full-text available
African trypanosomiasis is a vector-borne disease of animals and humans in the tsetse fly belt of Africa. Trypanosoma congolense (“nagana”) is the most pathogenic trypanosome in livestock and causes high morbidity and mortality rates among cattle. In the absence of effective preventative vaccines, the management of trypanosomiasis relies on chemoprophylaxis and/or -therapy. However, the trypanocides in clinical use exhibit poor oral bioavailability and toxicity, and therapeutic failures occur because of resistant strains. Because nitrofurantoin displayed, in addition to its clinical use, promising antiparasitic activity, the current study was conducted to evaluate the in vitro trypanocidal activity and preliminary in vivo treatment efficacy of previously synthesized nitrofuranylazines. The trypanocidal activity of these nitrofuran derivatives varied among the evaluated trypanosome species; however, T. congolense strain IL3000 was more susceptible than other animal and human trypanosomes. The nitrofurylazines 4a (IC50 0.04 μM; SI > 7761) and 7a (IC50 0.03 μM; SI > 9542) as well as the nitrothienylazine 8b (IC50 0.04 μM; SI 232), with nanomolar IC50 values, were revealed as early antitrypanosomal leads. Although these derivatives showed strong trypanocidal activity in vitro, no in vivo treatment efficacy was observed in T. congolense IL3000 infected mice after both oral and intraperitoneal administration in a preliminary study. This was attributed to the poor solubility of the test compounds in the in vivo testing media. Indeed, a challenge in drug discovery is finding a balance between the physicochemical properties of a drug candidate, particularly lipophilicity and water solubility, and maintaining adequate potency to provide an effective dose. Hence, future chemical modifications may be required to generate lead-like to lead-like nitrofuranylazines that possess optimal physicochemical and pharmacokinetic properties while retaining in vitro and, ultimately, in vivo trypanocidal efficacy.
Article
Full-text available
: Leishmaniasis is a major public health problem, and the alarming spread of parasite resistance has increased the importance of discovering new therapeutic products. In the present study, the antileishmanial activity of the methanolic extract of the leaf latex obtained from the Ethiopian plant Vernonia brachycalyx O. H. (family Asteraceae) was evaluated by in vitro testing against Leishmania aethiopica and . : Antileishmanial activity test was carried out using the Alamar Blue assay on promastigotes and axenic cultured amastigotes of and clinical isolates, and cell viability was fluorometrically determined. Amphotericin B was used as a positive control, and 1% dimethyl sulfoxide (DMSO) and the media were employed as a negative control. Moreover, preliminary phytochemical analysis of the extracts was performed. : Results of the study indicated that the latex possesses good activity against both parasites, with IC values of6.82 ± 0.18 and 6.34 ± 0.20μg/ml against promastigotes and 3.53 ± 0.33 and 2.61 ± 0.907μg/ml against axenically cultured amastigotes of and , respectively. The latex demonstrated selectivity indices (SIs) of 15.27 and 16.42 against promastigotes and 29.50 and 39.90 against axenically cultivated amastigotes of and . While, amphotericin B demonstrated SIs of 7.91 and 8.23 against promastigotes and 7.45 and 7.73 against axenically cultured amastigotes of and , respectively. Phytochemical screening demonstrated that the latex contains flavonoids, tannins, cardiac glycosides, terpenoids, saponins, alkaloids, and steroids. : The findings of this investigation attest that the latex of V. brachycalyx possesses promising antileishmanial activity against and , warranting further investigations into the active constituents.
