Drug Discovery: Innovations, Challenges and the Future of Medicine
eBook
Published: April 2, 2025

Credit: Technology Networks
The landscape of drug discovery is evolving rapidly, driven by breakthroughs in AI, high-throughput screening and precision medicine. As researchers work to develop safer, more effective therapies, they face challenges such as optimizing screening techniques, integrating complex in vitro models and navigating regulatory pathways. Addressing these obstacles is key to accelerating the development of life-changing treatments.
This eBook explores the latest advancements in drug discovery, from AI-driven precision medicine to the promise and risks of new weight loss drugs.
Download this eBook to discover:
- How AI is revolutionizing precision medicine and accelerating drug development
- Advances in high-throughput screening and complex in vitro models for better preclinical research
- Challenges and breakthroughs in developing next-generation therapeutics
Credit: iStock
DRUG
DISCOVERY:
Innovations, Challenges and the Future of Medicine
SPONSORED BY
How Is AI Speeding
Pharma’s Journey Toward
Precision Medicine?
Navigating the
Complexities of Impurities
in Pharmaceuticals
The Promise and Perils of
Weight Loss Drugs
CONTENTS
04
High-Throughput Screening:
Advances, Applications and
Combined Approaches
10
How Is AI Speeding Pharma’s
Journey Toward Precision Medicine?
13
Integrating Complex In Vitro Models
Into the Regulatory Pipeline
16
The Promise and Perils of Weight
Loss Drugs
21
How Broadly Neutralizing Antibodies
Are Driving Next-Gen Vaccines
29
Navigating the Complexities of
Impurities in Pharmaceuticals
35
The Evolving Treatment Landscape
for Alzheimer’s
39
Unveiling the Progress
and Challenges in the Fight
Against HIV
43
Contributors
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 3
TECHNOLOGYNETWORKS.COM
FOREWORD
The landscape of drug discovery is undergoing a remarkable transformation, driven
by scientific breakthroughs, technological advancements and a growing demand for
innovative treatments. With the rise of conditions such as Alzheimer's disease, HIV and
obesity, researchers are accelerating efforts to develop therapies that are not only effective
but also safer, more targeted and accessible to those in need.
The journey from identifying a promising drug target to delivering a life-changing
treatment is a complex and multidisciplinary endeavor. High-throughput screening, AI and
in vitro models are revolutionizing early-stage research, while novel biologics and precision
medicine approaches are shaping the future of patient care. Yet, challenges remain, from
navigating regulatory pathways to addressing the risks and limitations of new therapies.
This eBook brings together expert insights on the latest trends in drug discovery, from the
potential of broadly neutralizing antibodies in vaccine development to the promise and
perils of weight loss drugs. It also explores how AI is optimizing pharmaceutical research
and how emerging therapies for neurodegenerative diseases and infectious conditions are
redefining treatment possibilities.
The Technology Networks editorial team
4 DRUG DISCOVERY
High-throughput screening (HTS) is a drug discovery
approach that involves screening vast libraries of small
molecules to identify “hits” with therapeutic potential.
As a result of exciting past scientific breakthroughs,
HTS approaches have been miniaturized, standardized
and automated. This, alongside more recent advances
in cell culture and data analysis, has led to faster
more reproducible screening with targets that more
accurately reflect in vivo physiology.
In this article, we highlight various HTS strategies being
used to interrogate large libraries of compounds and
hear from researchers working to further streamline
specific approaches to increase speed and improve
quality and accuracy.
Let’s start by considering different approaches to HTS.
In vitro biochemical assays
A purified target (e.g., enzyme, receptor or hormone)
is screened against a compound library. The readout is
typically achieved using optical detection methods such
as absorbance, fluorescence or luminescence.
Considerations: These assays can identify hits
and quantify binding affinity but cannot reproduce a
phenotypic or functional response. They are unable to
provide information on cellular context.
Cell-based assays
Increasingly cell-based assays are used to provide a
phenotypic readout, e.g., a change in signaling measured
by a reporter gene or an effect on cell growth.
High-Throughput Screening:
Advances, Applications and
Combined Approaches
Joanna Owens, PhD
Credit: iStock
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 5
Considerations: They provide information
on how compounds affect function in a cellular
environment. However, drug compounds can
sometimes fail to perform in this setting if 2D
monolayer cultures are used, as they lack the native
tissue microenvironment1
that influences cell
behavior and function. These assays do not provide
information on how the compound is exerting its
effect – i.e., what the cellular target is, or whether it is
hitting the desired target or not.
RNA interference (RNAi) screening
RNAi screening can be used for high-throughput
analysis of gene function on a genome-wide scale.2
Considerations: This approach provides
insights into functional networks in cells that could
be targeted in drug discovery. It can be combined
with high-content imaging.
These approaches are key in the early stages of drug
discovery, offering researchers a diverse set of tools
that can be used in combination depending on the
specific requirements of the project and starting
materials (i.e., libraries, X-ray structures of targets,
available disease models). They each have their
own merits and limitations, but all have universally
benefitted from several key technological advances
(Table 1).
Despite these advances, researchers are still working
to optimize conventional HTS approaches and
develop platforms and software to combine different
methods. In the rest of this article, we’ll explore some
examples of innovation in screening methods.
Navigating challenges in HTS
The practical logistics of screening compound
libraries that can contain as many as three to four
million compounds remains a major challenge
in HTS.
Automation
HTS was made commercially feasible by the development of automated liquid handling workflows that
make it time and cost-efficient to process thousands of parallel samples. Today’s automated workflows
require limited supervision or manual intervention and improve screening quality and reproducibility.3,4
Microfluidic
systems
Miniaturized microfluidics devices make it possible to screen compounds in high-density arrays, using
small volumes of purified target and compound.5
This has increased the rate of screening and reduced the
amount of reagents required. Microfluidic channels also provide a basis for building different connected
compartments to mimic the interconnected nature of the human body.
3-dimensional
(3D) cell
culture
3D cell culture has enabled tissue spheroids or miniature organs (organoids) to be derived from cell lines,
primary cells or stem cells, which further enhance drug screening by better recapitulating the human
physiological environment compared to 2D cell lines or animal models.6
High content
screening
The addition of automated and/or live cell imaging methods alongside quantitative assays now makes
it possible to capture more detailed phenotypic outputs in high throughput, by taking multiple views
of a well under different light wavelengths and by labeling different targets within the individual cells.7
This can provide insights into cellular expression changes, or morphology changes at the organelle or
single-cell level. Multiple properties of individual cells can also be studied simultaneously.
HCS assays are typically conducted using 2D monolayer cultures to achieve higher throughput.
However, 3D cell culture systems (e.g., spheroids, organoids) can also be used.
Gene editing
Precise gene knockout using tools (e.g., CRISPR-Cas9) offer the potential to screen cells for targets that
can then be pursued therapeutically.2,8
As CRISPR guides are very easy to synthesize, you can design
customized libraries to knock out every gene in the genome or a particular set of genes, making the
approach amenable to HTS.
DNA-encoded
libraries
Advances in genetic sequencing allow compounds to be DNA barcoded, so that large compound libraries
can be screened within a single microplate well, accelerating hit identification and optimization.9,10
TABLE 1: Key advances in assay technologies for HTS
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 6
Physically screening each of these compounds against a
single target in replicate is a time-consuming and laborintensive undertaking, even with automation. Using
more focused libraries for well-characterized targets
is one option, but another route is to use mixtures of
compounds against a single target. To achieve this,
compounds can be attached to solid substrates such as
beads to allow the purification of “hits”, but this is still
not practical for the largest libraries. This has led to
the introduction of DNA barcoding to generate DNAencoded libraries.
Dr. Basilius Sauter is a postdoctoral researcher in Prof.
Dennis Gillingham’s lab at the University of Basel.
Sauter specializes in the areas of DNA-encoded libraries
and DNA-modifying drugs.
“Although the concept of DNA barcoding is not new, it
was only with advances in next-generation sequencing
that DNA-encoded libraries became viable for larger
library sizes,” he said. “Since then, the number of
different chemistries available has rapidly increased,
allowing the generation of more diverse DNA-encoded
libraries covering a greater chemical space.”
Optimizing DNA-encoded libraries
for HTS
One limitation of DNA-encoded libraries is that, unlike
conventional HTS, the assay can only detect binders
or non-binders, and you need to be able to separate
these. The conventional method is to use an affinity
purification step, where you attach your target protein
to a solid phase and wash the non-binders away. But
this has the disadvantage that you can lose compounds
that bind weakly which, with further optimization, could
have been potential drug candidates.
To circumvent this, Sauter and colleagues have recently
developed a new technique using an enzyme called
terminal deoxynucleotidyl transferase (TDT).
“TDT is a very special polymerase because it can
synthesize DNA without a template,” explained Sauter.
“We made a fusion of TDT with our target protein
of interest, such that when a DNA-encoded library
member binds to the protein of interest, TDT is in
close proximity. Then, when we incubate with only
dATP, the TDT preferentially extends the 3’ terminus
of the DNA barcode on the hit molecule and makes a
long poly dA tail.”
This makes it possible to select hits in solution, which
is an advantage when working with drug targets that
start to denature or behave differently when on a solid
phase for the affinity purification step. The polyA tail
is also proportionate to the binding affinity of the
compound, because it relies on the proximity of the
drug to the target.11
Perhaps the most exciting and surprising result,
however, was that the TDT method not only identified
all the binders that were determined using the standard
affinity purification step, but also found additional weak
binders that had evaded previous screens.
“When we tested it, it turned out to be a very weak
inhibitor of our target molecule,” said Sauter.” One of
the issues with DNA-encoded libraries is they often
identify hits that are strong binders but it’s not possible
to take them forward because they aren’t amenable to
lead optimization by medicinal chemists. We want to
DNA-encoded libraries
A short piece of DNA called a “headpiece”
is split into portions and modified with
different small molecules.9
A set of unique
DNA barcodes is added to each portion of
DNA, with each barcode corresponding to a
specific small molecule. The resulting DNAchemical conjugates are then pooled, split,
chemically diversified and encoded with
another set of DNA barcodes. This splitmodify-encode-pool is usually repeated
2-4 times and allows the generation of
many thousands of DNA-encoded small
molecules that can be screened against a
protein target.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 7
build on the work we’ve done so far to find a screening
assay that is sensitive for these weak binders, especially
for difficult targets where having a weak interaction is
better than none.”
Combining different screening
approaches and considering
alternatives
Selecting a specific HTS approach often involves a
compromise between using a method that provides
fundamental information about drug binding
characteristics to determine (e.g., specificity and
sensitivity) and gaining functional insights about
whether a drug’s action is achieving the desired
phenotypic results. To solve this problem, a team led by
Pavel Levkin at the Karlsruhe Institute of Technology
(KIT) in Germany has developed a combined highcontent, high-throughput platform that enables unified
on-chip chemical synthesis, characterization and
biological screening.12
The chip was developed as part of a European Research
Council Proof-of-Concept project to develop more
affordable and faster cell screening experiments
in diagnostics and personalized medicine. Levkin
and colleagues developed dendrimer-based surface
patterning that enables the generation of high-density
nanodroplet arrays. Each of the 50,000 droplets on a
plate function as an individual nanovessel. With these
vessels it is possible to conduct combinatorial chemical
synthesis and then run a range of analytical assays –
from on-chip detection using methods such as infrared
spectroscopy or matrix-assisted laser desorption/
ionization mass spectrometry, to high-content cellbased screening. By bringing together the traditionally
separate steps of high-throughput chemical synthesis,
reaction monitoring, compound characterization and
biological screening, the technique offers the potential
to unify the different approaches used in early-stage
drug discovery.
Another significant challenge in HTS is managing
large compound libraries and this, together with more
challenging targets, has led drug discovery researchers
to adopt fragment-based drug discovery (FBDD) as an
alternative to HTS. In FBDD you initially screen chemical
“fragments” to identify those with promising functionality,
and then build on the fragment “hit” either by adding
other chemistries or combining several fragments to
create a more potent lead compound. Pioneered in
the late 1990s by Professor Steve Fesik as “SAR by
NMR”, it now constitutes an essential toolkit for the
biopharma industry.13 However, despite this sensitivity,
NMR screening has other limitations – researchers
screening libraries of thousands of compounds quickly
find themselves with a data analysis bottleneck, having to
visually inspect thousands of spectra.
To address this problem, Geerten Vuister, professor
of structural biology at Leicester University and chair
of the Collaborative Computational Project for NMR
and his team have developed a computational pipeline
of analysis tools to support NMR-based screening.14
It started with tackling a fundamental challenge – the
complexity of a fragment library.
“Even relatively small fragment libraries contain several
hundreds of compounds and each has its own NMR
spectrum, so when you combine these molecules in
a single mixture, you might get overlapping spectral
regions and it becomes difficult to distinguish between
them,” explained Dr. Luca Mureddu, who developed the
pipeline. “Moreover, factors such as experimental error,
degradation of library compounds and varying buffer
requirements for different protein targets all contribute
to considerable variability between spectra.”
This led them to develop tools and workflows that
adjust for experimental factors and post-processing
that deals with the variability of the data improving
the hit rate identification. After running the algorithms
of the workflow, which can take as little as seconds
for a small library, to around ten minutes for a library
of a few thousand compounds, users receive a set of
scores indicating a level of confidence about whether a
compound is a hit or not.
“The most important premise is that we don’t want to
replace the researcher,” said Mureddu. “So, we don’t
provide a binary yes/no answer to whether something is a
hit.” Instead, based on a combination of scores, users can
not only identify reliable positive or negative hits, but also
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 8
easily inspect the data for compounds that fall into the
“uncertain” zone, potentially prioritizing these for followup screening using another established method.
“Drug companies tend to use a variety of screening
approaches, so it was important to develop a pipeline
that users can customize to their own specific screening
needs,” highlighted Dr. Vicky Higman, a Research
Fellow in Vuister’s team specializing in NMR.
“We’ve already seen that our collaborators are increasing
the size of their screening libraries, because they can now
analyze data much more quickly and reliably.” •
ABOUT THE INTERVIEWEES
Dr. Basilius Sauter is a postdoctoral researcher in Professor
Dennis Gillingham’s lab at the University of Basel. Sauter
specializes in the areas of DNA-encoded libraries and DNAmodifying drugs.
Dr. Luca Mureddu is a postdoctoral researcher at the University
of Leicester and part of the Collaborative Computational Project
for NMR working group. Mureddu focuses on focus on developing
software tools for fragment based-screening to enable and
accelerate high quality and reproducible research.
REFERENCES
1. Wei F, Wang S, Gou X. A review for cell-based screening
methods in drug discovery. Biophys Rep. 2021;7(6):504-
516. doi: 10.52601/bpr.2021.210042
2. Taylor J, Woodcock S. A perspective on the future
of high-throughput RNAi screening: Will CRISPR
cut out the competition or can RNAi help guide
the way? J Biomol Screen. 2015;20(8):1040-1051.
doi:10.1177/1087057115590069
3. Hansel CS, Plant DL, Holdgate GA, Collier MJ, Plant H.
Advancing automation in high-throughput screening:
Modular unguarded systems enable adaptable drug
discovery. Drug Discov Today. 2022;27(8):2051-2056. doi:
10.1016/j.drudis.2022.03.010
4. Green CP, Spencer PA, Sarda S. Advancing automation
in compound management: a novel industrial process
underpinning drug discovery. Drug Discov Today
2021;26(1):5-9. doi: 10.1016/j.drudis.2020.09.032
5. De Stefano P, Bianchi E, Dubini G. The impact of
microfluidics in high-throughput drug-screening
applications. Biomicrofluidics. 2022;16(3):031501. doi:
10.1063/5.0087294
6. Wang Y, Jeon H. 3D cell cultures toward quantitative
high-throughput drug screening. Trends Pharmacol Sci.
2022;43(7):569-581. doi: 10.1016/j.tips.2022.03.014
7. Blay V, Tolani B, Ho SP, Arkin MR. High-throughput
screening: today's biochemical and cell-based
approaches. Drug Discov Today. 2020;25(10):1807-1821.
doi: 10.1016/j.drudis.2020.07.024
8. Shalem O, Sanjana NE, Zhang F. High-throughput
functional genomics using CRISPR-Cas9. Nat Rev Genet.
2015;16(5):299-311. doi: 10.1038/nrg3899
9. Satz AL. What do you get from dna-encoded Libraries?.
ACS Med Chem Lett. 2018;9(5):408-410. doi: 10.1021/
acsmedchemlett.8b00128
10. Reddavide FV, Cui M, Lin W, et al. Second generation
DNA-encoded dynamic combinatorial chemical libraries.
Chem Commun (Camb). 2019;55(26):3753-3756. doi:
10.1039/c9cc01429b
11. Schneider LA, Sauter B, Dagher K, Gillingham D.
Recording binding information directly into DNA-encoded
libraries using terminal deoxynucleotidyl transferase.
J Am Chem Soc. 2023;145(38):20874-20882. doi: 10.1021/
jacs.3c05961
12. Benz M, Asperger A, Hamester M, et al. A combined highthroughput and high-content platform for unified on-chip
synthesis, characterization and biological screening. Nat
Commun. 2020;11:5391. doi: 10.1038/s41467-020-19040-0
13. Shuker SB, Hajduk PJ, Meadows RP, Fesik SW.
Discovering high-affinity ligands for proteins: SAR by
NMR. Science. 1996;274(5292):1531-1534. doi: 10.1126/
science.274.5292.1531
14. Mureddu LG, Ragan TJ, Brooksbank EJ, Vuister GW.
CcpNmr AnalysisScreen, a new software programme with
dedicated automated analysis tools for fragment-based
drug discovery by NMR. J Biomol NMR. 2020;74(10-11):565-
577. doi: 10.1007/s10858-020-00321-1
The most revolutionary serological pipet controller
integra-biosciences.com
PIPETBOY
GENIUS
Brings color into your lab!
Overfill protection
Aliquot faster, aspirate safer, pipet smarter.
Lightning-fast,
one-button
repeat dispensing
of aliquots
10 DRUG DISCOVERY
While generative artificial intelligence (AI) burst into
the public consciousness a little more than two years
ago with the release of ChatGPT, AI and machine
learning (ML) have been part of drug research and
development for far longer. AI and ML are now
being applied to every stage of biopharmaceutical
R&D including discovery, compound development,
trial design, regulatory submission, supply-chain
optimization and postmarket surveillance.
In the last two years, several published, peer-reviewed
articles have referred to precision medicine as
“futuristic” – a state that has not yet been fully realized
– but drug discovery is rapidly becoming more patientcentered with each new application of AI.
Although ChatGPT and other generative AI engines
built into smartphones, Internet search engines and
social media platforms have captured public attention,
generative AI is only one flavor of the technology.
The pharma industry has been using predictive
modeling for years. Investigators are turning to
increasingly advanced algorithms to identify patient
subgroups, forecast mechanisms of action, predict
differential drug responses including toxicity and
even adjust dosing strategies.
How Is AI Speeding
Pharma’s Journey Toward
Precision Medicine?
Neil Versel
Credit: iStock
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 11
Popular use cases and
therapeutic areas
In a 2023 report, the UK-based Wellcome Trust
identified five key families of use cases for AI in
drug discovery:
• Drug target identification and validation
• Small-molecule design and optimization
by pinpointing “hit-like or lead-like small
molecule compounds”
• Design and optimization of vaccines, particularly
mRNA vaccines
• Design and optimization of antibody structures
and properties
• Evaluation of safety and toxicity of promising
compounds
According to the report, more than 80% of published
articles on AI-enabled drug discovery in the preceding
five years were related to understanding disease, target
discovery and optimization of small-molecule compounds.
Of note, the organization said there has been a dearth of
publicly available data on safety and toxicity to train AI
models. Interviews with experts in the field included in
the report mentioned the challenges of predicting safety
and toxicity based on in silico data without sufficient
supporting clinical validation.
The Wellcome report also cited a 2021 Nature article
describing how AlphaFold, an AI/ML algorithm from
Google sister company DeepMind, predicted the
three-dimensional structure of human proteins with
“atomic” accuracy.
About 70% of private-sector investments in AI for
drug discovery between 2018 and 2022 were in the
“commercially tractable” therapeutic areas of oncology,
neurology and COVID-19, Wellcome added.
As noted in the American Journal of Managed Care
(AJMC), numerous drug companies have already
incorporated AI into drug R&D processes, particularly
to improve target identification, molecule discovery and
patient recruitment for trials.
“The integration of AI into the drug discovery process
offers immense potential for accelerating drug
development, reducing costs, and improving patient
outcomes,” the October 2024 article concluded.
“However, the successful implementation of AI requires
addressing knowledge gaps, ensuring data quality, and
navigating regulatory challenges.”
At least one major drug company has said that it is
looking at AI for developing precision therapeutics in
oncology, immunology and neuroscience, all popular
therapeutic areas for AI application among Big Pharma.
However, the Wellcome report cited COVID-19
response as a shining example of the power of this
evolving technology.
In the early days of the pandemic in March 2020, AI was
used to research monoclonal antibodies derived from
convalescent plasma of COVID-19 patients. About 2,000
potential candidates were quickly narrowed down to
24, and the most promising compound, bamlanivimab,
entered into clinical trials within three months.
The U.S. Food and Drug Administration (FDA) granted
Emergency Use Authorization to bamlanivimab in
November 2020, just eight months after research
commenced. Though the FDA revoked the authorization
in April 2021 after subsequent SARS-CoV-2 variants
proved more resistant to the therapy, this episode
highlighted just how rapidly drug-makers could develop
narrowly targeted treatments.
Reality and promise
AI analyzes large genomic and multiomic datasets
faster than ever before to help target therapies.
A study published in Science in September 2023
explained how bioinformaticians have been able to
build upon each other’s work to extend the capabilities
of popular AI algorithms including AlphaFold to
improve prediction of pathogenicity of missense
variants in the human proteome.