Article
Human African trypanosomosis (HAT) which is also known as sleeping sickness is caused by Trypanosoma brucei gambiense that is endemic in western and central Africa and T. b. rhodesiense that is endemic in eastern and southern Africa. Drugs used for treatment against HAT first stage have limited effectiveness, and the second stage drugs have been reported to be toxic, expensive, and have time-consuming administration, and parasitic resistance has developed against these drugs. The aim of this study was to evaluate the anti-trypanosomal activity of nitrofurantoin-triazole hybrids against T. b. gambiense and T. b. rhodesiense parasites in vitro. This study screened 19 synthesized nitrofurantoin-triazole (NFT) hybrids on two strains of human trypanosomes, and cytotoxicity was evaluated on Madin-Darby bovine kidney (MDBK) cells. The findings in this study showed that an increase in the chain length and the number of carbon atoms in some n-alkyl hybrids influenced the increase in anti-trypanosomal activity against T. b. gambiense and T. b. rhodesiense. The short-chain n-alkyl hybrids showed decreased activity compared to the long-chain n-alkyl hybrids, with increased activity against both T. b. gambiense and T. b. rhodesiense. Incorporation of additional electron-donating substituents in some NFT hybrids showed increased anti-trypanosomal activity than to electron-withdrawing substituents in NFT hybrids. All 19 NFT hybrids tested displayed better anti-trypanosomal activity against T. b. gambiense than T. b. rhodesiense. The NFT hybrid no. 16 was among the best performing hybrids against both T. b. gambiense (0.08 ± 0.04 μM) and T. b.rhodesiense (0.11 ± 0.06 μM), and its activity might be influenced by the introduction of fluorine in the para-position on the benzyl ring. Remarkably, the NFT hybrids in this study displayed weak to moderate cytotoxicity on MDBK cells. All of the NFT hybrids in this study had selectivity index values ranging from 18 to greater than 915, meaning that they were up to 10-100 times fold selective in their anti-trypanosomal activity. The synthesized NFT hybrids showed strong selectivity >10 to T. b. gambiense and T. b. rhodesiense, which indicates that they qualify from the initial selection criteria for potential hit drugs.
Article
Full-text available
Knowledge of the three-dimensional structures of protein targets now emerging from ge- nomic data has the potential to accelerate greatly drug discovery, but technical challenges and time constraints have traditionally limited their use primarily to lead optimization. Their application is now being extended beyond structure determination, into new ap- proaches for lead discovery (for review see Blundell et al.,2002). Virtual screening coupled with high throughput X-ray crystallography is focused on identifying one or more weakly binding small-molecule fragments from compound libraries consisting of hundreds of small- molecule fragments. The high-resolution definition of this binding interaction provides an information-rich starting point for medicinal chemistry. The use of high throughput X-ray crystallography does not end there, as it becomes a rapid technique to guide the elabora- tion of the fragments into larger molecular weight lead compounds.
Article
Full-text available
The movement of ideas and innovation from academia into the world of business has a long and fruitful history. Ironically, it might be argued that the recent pressure put on universities and basic research organisations to protect and exploit their intellectual property has, in many ways, created a less conducive environment to successful commercialisation than existed 30 years ago. This movement has been concurrent with the drift of the Pharmaceutical industry towards a more risk-averse R&D strategy in which it has increasingly concentrated its resources on a reductionist drug discovery process and later stage clinical development. In effect, these two strategies have created a discontinuity between academic scientific output and industry at a time when academia as a source of innovation is perhaps more important to industry than ever.
Chapter
Full-text available
A number of criteria must be met before an enzyme or receptor can be regarded as a suitable target for drug discovery and drug development. This article considers the various approaches to target discovery, critically examines the criteria for target selection and compares the strengths and weaknesses of target validation by chemical and genetic methods. Particular reference is made to the prospects of drug development against trypanothione reductase, a target common to diseases caused by species of trypanosomes and the leishmania
Article
Combinatorial chemistry and high-throughput screening (HTS) research programs are widely used in medicinal chemistry and agro-chemistry, in order to select new leads. But, due to the high cost associated to synthesizing and screening a very large number of compounds, there is an increasing need of efficient tools allowing to design and to classify large chemical libraries in order to get enhanced information content. This objective, defined as « data base mining » (DBM), can be achieved by analyzing molecular diversity in large databases with help of the most up-to-date methods based on Kohonen Self Organizing Maps, Fuzzy Logic techniques and Genetic Algorithm. In fact, the proposed methods allow to get a friendly representation of the compound distribution in the hyperspace derived from their molecular descriptors. These derived models of virtual high-throughput screening (v-HTS), linking the structures of the compounds with their biological properties, are suitable to predict activity values for new untested molecules.