A May 2024 review article published in the journal
Fundamental Research suggested that AI, in the form of
unsupervised machine learning, is a more efficient way
of generating patient clusters and phenotype candidates
than human training of datasets in the development
of gene therapies. AI is also useful for optimizing drug
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 12
dosing based on individuals’ genetic profiles, the
international team of authors wrote.
A July 2023 article in Pharmaceutics noted that an AI
technique called clustering – which groups similar
datapoints to find subgroups of patient data, gene
expression profiles, chemical structures and other
pertinent information – is useful for identifying drug
targets and stratifying patients. Other AI algorithms can
sift through complex stores of data in search of anomalies.
“AI is being utilized to advance precision medicine
approaches. By analyzing patient data, including
genomics, proteomics, and clinical records, AI
algorithms can identify patient subgroups, predict
treatment responses, and assist in personalized
treatment decision-making. AI also contributes to the
development of biomarkers for disease diagnosis and
prognosis,” the authors, academic researchers from
India and Northern Ireland, wrote.
“AI might revolutionize the pharmaceutical industry
in the future to accelerate drug discovery and drug
development,” they continued, offering a clear caveat
with their word choices. “AI-enabled precise medicine
could categorize patients, predict therapy responses,
and customize medicines by analyzing genomes,
proteomes, and clinical records.”
They saw promise in what they called “virtual screening”
to accelerate identification of lead compounds by
selecting therapeutic candidates with the necessary
characteristics from massive chemical databases.
“Scientists may create innovative compounds with
target-binding characteristics using deep learning and
generative models, improving medication effectiveness
and lowering adverse effects,” the Pharmaceutics
article said. “AI improves clinical trial design, patient
selection, and recruitment. AI algorithms will use
electronic health records, biomarkers, and genetic
profiles to find appropriate patients, lower trial costs,
and speed up approval.”
But, as the AJMC article stated, AI performance is only
as good as the data the algorithm is trained on — the old
“garbage in, garbage out” maxim from computer science.
One potential tripwire is data bias. Writing in NPJ
Digital Medicine, investigators from the Berlin Institute
of Health at Charité and Harvard Medical School
noted that AI data is often trained on datasets that
have under-represent women, people of color, lowincome groups and other historically disadvantaged
demographics. “For example, an AI algorithm used
for predicting future risk of breast cancer may suffer
from a performance gap wherein black patients are more
likely to be assigned as ‘low risk’ incorrectly,” they said.
Regulation
Regulation rarely keeps up with technological
advancements, and AI is no different in this regard.
While enabling legislation might be behind the times,
regulators are at least attempting to stay ahead of the
curve by offering guidance to constituents including
drug developers.
For example, the European Medicines Agency
(EMA) recently spent more than a year developing a
“reflection paper” offering guidance on using AI/ML
in pharmaceutical discovery, development, approval
submissions, manufacturing and outcomes assessment.
This guidance document spells out how existing
European Union and national laws cover AI/ML across
pharma lifecycles, including when technologies might be
treated as medical devices.
The EMA pointed out the importance of paying
attention to data quality and relevance in training
algorithms. ML developers should take a “humancentric approach” to design be able to explain their
methodologies to allow the agency to review and
monitor “black box” models, the paper said.
“This requires not only that active measures are
taken during data collection and modelling … but also
that both user and patient reported outcome and
experience measures are included in the evaluation
of AI/ML tools when they interface with an individual
user or patient,” the EMA explained. •
13 DRUG DISCOVERY
Complex in vitro models (CIVMs), such as
microphysiological systems (MPS), organoids,
spheroids and organ/tissue-on-a-chip, present an
opportunity to enhance the efficiency and accuracy of
drug discovery and development in an ethically manner.
The Critical Path Institute (C-Path), a nonprofit
organization created to improve drug development, has
been working to facilitate the adoption of MPS as drug
development tools (DDT) in regulatory science.
At this year’s Society for Laboratory Automation
and Screening (SLAS) annual meeting, Dr. Graham
Marsh, scientific director at C-Path, presented the
organization’s framework to support a US Food and
Drug Administration (FDA) qualification of CIVMs
for use in regulatory submissions.
Technology Networks had the pleasure of speaking with
Marsh to discuss how the framework was developed
and how automation and AI can increase the confidence
in CIVMs.
Q: What is C-Path, and what are its
core aims?
A: C-Path's mission is to lead collaborations that
advance better treatments for people worldwide.
Globally recognized as a pioneer in accelerating drug
development, C-Path has established numerous
international consortia, programs and initiatives that
currently include more than 1,600 scientists and
representatives from government and regulatory
agencies, academia, patient organizations, disease
foundations and pharmaceutical and biotech companies.
Integrating Complex In
Vitro Models Into the
Regulatory Pipeline
Molly Coddington
Credit: iStock
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 14
Uniquely different to a contract research organization
or service provider, our neutral collaborations offer
a unique front-row seat to the dialogue on the latest
in regulatory science from FDA and the Europeans
Medicines Agency.
These collaborations allow for information and data
sharing, which serves as the foundation for C-Path to
spearhead the transformation of such information and
data into actionable solutions that address specific
unmet needs in the drug development process. Such
solutions can include data resources, biomarkers,
clinical outcome assessment tools, clinical trial
simulators and other quantitative tools.
Q: Can you discuss how CIVMs
could supplement or even replace
existing models?
A: Regulatory agencies rely on models for demonstrating drug efficacy and safety. These models have
gone through rigorous characterization to ensure
confidence in the data that they produce. The stance
of regulatory bodies – and I think rightly so – is to be
conservative and cautious to choose the in vitro and
in vivo data from models that has been qualified and
routinely used when evaluating new drugs.
At the same time, scientists are rapidly developing
new methods that promise to better represent human
biology. MPS bring multiple cell types together with
relevant cell–cell interactions and shear forces, and
more faithfully reproduce the biology observed in vivo.
We have been working to bridge these two camps and
enable the robust characterization of new models so
that they can be integrated into the regulatory pipeline.
There is a huge opportunity for these systems to
supplement the data that are currently being generated
in animals with more human-relevant data. This is
particularly important for biologics and gene therapies
where the modalities are incredibly human specific.
We are looking for specific contexts of use for these
tools where they have a significant value add/or
demonstrated improvement over the current standard
assay. We believe that’s where we should focus to
qualify potential drug development tools.
Q: What is the stance of regulatory
bodies on the use of MPS in drug
discovery?
A: The FDA is willing to accept any data that the
applicant wants to include in their submission, and, to
date, some drugs have moved into clinical trials on the
weight of evidence provided by MPS. A nice example
is the Sanofi/Hesperos case, where the MPS data from
their myelination chip were used to show efficacy and
advance a therapeutic.
The FDA has also formalized the Innovative Science
and Technology Approaches for New Drugs
(ISTAND) pathway for qualifying MPS models as drug
development tools.
There are a number of reasons why a developer might
choose to qualify their model through this pathway,
but I hope that the work that we are doing will make it
easier for developers to create validation packages for
models that are being included in submissions, and find
appropriate contexts of use and drafting qualification
plans if they want to move their model into the drug
development tool pipeline.
The impression that I get from talking to some
colleagues in regulatory agencies is that they are excited
by the possibility that these tools will improve patient
safety, but we need to make sure they are properly
characterized to ensure their appropriate use.
These tools and
solutions help de-risk
decision making in
the development and
regulatory review
process of novel
medical products.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 15
Q: At SLAS 2025, your talk discussed
a novel framework to support FDA
qualification of CIVMs for use in
regulatory submissions. Can you tell
us about this framework, how it was
developed and its current status of use?
A: Our group, the Predictive Safety Testing
Consortium (PSTC), was the recipient of a Broad
Agency Announcement project contract from the FDA.
The project contract was to fund work with the team in
the Division of Applied Regulatory Science to perform
a landscape analysis of the regulatory readiness of
commercially available MPS models and to host a series
of public workshops to bring together stakeholders
from regulatory agencies, 3D tissue model developers,
academics and pharmaceutical industry scientists.
The goal of these public meetings has been to open lines
of dialogue between the groups, identify areas of unmet
regulatory need and highlight gaps in existing models
where CIVMs would improve regulatory evaluation
of drugs, whether that’s by improving the ability of the
models to predict drug efficacy in a disease model or
demonstrating drug safety for moving a new molecule
into the clinic.
We’ve taken the output from the first meeting and
written a whitepaper on the pathway to use CIVM for
regulatory applications. The output from the second
workshop is being drafted as a manuscript and updated
to the existing whitepaper document.
Q: What role can automation and AI
play in advancing the role of MPS in
drug discovery and development?
A: The benefit that automation can provide is
increasing the reproducibility of MPS assays and
thus increasing the confidence in the models. A key
component of validating MPS models for regulatory
assessment is building confidence that the data they
produce is robust and reproducible. The tools being
developed for laboratory automation and liquid
handling can eliminate a key source of variability in
these systems.
Combining AI/ML models with MPS models has great
potential to produce datasets with increased depth and
predictivity, as the algorithms promise to find higher
order trends in the data. Computer vision systems and
automated analysis tools will enable faster and more
thorough processing of large datasets to enable higher
throughput screens and the generation of larger and
more reproducible and relevant datasets. •
Dr. Graham Marsh was speaking with Molly Coddington, Senior
Writer and Newsroom Team Lead for Technology Networks.
ABOUT THE INTERVIEWEES
Dr. Graham Marsh is a scientific director in the Critical Path
Institute's Predictive Safety Testing Consortium. He is working
to enable the use of complex in vitro models for regulatory
applications by establishing evidentiary considerations for their
qualification as drug development tools.
What is the ISTAND Pilot
Program?
ISTAND, which launched in 2020, is a
program that supports the development
of new DDTs to be used in regulatory
applications for novel medical products.
Examples of submissions that might be
considered for ISTAND include the use
of MPS, AI-based algorithms or the use of
novel digital health technologies, such as
wearables, for patient assessment.
16 DRUG DISCOVERY
Though originally approved for treating type 2 diabetes,
semaglutide (Ozempic™ and Wegovy™) and tirzepatide
(Mounjaro™) have garnered increased attention over
the last few years due to their weight loss effects.
These drugs belong to a class known as glucagon-like
peptide 1 receptor agonists (GLP-1RAs).
This article will explore the benefits and risks of
GLP-1ARs, discussing their future implications and
potential alternatives.
GLP-1RAs – successes in diabetes
and beyond
Before their use as a weight loss intervention, GLP1RAs were trialed and approved for their ability to
reduce glucose levels effectively in type 2 diabetes.
The first GLP-1RA to be approved by the US Food
and Drug Administration (FDA), for type 2 diabetes,
exenatide, required twice daily injections. In 2019,
semaglutide became the first oral medication of this class
to be approved by the FDA. This oral form (Rybelsus™)
must be taken once daily. The injectable form (Ozempic
and Wegovy) is administered once weekly.
The Promise and Perils of
Weight Loss Drugs
Kate Robinson
Credit: iStock
How do GLP-1RAs work?
Glucagon-like peptide 1 (GLP-1) is a
hormone that triggers the release of
insulin (lowering the amount of glucose
in the blood), blocks the secretion of
glucagon (which raises blood sugar levels),
slows the emptying of the stomach and
increases satiety. GLP-1 agonist drugs
mimic this hormone.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 17
As the use of GLP-1RAs for diabetes management
has increased, the discovery of their impact on
weight loss has sparked interest in their potential
for obesity treatment.
A study published in the New England Journal of
Medicine found that adults with obesity had a mean
weight loss of 14.9% with semaglutide and lifestyle
intervention. This exceeded the loss with placebo plus
lifestyle intervention by 12.4 percentage points.
Tirzepatide, a dual-acting GLP-1/glucose-dependent
insulinotropic polypeptide (GIP) agonist, has shown
greater glucose and weight lowering effects than
semaglutide. The drug is also effective at reducing
blood pressure, waist circumference, body weight and
liver fat content.
“GLP-1RAs have shown impressive clinical benefits in
multiple trials. They can lead to an average weight loss
of 15%–20%, improve cardiometabolic risk factors and
prevent secondary cardiovascular events. They also
slow the progression of kidney disease and reduce the
severity of obesity-related conditions, like obstructive
sleep apnea,” said Dr. David D. Kim, assistant professor of
medicine and public health at the University of Chicago.
GLP-1RAs could also provide benefits outside of
diabetes and obesity management. A recent study,
published in Nature Medicine, found an association
between GLP1-RAs and benefits to neurological
and behavioral health, with reduced risks of seizures
and addiction to alcohol, cannabis, stimulants and
opioids. Participants taking the drugs also experienced
decreased risks of suicidal ideation, self-harm, bulimia
and psychotic disorders such as schizophrenia.
Further studies have shown potential benefits of GLP1RAs in fatty liver disease, chronic kidney disease and
heart disease.
Could GLP-1Ras exacerbate
eating disorders?
The success of GLP-1RAs has led to an increase in
demand and supply shortages, leading to a rise in
counterfeit versions of these products being sold.
These products can contain any number of ingredients,
potentially leading to unknown adverse events or
accidental overdoses.
Legitimate GLP-1RAs can cause nausea, vomiting,
diarrhea, dizziness, mild heart rate increase, infections,
headaches and indigestion. Rare side effects include
pancreatitis, thyroid cancer, kidney injury and
worsening diabetes-related retinopathy.
One potential area for concern, according to Dr.
Riccardo Dalle Grave, director of the Department
of Eating and Weight Disorders at Villa Garda Hospital,
Italy, is that these drugs could potentially contribute to
the onset or exacerbation of eating disorder symptoms.
“By enhancing feelings of satiety and suppressing
hunger, GLP-1RAs may exacerbate dietary restriction
(physiological undereating) and dietary restraint (the
cognitive effort to limit food intake),” Dalle Grave said.
“This can encourage the adoption of rigid and extreme
eating patterns, such as skipping meals, consuming
minimal portions or avoiding specific foods altogether.
These behaviors are core characteristics and
perpetuating factors of eating disorders. Moreover,
common side effects of GLP-1RAs, such as nausea,
vomiting and diarrhea, can further suppress appetite,
reinforcing restrictive eating behaviors.”
For those with existing eating disorders, Dalle Grave
explained how “the appetite-suppressing effects of GLP1RAs may conflict with key eating disorder treatment
approaches, which emphasize re-establishing regular
eating habits and reducing restrictive behaviors”.
Conversely, some researchers believe GLP-1RAs could
be used to treat binge-eating disorder (BED), which
is characterized by recurrent binge episodes without
compensatory behaviors such as excessive exercise,
inducing vomiting or using laxatives.
“Although some evidence suggests they may reduce
binge eating, GLP-1RAs are not approved for BED
treatment. Their use should be limited to cases where
binge episodes are substantially reduced, regular eating
patterns are established and healthy lifestyle changes
are adopted. Close monitoring is essential to detect
worsening eating disorder symptoms, such as rigid
dietary rules or meal skipping,” said Dalle Grave, “the
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 18
role of GLP-1RAs in obesity when it coexists with BED
requires further research.”
The full impact of weight loss drugs on other mental
health conditions remains unclear. A Scientific Reports
study found that liraglutide and semaglutide increased
anxiety and suicidal ideations in females and younger
participants in real-world settings. In one case, a
patient with a history of depression experienced
depressive symptoms after four weeks of treatment with
semaglutide, which ceased when treatment stopped.
Another study of the reported adverse events for
semaglutide, liraglutide and tirzepatide over a period
of 28 months found that while only 1.2% of the total
reports were psychiatric adverse events, there were 9
deaths and 11 life-threatening outcomes due to suicide
attempts in this time period. Depression, anxiety and
suicidal ideation were also reported.
The future of weight loss
Weight loss medications haven’t been around long
enough to have a full picture of their long-term effects.
“While clinical trials and post-market studies have
provided robust evidence of efficacy and safety over
a few years, there is still limited data on the long-term
effects of GLP-1RA use. This uncertainty is something
we’ll need to continue monitoring as more people use
these medications for extended periods,” said Kim. It
remains to be seen if these drugs could have impacts on
metabolism, mental health or other chronic conditions.
“By reducing obesity-related complications, GLP-1RAs
can lower long-term healthcare costs. However, at their
current net price in the US, the long-term cost savings
don’t fully offset the increased spending on these
medications. So, while they offer tremendous health
benefits, even with insurance coverage, patients often
face substantial out-of-pocket expenses.”
The high cost associated with these drugs is likely one of
the factors leading over 50% of users to discontinue use
before the one year mark. Over 84% of users without
type 2 diabetes discontinued use before two years.
According to Kim, the financial burden and potential
side effects of long-term GLP-1RA use highlight the
need for an alternative strategy.
Studies have also shown that patients can gain weight
after discontinuing GLP-1RAs. One study found a
significant association between weight re-gain and
reinitiation of treatment. According to Kim, “this occurs
because GLP-1RAs not only help reduce appetite and
improve metabolism while they're active, but they also
influence hormonal pathways that regulate hunger and
satiety. Once the medication is stopped, these pathways
often revert to their pre-treatment state, making it
harder for patients to sustain their progress without
additional support.”
A paper co-authored by Kim in 2024 suggested that,
while potentially less effective than weight loss drugs,
weight-maintenance strategies could optimize the
clinical benefits of reducing unhealthy weight more
efficiently and equitably.
“Weight-maintenance programs, which emphasize
lifestyle modifications like diet, exercise and behavioral
counseling – often following an initial weight loss phase
achieved with GLP-1RAs – offer a more sustainable
and affordable alternative to long-term medication
use. These programs avoid the high recurring costs
associated with GLP-1RAs and eliminate the risks of
medication-related side effects,” Kim said.
“This approach is particularly important for disadvantaged
populations, who are disproportionately affected by
obesity and its complications, and who stand to benefit the
most from equitable access to these interventions.”
One month supply list prices
Ozempic 1 mg (semaglutide): 936 USD
Wegovy 2.4 mg (semaglutide): 1,349 USD
Rybelsus 7 mg (semaglutide): 936 USD
Mounjaro 15 mg (tirzepatide): 1,023 USD
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 19
GLP-1RAs offer transformative health benefits to
many patients. However, these medications are not
without risks. It will be critical to address gaps in our
understanding of their long-term effects and to explore
how these medications can be integrated into broader,
more sustainable weight management strategies.
The future of weight loss treatments lies not only in
advancing pharmacological options but also in ensuring
these solutions are accessible, affordable and part of a
comprehensive strategy tailored to the diverse needs of
patients worldwide. •
ABOUT THE INTERVIEWEES
David D. Kim, PhD is an assistant professor of Medicine and
Public Health Sciences at the University of Chicago. As a health
economist, Dr. Kim's primary focus centers around measuring the
value of health interventions and providing guidance for valuebased health care decisions.
Riccardo Dalle Grave, MD is the director of the Department of
Eating and Weight Disorders at Villa Garda Hospital (Garda,
VR, Italy). Currently, the focus of his research is evaluating the
cognitive behavior therapy of adult and adolescent patients with
eating disorders and obesity, respectively, both in outpatient and
inpatient settings.
Choose Lonza for Optimal Cell Performance
Lonza’s primary cells and media are globally renowned
for quality and reliability. With over 40 years of experience and referenced in 10 times more publications than
other suppliers, Lonza can deliver the quality you need
to obtain reliable, and biologically relevant reults every
time.
Our comprehensive cell type offering is unmatched
and spans across research areas such as respiratory,
neurobiology, cancer, stem cells, cardiovascular,
toxicology, and others.
Primary Cells and Media Portfolio:
• 100+ cell types – cryopreserved, human and animal
• Broad donor variety – large selection of donors for
most cell types
• Cells from normal and diseased tissues
• Optimized Growth Media BulletKit® for each cell type
Your Trusted Source for Primary Cells
lonza.com/primary-cells
All trademarks belong to Lonza, and are registered in the USA, EU and/or CH or belong to
third-party owners and are used only for informational purposes. However, no warranty is made,
either expressed or implied. For more details: http://www.lonza.com/legal.
© 2025 Lonza. All rights reserved. CD-AD154 03/25
Harvest
40 Years of
Primary Cell
Expertise
Enabling a Healthier World
Learn more.
21 DRUG DISCOVERY
Antibodies have become an integral part of research
and medicine. Therapeutic applications of antibodies
began to emerge following the publication of Köhler
and Milstein’s groundbreaking 1975 paper in Nature
describing the hybridoma technique to produce
monoclonal antibodies (mAbs).
Introducing mAb technology enabled researchers to
produce a single antibody to a specific antigen and
replicate it in culture. In 1986, the Food and Drug
Administration (FDA) approved the first therapeutic
mAb designed to prevent transplant rejection. Since
then, more than 100 mAbs have been approved
for treating a range of diseases including cancer,
autoimmune and chronic inflammatory diseases.
mAbs synthesized in the laboratory mimic natural
antibodies produced by the body and are designed
to target and neutralize pathogenic proteins or
antigens selectively. A specific class of mAb,
capable of broad neutralization, has become highly
valued in vaccine development for its ability to
neutralize multiple virus strains.
Broadly neutralizing antibodies (bNAbs) were first
discovered in the '90s when it was observed that HIVinfected individuals possessed antibodies that could
recognize and neutralize different subtypes of HIV.
bNAbs are now being heavily researched, not only for
their potential in the prevention of HIV, but also for
other rapidly mutating viruses such as influenza and
SARS-CoV-2.
How Broadly Neutralizing
Antibodies Are Driving
Next-Gen Vaccines
Blake Forman
Credit: Technology Networks
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 22
Developing vaccines against an
evolving threat
During replication, the nucleic acid of many viruses
will mutate, reducing the protective ability of vaccines.
Combating the ability of mutated viruses to escape
the immune system remains one of the most pressing
challenges faced by vaccine developers.
“Viruses are constantly under selective pressure from
the human immune response,” Dr. William Voss, a
postdoctoral fellow at The University of Texas at Austin
told Technology Networks. “A particular virion that is
neutralized by an antibody during infection cannot
enter its host cell and replicate. This drives the process
of immune escape, where viral variants that mutate the
epitope targeted by a given neutralizing antibody, such
that it no longer binds or neutralizes, survive and pass
on their genetic material.”
The development of broad-spectrum vaccines that can
induce bNAbs against various virus strains has been
identified as one of the most promising ways to combat
these mutations.
“When it comes to bNAbs potential in vaccines,
two approaches are the subject of investigation. The
first involves the concept of passively transferring
vaccine-like antibodies into an individual. This would
only be possible if you can extend the half-life of
said antibodies,” Dr. Michael Diamond, the Herbert
S. Gasser Professor in the department of medicine,
molecular microbiology, pathology and immunology
at Washington University School of Medicine told
Technology Networks.
“This approach was taken by AstraZeneca when it
produced Evusheld® (tixagevimab/cilgavimab), a long
half-life mAb combination that could be administered
prophylactically to combat emerging waves of
SARS-CoV-2. The problem with Evusheld, and
most other therapeutic anti-SARS-CoV-2 mAbs,
was that it lost its effectiveness against newer variants
and resistance emerged.”
Another concept is to induce bNAbs by providing a
tailored antigen designed to focus the immune response
on conserved epitopes of a virus. “There are vaccines
that are trying to do this, and although they've been able
to generate broadly neutralizing responses there hasn’t
been much success generating bNAbs consistently,”
explained Diamond.
Unraveling the mechanism of
these antibody “heroes”
Like most antibodies, bNAbs are Y-shaped proteins
(Figure 1) generated by a type of white blood cell called B
What are conserved epitopes?
Conserved epitopes are portions of a
foreign protein or antigen retained by
multiple strains of a virus that binds with a
complementary site of an antibody – such
as the S2 domain of SARS-CoV-2.
FIGURE 1: Structure of an antibody.
Credit: Technology Networks
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 23
cells. The exact mechanism of action of bNAbs has been
an ongoing area of investigation. However, research has
shown that most bNAbs target conserved epitopes.
In 2022, researchers from The Pasteur Institute’s Virus
and Immunity Unit described how anti-HIV bNAbs
bind to viral particles to form aggregates at the surface of
immune cells. These aggregates effectively block cellto-cell transmission by preventing the release of viral
particles and the formation of synapses used by viruses to
move from one cell to another.
Producing bNAbs naturally in the human body is a
long process in an antibody–virus race for dominance.
For example, potent bNAbs develop in people living
with HIV, but only rarely and after many months or
even years after transmission.
“Dr. Dennis Burton's group and others have shown
that you require a substantial amount of somatic
hypermutation over the context of many exposures to
evolve these bNAbs. Work from Dr. Michel Nussenzweig
has also shown this elegantly at the single B-cell level,”
said Diamond.
As you could expect, identifying and characterizing
bNAbs is no easy task; scientists have to comb through
thousands of antibodies to identify those likely to be
active against most viral strains.
Typical approaches for the high-throughput screening
of antibodies include phage display, whereby highaffinity interactions between antibodies and antigens
are identified by displaying proteins on the surface of
bacteriophages. Techniques such as enzyme-linked
immunosorbent assays (ELISAs) are often employed in
hybridoma screening to identify monoclonal antibodies
with high antigen affinity.
Diamond explained how antibody screening can also be
done at a single B-cell level: “You can identify specific
B cells that interact with a viral protein, and then make
the antibody that single B cell encodes. Now you have
a single antibody from a single B cell that binds to your
protein of interest. Depending upon the sophistication of
your screen, you may even be able to know right away if
that's a neutralizing anybody.”
More recently, artificial intelligence (AI) and machine
learning approaches have been suggested. AI could be
useful for screening methods such as epitope/paratope
mapping to predict the areas of the antibody (the
paratope) and antigen (the epitope) involved in binding.
However, AI methods can lack accuracy and still require
experimental input.
In the context of mutating viruses, researchers have
used machine learning to computationally redesign an
existing antibody against an emerging strain of SARSCoV-2 while maintaining efficacy against the dominant
variant. The study's findings suggest that computational
approaches can be used to optimize an antibody to target
multiple escape variants.
Identifying bNAbs and utilizing them in vaccine technology has proven challenging, however, researchers
have made multiple strides this year alone in disease
areas such as COVID-19 and HIV.
Towards a preventative
HIV vaccine
Today, infection with HIV is manageable using
antiretroviral medications, however, a successful vaccine
has alluded developers since the ‘80s. A critical roadblock
in developing a preventative vaccine has been the
inability to induce B-cell lineages of bNAbs that target the
rapidly evolving virus.
A vaccine candidate developed at the Duke Human
Vaccine Institute has now reportedly triggered low
levels of HIV bNAbs among a small group of people in a
clinical trial.
The vaccine targets an area on the HIV-1 outer envelope
called the membrane-proximal external region (MPER),
which remains stable even as the virus mutates. In the
study, the researchers analyzed data from the HVTN 133
Phase 1 clinical trial, which involved 20 healthy, HIVnegative individuals.
“One of the questions we have worried about for many
years is if it will take years to induce bNAbs with a
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 24
vaccine like it takes for bNAbs to develop in people living
with HIV,” Dr. Barton Haynes, director of the Duke
Human Vaccine Institute previously told Technology
Networks. “Here we found that bNAb lineages developed
after the second immunization.”
This work shows the feasibility of inducing antibodies
that neutralize the most difficult strains of HIV, however,
the researchers stress that there is still more work to be
done to create a more robust response.
A separate study, conducted by the Scripps Consortium
for HIV/AIDS Vaccine Development, utilized germline
targeting to stimulate animals’ immune systems to induce
rare precursor B cells of a class of HIV-targeting bNAbs
called 10E8.
The 10E8 bNAb binds to a conserved region of the
glycoprotein gp41 on HIV’s surface. Designing an
immunogen to stimulate the production of 10E8 bNAbs
has been challenging because the binding region of gp41
is hidden in a recessed crevice on the virus’ surface.
To address this challenge, the researchers engineered
immunogens on nanoparticles that mimic a specific part
of gp41. Vaccinations in rhesus macaque monkeys and
mice with these immunogens elicited responses from
the 10E8 B cell precursors, and induced antibodies that
showed signs of maturing into bNAbs that could reach
the gp41 region.
The research forms part of the group's larger work to
develop a germline-targeting strategy for priming the
immune system to elicit a bNAb called VRC01, which was
discovered by NIAID researchers almost 15 years ago.
bNAbs presence in COVID-19
research
The first vaccine a person receives promotes a strong
primary immune response that shapes future interactions
with the virus, a process known as imprinting.
Diamond and colleagues at Washington University
School of Medicine in St. Louis recently detailed how
imprinting affects subsequent COVID-19 vaccinations.
Unlike the influenza virus, where immunity elicited
by one year’s flu shot can interfere with subsequent
immune responses, the researchers found that repeat
vaccinations resulted in the production of bNAbs capable
of neutralizing a wide range of SARS-CoV-2 variants.
The researchers concluded that these cross-reactive
antibodies may confer substantial protection against
future pandemics caused by a related coronavirus.
One bNAb to neutralize all variants
Researchers at The University of Texas at Austin
recently discovered and isolated a particular bNAb,
called SC27, which appears effective at neutralizing the
numerous variants of SARS-CoV-2. “This research –
including understanding how SC27 achieves its broadly
neutralizing activity – helps inform vaccine development,”
explained Voss. “Future vaccines that can trigger the
production of such antibodies could protect us more
broadly against emerging viral variants as SARS-CoV-2
continues to evolve. Additionally, SC27 itself could
potentially be used as a therapeutic antibody.”
SC27 recognized the different characteristics of the spike
proteins in the many COVID variants. “A technique
called deep mutational scanning quantified the potential
“Antibodies such as
SC27 that may retain
neutralizing activity
despite future viral
evolution represent
a promising area of
drug development,”
said Voss.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 25
for an array of spike mutations to escape SC27 and
demonstrated that such mutations are rarely observed in
circulating SARS-CoV-2,” described Voss.
“Several key amino acid residues within the epitope
targeted by SC27 show minimal natural variation across
SARS-CoV-2 variants, suggesting that SC27 may remain
potently neutralizing moving forward.”
To isolate and identify the antibody the researchers
developed a novel technique combining single-B cell
sequencing and immunoglobulin proteomics, a method
they named Ig-Seq.
Beyond COVID-19 research, Voss believes this approach
could be used to study other rapidly mutating viruses: “As
a high-throughput method for characterizing the plasma
antibody repertoire and facilitating the cloning and
characterization of specific antibodies of interest, Ig-Seq
could be used to identify broadly neutralizing epitopes on
a variety of pathogens, including influenza.”
Voss continued, “Identifying such regions of viral proteins
and understanding the mechanism of antibodies that
target them could inform the development of vaccines
that aim to elicit bNAbs.”
How close are we to “one shot”
vaccines?
Advancements in structural biology and immunologic
technologies have brought researchers closer than ever
to understanding bNAbs and the mechanisms that drive
their production.
“To get a single shot vaccine to prevent infection
in a sustained way, we also need to accelerate the
development of mucosal vaccines. The emerging
consensus is that most vaccines administered by an
intramuscular route don’t induce enough upper airway
immunity, especially in the setting of evolving variants that
compromise neutralizing activity,” Diamond said of the
possibility of a “one shot” vaccine for respiratory viruses.
Diamond envisions two approaches could translate
bNAbs to the clinic in the next few years. The first
involves the concept of vaccine-like antibodies with an
extended half-life. Instead of vaccinating and inducing
an immune response, this would involve giving a patient
a bNAb.
“The advantage of this approach is that elderly and
immune-compromised people wouldn’t have to rely on
a dysfunctional immune system making an antibody,”
explained Diamond. “The downside is that you run the
risk that if a virus is highly evolving you could generate
escape in a population relatively quickly, as was the case
with Evushield.”
The second approach would involve using vaccines to
induce the production of bNAbs in a patient.
“Work still needs to be done to improve our
understanding of how to focus and display epitopes
recognized by bNAbs to be able to induce antibodies
specifically and with high precision. We are still struggling
as a field in the best way to do that. The rules that may
work for flu may not be the same for HIV because the
epitopes are different and must be displayed differently.
“Alongside targeting specific epitopes with
bNAbs, a successful ‘one shot’ vaccine will also
need to induce ancillary immune responses such
as T-cell responses and Fc effector responses,”
said Diamond.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 26
Unfortunately, the lessons we continue to learn from
one virus might not translate across different viruses,”
Diamond concluded. •
ABOUT THE INTERVIEWEES
Dr. Michael Diamond is the Herbert S. Gasser Professor in the
Department of Medicine, Molecular Microbiology, Pathology and
Immunology at Washington University School of Medicine. He
earned his PhD in cell and developmental biology from Harvard
University and an MD from Harvard Medical School. His research
group studies the molecular basis of disease of globally emerging
RNA viruses, focusing on the interface between pathogenesis and
host immunity.
Dr. William Voss is a postdoctoral fellow at The University of
Texas at Austin. He earned his PhD in cell and molecular biology
from The University of Texas at Austin during which he was
awarded the Outstanding Research Paper Award, Department
of Molecular Biosciences, The University of Texas at Austin,
April 2022 for the 2021 publication in Science titled “Prevalent,
protective, and convergent IgG recognition of SARS-CoV-2
non-RBD spike epitopes”.
27 DRUG DISCOVERY
Gain More Insights.
Make Better Decisions.
Sartorius accelerates the development of breakthrough therapeutics
with innovative solutions for lab filtration, cell/protein analysis, and more.
Ensure confidence in your biotherapeutic discovery and development
program through:
• Process streamlining
• Multiplexing
• Information-rich data
• Biologically relevant insights
www.sartorius.com/biologics
29 DRUG DISCOVERY
Impurities in pharmaceuticals are a major concern
for drug manufacturers, as they can significantly
impact the quality, safety and effectiveness of the final
product. Even a single unknown impurity discovered
late in production can lead to the rejection of entire
batches. The International Council for Harmonisation
(ICH) defines an impurity as any substance in a
drug product that is not the active pharmaceutical
ingredient or the excipients.
Impurities are categorized into organic impurities,
inorganic impurities, other materials and residual
solvents. They can originate at various stages – from
manufacturing to transportation and storage. During
production, impurities may arise from interactions
between the active pharmaceutical ingredient (API)
and excipients, or from contact with packaging
materials. Understanding these sources is crucial for
maintaining the purity and safety of pharmaceuticals
throughout their lifecycle.
Impact of impurities on product
approvals and regulatory actions
The presence of impurities can delay product
approvals during manufacturing and prompt recalls
once products are on the market, triggering regulatory
actions. Recent incidents involving substances like
N-nitrosodimethylamine (NDMA) in medications such
as ranitidine and valsartan have resulted in widespread
recalls, highlighting the ongoing challenges in effectively
managing pharmaceutical impurities.
Navigating the Complexities of
Impurities in Pharmaceuticals
Neeta Ratanghayra, MPharm
Credit: iStock
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 30
Naiffer Romero, USP Principal Scientist and Community
Manager of the Nitrosamines Exchange says, “Since
nitrosamines were first found in drug products in
2018, there has been a coordinated effort among
international regulatory and health agencies to advance
the development of guidance to mitigate their presence,
collaboration with industry and manufacturers to
deepen the science and understanding of identifying
and testing these impurities in medicines. Although
our understanding of the issue has come a long way,
it continues to evolve as new challenges arise, and the
problem facing industry is far from over. Regulatory
agencies initially aimed to unify their approach to
nitrosamines in pharmaceuticals, but global variation in
testing capabilities has led to differing stages of managing
and reducing these impurities. This highlights the
importance of discussions and educational efforts among
regulators, drug manufacturers and industry stakeholders
to address the issue.”
ICH guidelines for managing
impurities in drug substances
and products
The quantification, qualification, identification and
control of impurities are critical in drug development to
ensure the safety and purity of the final drug product.
International and regional guidelines play a key role
in guiding drug developers and regulatory agencies
in evaluating and managing impurities in both drug
substances and products.
The ICH has established several key guidelines for
managing impurities. The ICH Q3A focuses on impurities
in new drug substances, while ICH Q3B addresses
impurities in new drug products. These guidelines
outline essential criteria including identification and
characterization, accurate quantification, established
limits and understanding the origin of impurities.
They also emphasize the importance of conducting
toxicological assessments, evaluating the stability of the
drug in the presence of impurities, ensuring regulatory
compliance, performing thorough risk assessments and
implementing effective control strategies.
While reducing impurities to practical minimums is ideal,
complete elimination is often not feasible, necessitating
the establishment of specific impurity criteria. The ICH
Q3A and Q3B guidelines provide essential thresholds
for reporting, identifying and qualifying impurities in new
drug substances and new drug products respectively.
Impurity investigations by phase
of development
Managing impurities is crucial throughout the
entire product lifecycle, from initial development to
manufacturing and distribution. Yaman Abdin, research
fellow at Saarland University, says, “Impurities are
everywhere! This simple truth keeps colleagues in
quality control, from devising guidelines to implementing
strategies, very busy. Impurities are an inevitable
challenge in chemistry, especially in pharmacy, and the
field is dynamic and evolving.”
The level of scrutiny in impurity investigations depends
on the project's development phase. In the early
stages of development, it's impractical to investigate
all impurities; the focus should be on the most likely
potential impurities observed in the initial phases (e.g.,
intermediates, solvents, predictable by-products from
stress studies). In the later phases of development, more
detailed studies are conducted to understand impurity
origins and behaviors, guiding decisions on which
impurities require routine monitoring with defined limits.
Stress degradation studies in later phases further identify
degradation products, helping to inform formulation and
storage requirements.
“We're constrained by the accuracy and precision of our
analytical methods as well as the fact that we only find
what we look for. To manage impurities effectively, we
must focus on the purity of starting materials, controlled
processing environments, and proper handling and
storage. Adopting a “quality by design” approach is key to
achieving better purity profiles,” explains Abdin.
Analytical techniques for
impurity detection
A variety of analytical techniques are employed to
identify and quantify impurities throughout the drug
development process. Each method offers distinct
advantages and is suited to specific types of analyses.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 31
Below, we explore several key analytical methods used for
impurity detection in pharmaceutical products.
Advances in NMR‐based
characterization of
drug impurities
NMR facilitates the identification and confirmation of
molecular structures, aiding in the detection of impurities.
NMR helps elucidate the mechanisms behind the
formation of process and degradation impurities, which
is vital for maintaining drug quality. However, challenges
remain. Gary Martin, adjunct professor at the Stevens
Institute of Technology, explains, “Undoubtedly, the
biggest challenge associated with the identification
and characterization of drug impurities is the size of
the sample that can be isolated vs. the NMR probe
technology available where the research is being done.
While sensitivity has never been an issue for mass
spectrometric methods, quite the opposite is true for
NMR spectroscopy, which is notoriously sensitivitychallenged. Obviously, higher-field NMR spectrometers
are advantageous.”
Highlighting the recent advances, Martin says “Probably
one of the most significant recently developed techniques
that can be used in the characterization of drug impurities
was the development of the i-HMBC experiment, which
allows the unequivocal differentiation of two-bond from
three-bond long-range or longer-range correlations.
The full potential of this experiment and the range of
applications has only just begun to be realized.”
Method Description
Thin layer
chromatography
Cost-effective method for separating drug components and identifying impurities; minimal
sample preparation and high sample loading capacity.
High-performance
liquid chromatography
Offers excellent specificity and precision; often combined with mass spectrometry (LC-MS) for
impurity analysis. Requires extensive system suitability testing.
Gas chromatography Effective for analyzing volatile organic compounds; derivatization expands its application to nonvolatile substances. Used to analyze residual solvents and process-related impurities.
Near-infrared
spectroscopy
Rapid, non-destructive method suitable for multi-component analysis with minimal sample
preparation. Ability to extract multiple parameters from a single spectrum.
Nuclear magnetic
resonance (NMR)
spectroscopy
A valuable tool for identifying and confirming molecular structures, thus helping detect
impurities. It also helps in decoding the mechanisms behind their formation and degradation.
Electrochemical
methods
Gained popularity for drug analysis due to advanced instrumentation and improved
understanding; includes methods like cyclic voltammetry and adsorptive stripping voltammetry.
Kinetic methods Focuses on measuring concentration changes over time; applicable techniques include fixed-time
and initial rate methods.
Electrophoretic
methods
Capillary electrophoresis offers effective separation of charged analytes; efficient and requires
minimal sample volume.
Flow injection and
sequential injection
analysis
Flow injection analysis (FIA) automates chemical procedures by injecting samples into a
continuous liquid stream, allowing real-time measurement. Sequential injection analysis builds
on FIA principles with programmable flow, enhancing automation in pharmaceutical analysis.
Both methods improve sampling rates.
Hyphenated techniques
Combine separation methods with online detection. Includes LC-MS, GC-MS (gas
chromatography-mass spectrometry), CE-ICP-MS (capillary electrophoresis-inductively
coupled plasma mass spectrometry) and CE-MS (capillary electrophoresis-mass spectrometry).
TABLE 1: Analytical methods used for the detection of impurities in drug products.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 32
Strategies for minimizing
drug impurities
To minimize drug impurities, several effective strategies
can be implemented. Firstly, optimizing synthesis
routes and process controls ensures efficient pathways,
significantly reducing impurity formation. Additionally,
using high-quality starting materials with low impurity
profiles helps to limit contaminants in the final product.
Strict adherence to good manufacturing practices
(GMP) is essential for maintaining high production
standards and minimizing impurities. Controlled storage
and handling conditions, including proper temperature
and humidity management, prevent degradation and
the formation of impurities. Lastly, thorough validation
of cleaning procedures ensures that equipment and
facilities are free from cross-contamination, further
safeguarding product integrity.
“In addition to the general requirements manufacturers
must meet to analyze their products for impurities, to
identify those that may arise during manufacturing, and to
conduct stability studies – when it comes to nitrosamines
specifically – there is a need for more preemptive actions
to effectively head off the risks,” Romero explains.
As per Romero, pharmaceutical companies can take the
following proactive actions to mitigate the risks:
• Develop and implement risk-based assessments
to evaluate chemical structures, manufacturing
processes, raw materials and historical data
to help determine which products are more
vulnerable to the formation of nitrosamines.
• Adopt a culture of continuous improvement,
regularly reviewing and refining their risk
management strategies, manufacturing processes
and testing protocols.
• Proactively test medicines for impurities
throughout a product’s life cycle for better
detection, assessment and action to keep the
global medicines supply chain safe.
Regardless of the source of the impurities – whether
from manufacturing to transportation or storage –
comprehensive risk assessment and mitigation ultimately
helps determine the safety of a product in the hands of a
patient, adds Romero.
Promoting collaboration
and innovation in addressing
drug impurities
By adopting innovative strategies and fostering
partnerships across the industry, stakeholders can
effectively address the emerging issues of drug
impurities and prevent unforeseen product recalls.
Romero says, “Information and knowledge sharing
allows for open discussions regarding recent findings
and updated regulatory requirements. To facilitate
collaboration and information sharing among the
global network of professionals concerned with the
issue, USP created the Nitrosamines Exchange, a
dedicated knowledge-based community designed
to allow industry subject matter experts, along with
representatives from international regulatory and health
agencies, to discuss recent updates and engage in active
problem-solving. Members of the quickly growing
community of 4,000+ share information and solutions
to mitigate nitrosamines through thread conversations,
questions and anecdotal experiences. By participating in
these types of conversations, industry can help prevent
future nitrosamine-related recalls and help ensure
medicines remain safe for patients.”
Abdin adds, “Global harmonization of regulatory
standards and constantly improving analytical techniques
have significantly enhanced our ability to detect and
control impurities. However, emerging challenges such as
microplastic and nanoplastic impurities could find their
way or maybe already found their way to drug products
and hence represent the next frontier. Addressing these
new types of contaminants will require innovative
methods and continued global collaboration to ensure the
safety and efficacy of pharmaceuticals.” •
ABOUT THE INTERVIEWEES
Gary Martin is an American chemist and expert in NMR spectroscopy
and medicinal chemistry. Formerly with the Structure Elucidation
Group at Merck, he is now an adjunct professor of Chemistry at
both Seton Hall University and Stevens Institute of Technology.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 33
His research focuses on developing new NMR methods for
characterizing the molecular structure of pharmaceutical impurities,
degradants, drug metabolites and natural products. Martin has
written a widely used monograph on 2D NMR methods and coedited
two volumes on modern NMR applications in natural product
structure elucidation. He has published over 325 papers, more than
45 invited reviews and chapters, and delivered over 500 seminars
and lectures at national and international meetings.
Naiffer Romero is a Principal Scientist at the U.S. Pharmacopeia (USP),
where he leads scientific outreach and technical engagement on
national health priorities in Latin America and the United States. With
more than 20 years of experience in the pharmaceutical industry and
an expert in nitrosamine impurities, he manages USP’s Nitrosamines
Exchange online community and serves as USP representative on
several international nitrosamines workstream committees.
Yaman Abdin is a postdoctoral researcher at the Institute for
Bioorganic Chemistry, School of Pharmacy, Saarland University,
Germany. His research focuses on the philosophical, social and
psychological aspects of pharmacy, encompassing drug development,
regulation and application. He leads the 'Pharmasophy' group at the
Institute for Bioorganic Chemistry.
Reserve Your Place for Early Access
Be among the first to secure your spot at our exclusive online event on drug discovery and
development! Gain early access to cutting-edge discussions on small molecule and biologic
drug development, high-throughput screening and novel target identification strategies.
REGISTER NOW
Brought to you by the publication
September 24-25, 2025
8AM PDT | 11AM EDT | 4PM GMT
Advances in Drug Discovery &
Development 2025
ONLINE SYMPOSIUM
TECHNOLOGY SPOTLIGHT SPONSORS STANDARD SPONSORS
PLATINUM SPONSOR
• Be one of the first to gain access to scientific
discussions
• Receive exclusive updates directly from our
event platform
• Get event insights as content is ready
• Stay informed as talks and speakers are
confirmed
• Enjoy priority Q&A: Submit your questions
early and they will be presented during the
online event on a first-come, first-served basis
Benefits of Early Access
35 DRUG DISCOVERY
In recent years, we have seen significant advances
in the development of drugs to tackle Alzheimer’s,
particularly in the wake of two decades of largely
unfruitful research. Though some such treatments are
being hailed as a success, not all researchers remain
convinced. Regulators around the world have had
differing receptions to the drugs, but despite their
problems, research now seems to be trending in a
positive direction.
Emerging new treatments
The past two years has seen regulators approve new
drugs lecanemab and donanemab to treat Alzheimer’s
disease, though their development hasn’t been plain
sailing. Both are designed to target a protein called
amyloid-beta in the brain that forms abnormal clumps
called plaques. These plaques are thought to be the root
cause of the disease as part of the “amyloid hypothesis”,
though this idea has also been disputed by some.
The first amyloid-targeting antibody to gain approval
from the US Food and Drug Administration (FDA) –
aducanumab – was given the green light in 2021. But it
was not without controversy; three FDA advisors, whose
committee had advised against aducanumab’s approval
due to a lack of evidence of efficacy, resigned shortly
after the decision. Aducanumab’s drugmaker, Biogen,
later discontinued the drug in January 2024 to focus on
the development of their new offering, lecanemab.
The Evolving Treatment
Landscape for Alzheimer’s
Sarah Whelan, PhD
Credit: iStock
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 36
Data from clinical trials suggest that amyloid-targeting
drugs like lecanemab and donanemab provide modest
benefits for slowing the disease’s progression, mostly
during the early stages of the disease. Despite their costs
– both risk-wise and financially – their approval heralds
some hope for those affected by Alzheimer’s, and it is a
positive step after years of trials finding that previous
drug candidates failed to impress.
There are several reasons why finding effective drugs
to combat Alzheimer’s has been so difficult, as Andrew
Doig, a professor of biochemistry at the University of
Manchester, told Technology Networks: “Firstly, the
target that has received most attention, amyloid-beta,
is not at all easy to hit.”
Doig explained that amyloid-beta likely exists in the
brain in the form of multiple units stuck together in
structures called oligomers. Each different oligomer
can vary in toxicity and structure, making finding drugs
that effectively bind to them a much more difficult task.
“Even if we do have good structures of oligomers, it isn’t
clear which would be the most important (i.e., the most
toxic). It is also unlikely that a drug would bind to all the
toxic forms,” he said.
The complex pathologies in the brain and the
mechanisms at play that underpin Alzheimer’s
development also make it challenging to target, as the
disease’s characteristic brain abnormalities can emerge
decades before any apparent symptoms.
“By the time it is diagnosed, it could be too late to
treat, as the brain has already undergone significant
irreversible damage,” Doig said. To increase the chances
of success, patients in the earlier stages of dementia –
such as mild cognitive impairment – are now beginning
to take center stage for clinical trials.
Amyloid-beta’s other knock-on effects could also
indicate that early treatment may be best, Doig
explained, as the accumulated amyloid-beta likely
kickstarts feedback loops in the brain involving
oxidative stress, inflammation and tau – another key
protein implicated in Alzheimer’s development.
“Once these loops are triggered, removing toxic forms
of amyloid-beta may have little effect,” he explained.
Drawbacks for newly
approved therapies
Nonetheless, somewhat encouraging data from clinical
trials have provided support for the approval of amyloidtargeting antibodies, suggesting they could have modest
benefits for slowing down the disease’s progression.
Lecanemab, for example, slows the progression of
the disease in its earlier stages, rather than halting or
reversing it. “All it does is to slow down the rate at which
the disease progresses, as measured by cognitive ability,”
Doig explained. “In effect, patients who take lecanemab
see a delay in progression to more severe dementia by
about six months.”
These antibodies are delivered into a vein with the
aim of binding to and clearing the brain’s amyloid-beta
plaques; but not all are convinced as to whether this
necessarily translates into meaningful benefits for the
patients taking it.
“The benefits of lecanemab are so modest as to be
undetectable in an individual treated patient,”
said Robert Howard, professor of old age psychiatry
at University College London. “Although 27% slowing
of disease course sounds impressive, this is not
strictly what the analysis of the trial data showed.”
Another drawback to these treatments is finding
patients who are eligible to receive the drug.
This requires undergoing expensive or invasive
procedures such as PET scans and lumbar punctures,
notwithstanding the eyewatering costs of the drug
alone, which the manufacturers have set in the region
of $26,500 a year.
“The benefits are small, however, and there are
concerns with the drug,” Doig adds, as, even after
considering the costs, the drug carries the risk of
potentially severe side effects.
“There is a small but real risk that lecanemab can cause
brain hemorrhage. A few patients on the trial died in this
way,” Doig said, referring to amyloid-related imaging
abnormalities (ARIA), which cause potentially fatal
microbleeds or clots in the brain.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 37
Weighing the risks versus the benefits is therefore a
difficult task. The patients likely to benefit most from
the drug are those in the earliest stages of the disease,
who, coincidentally, have the most to lose considering
the risks.
Weighing the benefits and risks
This decision on whether to approve these drugs has
proved arduous for many regulators worldwide, with
some taking more conservative stances than others.
The FDA was the first to give the green light in July
2023, while the European Medicines Agency (EMA)
later decided against its approval in July 2024. The UK
was next to follow in the FDA’s footsteps to approve
lecanemab. However, they ruled against making it
available on the National Health Service, reasoning that
the small benefits do not justify the cost – meaning only
those able to afford private healthcare can receive it.
Another anti-amyloid antibody, donanemab, which also
modestly slows cognitive decline at the risk of ARIA,
earned FDA approval in July 2024.
“The benefits are marginal, the costs are high and there
is some risk with these drugs, so it is not surprising that
many regulatory bodies are saying no,” Doig summarized.
“I can understand why some do approve them, however,
as we desperately need new therapies for Alzheimer’s,
even if they are not especially effective.”
Though further research could provide safer and more
effective drug candidates, the approval of these first
few drugs may be the starting point to show that we
are on the right track. “Patients and carers can take
heart that it is possible to slow cognitive decline caused
by Alzheimer’s and that amyloid-beta is indeed an
appropriate [drug] target,” Doig said. “It therefore points
the way to a future where Alzheimer’s can be treated,
bringing benefit to millions of people.”
Potential for other
treatment avenues
Aside from amyloid-targeting antibodies, there are also
other avenues that may be worth pursuing to add more
tools to combat the disease.
Doig described these in some of his predictions for the
future of research into Alzheimer’s treatments over the
coming decade: “We will bring forward a wider range of
targets, rather than solely trying to clear amyloid-beta.
Tau is next, but anti-inflammatories and antivirals also
show promise. Many other pathways are affected by
Alzheimer’s and might be useful drug targets.”
Drug repurposing, like that of anti-inflammatories and
antivirals, is one such approach that could be used to
identify useful drug targets. Repurposing is the process of
finding new applications for existing drugs: a significantly
cheaper and potentially faster method to find new and
effective treatments for many diseases.
“Some therapies aimed at other conditions are showing
real benefit for Alzheimer’s. These are the shingles
vaccine, Shingrix®, and the anti-obesity drugs, such
as semaglutide,” said Doig.
The shingles vaccine targets a protein called HSV-1
that causes both chicken pox and shingles. Doig’s
“Patients and carers can take heart that it is
possible to slow cognitive decline caused by
Alzheimer’s and that amyloid-beta is indeed an
appropriate [drug] target,” Doig said.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 38
colleague, Ruth Izhaki, pioneered the investigation
into the link between Alzheimer’s and HSV-1 for
decades. However, her work has only recently gained
wider acceptance.
“The Shingrix vaccine seems to be roughly as effective
as lecanemab at a much lower cost, without fortnightly
administration and the worrying side effects,” Doig said.
Meanwhile, tackling other conditions closely associated
with Alzheimer’s could be another promising option to
limit the disease’s incidence and progression: “We know
that cardiovascular problems in middle age increase the
risk of Alzheimer’s later on,” Doig said. “Tackling type 2
diabetes via suppressing appetite shows many benefits,
including lowering the risk of Alzheimer’s. These are
very exciting recent discoveries which offer cheap and
effective ways to reduce Alzheimer’s.”
The first step?
Though the effort to find treatments for Alzheimer’s
has not been straightforward, the emergence of these
new drugs represents what is hopefully the first step
on the journey to safer and more effective treatments.
With diagnoses for people aged 65 and over projected
to grow to 12.7 million by 2050, the pressing need for
breakthroughs to prevent or cure the disease is important
now more than ever. •
ABOUT THE INTERVIEWEES
Andrew Doig is a professor of biochemistry at the University
of Manchester. He holds an MA in natural science and PhD in
chemistry from the University of Cambridge, and carried out
postdoctoral work at Stanford University.
39 DRUG DISCOVERY
It is estimated by the World Health Organization (WHO)
that by the end of 2022, 39 million people were living
with human immunodeficiency virus (HIV), and in that
year alone, 630,000 people had died from HIV-related
causes and 1.3 million people had acquired the disease.
With the assistance of 11 UN cosponsors, UNAIDS is
hoping to end the HIV and AIDS epidemic by 2030.
This article will explore the difficulties faced in
attempting to treat HIV, an innovative prevention
strategy and the possibility of a cure.
Challenges in HIV management
HIV is a retrovirus and can integrate its genetic material
into host genomes, making it difficult to treat, and
currently lacking a cure. If left untreated, HIV can lead
to acquired immunodeficiency syndrome (AIDS).
Unveiling the Progress
and Challenges in the Fight
Against HIV
Kate Robinson
Credit: iStock
What is AIDS?
AIDS is a disease that occurs when the
immune system is damaged by HIV.
When treated, HIV is prevented from
progressing, so many people with HIV do
not develop AIDS.
A person with HIV is considered to have
AIDS either when the number of CD4 (a
type of white blood cell) cells drops below
200 cells per cubic millimeter (A healthy
person has anywhere from 500 to 1,600
cells per cubic millimeter) or if they develop
one or more opportunistic infections.
Without treatment, people with AIDS
typically survive around 3 years.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 40
Even though treatment can help to effectively manage
the condition, some immune cells go into a dormant
state but still contain viral DNA and can reemerge once
treatment ceases.
“HIV can establish latent reservoirs within our body.
What this means is that they can hide within cells in
our body and lay dormant for a long period of time (e.g.,
months or years). Once they become active, they can
immediately replicate and infect other immune cells,”
said Dr. Emmanuel Ho, a professor at the University of
Waterloo. “HIV is able to alter its genetic makeup to
avoid our immune system or develop resistance against
anti-HIV drugs. HIV can also infect and kill immune
cells in our body. As a result, these immune cells will no
longer be able to participate in the fight against HIV.”
Combined antiretroviral therapy (cART) is currently
the only treatment for HIV. cART uses a mix of different
antiviral drugs to stop HIV replicating.
“cART targets the virus at several key stages of its
infection and replication cycle. By doing so, cART
reduces the amount of virus in the body to undetectable
levels, thereby protecting and preserving these cells
and the host's immune system,” said Dr. Jamie Mann,
senior lecturer in vaccinology and immunotherapy at
the University of Bristol. “When HIV infects T cells,
on rare occasions, rather than the virus replicating and
killing the host cell, the virus becomes dormant. In this
dormant state, the virus is not susceptible to cART and
can exist for a very long time.”
Although cART is the main treatment for those living with
HIV, this therapy cannot eradicate the latent reservoirs
developed by the virus, so the treatment must be ongoing
to be effective. While necessary, long-term use of cART is
associated with toxicity and drug resistance.
There is also a global disparity in access to treatments:
• Approximately 10 million people living with
HIV still do not have access to antiretroviral
therapy. According to the 2023 UNAIDS
report, only 43% of children living with HIV
have access to life-saving medicine.
• Over 25 million people living with HIV at the
end of 2022 were in Africa. Gender inequalities
continue to make an impact on the HIV/AIDS
response, particularly in sub-Saharan Africa. In
2022, women in the region accounted for 63% of
new infections. Worldwide, this figure was 46%.
• Adolescent girls and young women in subSaharan Africa are at a higher risk of infection
than other groups, in part due to violence,
stigma, discrimination and harmful laws and
practices. Other factors impacting the odds of
infection include unstable housing, lower levels
of education, poverty and food insecurity.
• Across the globe, HIV prevalence was 14 times
higher among transgender people, 11 times
higher among men who have sex with men, 7
times higher among people who inject drugs and
4 times higher among sex workers in 2022 when
compared with adults in the general population.
“Stigma, discrimination, healthcare access and
treatment adherence issues will all contribute to
different populations facing distinct challenges when
attempting to cure HIV,” said Mann.
Protecting against sexual
transmission of HIV
Dr. Emmanuel Ho leads a research group focused on
developing and characterizing innovative drug delivery
strategies, including nanomedicines, medical devices and
biomaterials, for the treatment and prevention of HIV/
AIDS, among other diseases. Ho recently developed a
siRNAs are non-coding doublestranded RNA molecules, typically
used to silence a gene of interest.
These RNA interference tools
are useful in the study of gene
function and can even be utilized
for the treatment of diseases such
as cancer.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 41
novel nanomedicine loaded with genetic material called
small interfering RNAs (siRNAs) to fight HIV.
“siRNAs will play an important role in the development
of novel HIV therapeutics. If we can identify a ‘target’
(e.g., gene) in our body that promotes or enhances HIV
infection, as long as we know the genetic sequence of
the target, we can easily design siRNAs to bind and
knockdown the expression of that target,” said Ho.
The nanomedicine is intended to reduce the expression
of CCR5 – a gene that encodes a protein expressed by
T cells and macrophages, known to be an important
co-receptor for HIV to enter host cells - to prevent HIV
from attaching to and entering host cells. “This gene
plays a role in the HIV infection process. By reducing the
expression of CCR5, we hope to reduce HIV infection,”
explained Ho.
By releasing a second siRNA to reduce the expression
of Nef, a protein produced by HIV to inhibit autophagy,
the nanomedicine can reactivate autophagy to eliminate
the HIV still present in the cells. As Ho explained,
“If, unfortunately, HIV is still able to infect immune
cells within the body, we hope that our nanoparticles
can reactivate autophagy.”
When administered directly into the vagina, naked
siRNAs have difficulties in achieving efficient mucosal
uptake due to their rapid degradation. To get around
these issues, Ho’s team encapsulated the siRNA in PEGPLGA polymer nanoparticles.
The nanomedicine was developed to be delivered
intravaginally to protect against sexual transmission
of HIV. However, intravaginal delivery can come with
challenges. “In order to deliver nanoparticles to the
submucosal layer where immune cells are located, it will
first have to move across a layer of cervicovaginal mucus,
which can trap the nanoparticles. Afterwards, it will
have to penetrate across an epithelial layer. Finally, it is
important to have the nanoparticles enter the appropriate
cells,” explained Ho.
The team plans to optimize the nanoparticle system to
attempt to achieve 100% protection against infection.
Looking ahead: Is a cure on
the horizon?
While infection with HIV is treatable, there is no cure
available. Dr. Jamie Mann is interested in addressing this
issue with the development of novel therapeutic and
prophylactic vaccines against HIV.
“The global distribution of HIV subtypes varies, with
certain subtypes being more prevalent in specific regions.
This variation presents unique challenges for an HIV
cure, as transmission, disease progression and responses
to treatments differ. Ultimately, the cost and complexity
of the treatment must be such that it is easily accessible
for all who require it,” said Mann.
Mann recently co-led an international study to
demonstrate the ability of a new therapeutic to cure
HIV. The therapeutic, an HIV-virus-like particle (HLP),
can reactivate the virus when dormant, rendering it
susceptible to cART and the immune system.
Using blood samples from 32 participants living with
chronic HIV, who were on stable cART for a median of
13 years, the team found that HLP was able to specifically
target just the immune cells containing latent HIV
reservoir and purge these cells of their HIV. “Our data
shows that divergent strains of HIV are also susceptible
to the same HLP treatment, suggesting that the HLP
could have global applications as a therapy,” said Mann.
HLP are engineered to resemble HIV and lack a viral
genome, making them incapable of causing an infection
Research is driving
the development of
new treatments and
could introduce a cure
for HIV, but ending
this epidemic will be a
global effort.
TECHNOLOGYNETWORKS.COM
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 42
or replicating. These particles can be administered
by intramuscular injection. The study shows that the
HLP reverses latency irrespective of the subtype of the
individual’s infection.
Mann and the team plan on transitioning this work from
the lab to clinical trials. “These trials will allow us to
rigorously evaluate the effectiveness of our strategy in
a controlled setting and hopefully bring us another step
closer to making a cure a reality.”
Research is driving the development of new treatments
and could introduce a cure for HIV, but ending this
epidemic will be a global effort, requiring improved
access to preventatives, therapeutics and education for
at-risk groups. •
ABOUT THE INTERVIEWEES
Dr. Emmanuel Ho is a professor at the University of Waterloo.
His research group is interested in the development and
characterization of innovative drug delivery strategies including
nanomedicines, medical devices and biomaterials for the
treatment and prevention of HIV/AIDS, cancer and chronic wound
healing.
Dr. Jamie Mann is a senior lecturer in vaccinology and
immunotherapy at the University of Bristol. His research centers
around the development of novel therapeutic and prophylactic
vaccines against mucosal pathogens such as HIV and Influenza.
DRUG DISCOVERY: INNOVATIONS, CHALLENGES AND THE FUTURE OF MEDICINE 43
TECHNOLOGYNETWORKS.COM
CONTRIBUTORS
Blake Forman
Blake pens and edits breaking news, articles and
features on a broad range of scientific topics with a
focus on drug discovery and biopharma. He holds
an honors degree in chemistry from the University of
Surrey and an MSc in chemistry from the University
of Southampton.
Joanna Owens, PhD
Joanna Owens holds a PhD in molecular
toxicology from the University of Surrey. She
has over 20 years’ experience writing about a
wide range of scientific topics in biosciences,
pharmaceuticals and biotechnology.
Kate Robinson
Kate graduated from Sheffield Hallam University
with a bachelor's degree in biomedical sciences in
2020. She joined the editorial team at Technology
Networks in 2021.
Molly Coddington
Molly Coddington is a senior writer and newsroom
team lead for Technology Networks. She holds a
first-class honors degree in neuroscience. In 2021
Molly was shortlisted for the Women in Journalism
Georgina Henry Award.
Neeta Ratanghayra, MPharm
Neeta Ratanghayra holds a Master’s degree in
pharmacy. She currently works as a freelance
medical writer specializing in developing content
for the pharma, biotech and healthcare industries.
Neil Versel
Neil Versel is a healthcare and life sciences
journalist, specializing in bioinformatics, information
technology, genomics, patient safety, healthcare
quality and health policy.
Molly Coddington
Molly Coddington is a senior writer and newsroom
team lead for Technology Networks. She holds a
first-class honors degree in neuroscience. In 2021
Molly was shortlisted for the Women in Journalism
Georgina Henry Award.
Sarah Whelan, PhD
Sarah joined Technology Networks in 2022 after
completing a PhD in cancer biology, where
her research focused on the development of
colon cancers.
Sponsored by




Download the eBook for FREE Now!
*This also covers the processing of my personal data provided above for these purposes, including profile building to better tailor the communication to my personal interests based on my previous and predicted choices. Learn More
You can withdraw your consent at any time with effect for the future. This does not affect the lawfulness of the processing up to the time of withdrawal.
Further information on the processing of personal data can be found in the Sartorius Privacy Notice.
Technology Networks may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
You can withdraw your consent at any time with effect for the future. This does not affect the lawfulness of the processing up to the time of withdrawal.
Further information on the processing of personal data can be found in the Sartorius Privacy Notice.
Technology Networks may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.