You are on page 1of 8

DEVELOPMENTAL THERAPEUTICS—MOLECULARLY TARGETED AGENTS AND TUMOR BIOLOGY

Taking Aim at the Undruggable


Niamh Coleman, MD, PhD, and Jordi Rodon, MD, PhD

The term “undruggable” is used to describe a protein that is not pharmacologically capable of being targeted;
overview

recently, however, substantial efforts have been made to turn these proteins into “druggable” targets. Thus,
“difficult to drug” or “yet to be drugged” are perhaps more appropriate terms. In cancer, a number of elusive
targets fall into this category, including transcription factors such as STAT3, TP53, and MYC. Pharmaco-
logically targeting these intractable proteins is now a key challenge of modern drug development, requiring
innovation and the development of new technologies. In this article, we discuss some of the recent technologic
and pharmacologic advances that have underpinned the erosion of the concept of undruggability. We describe
recent successes in drugging the undruggable RAS (KRAS G12C and HRAS), and discuss the advances that
have led to the validation of further targets previously believed to be undruggable, such as HIF-2α, BCL-2,
MDM2, and MLL. Finally, we look to the future and describe important advances that are likely to have a major
impact on targeting undruggable targets, such as the advent of proteolysis-targeting chimeras and protein-
protein modulators, which are leading to considerable excitement surrounding the development of cancer
targets.

CONCEPTS development risks, and patient convenience, that


If we look at the relationship among the human pro- enable the development of a drug candidate.
teome, diseases, and drugs (approved or in develop- In cancer, some of these disease-modifying targets
ment), it is striking to realize that only between 1% and seem difficult to address; they seem “undruggable”
2% of disease-modifying proteins are known, or pre- because they lack accessible deep hydrophobic
dicted, to be druggable. In therapeutics, the most pockets where ligands and drugs can bind, or they lack
frequent drug target is catalytic sites of proteins, but enzymatic activity (have no active site), referring to the
there are other entities that could be modulated for loss of function of the target. In cancer, the classic
therapeutic intent, such as nucleic acids (and nucleic undruggables include β-catenin, RAS, and transcrip-
acid complexes [e.g., ribosomes]), intracellular small tion factors (e.g., STAT3, TP53, MYC).2 Some recent
molecules, and protein-protein or protein–nucleic acid technologic advances have eroded the concept of
interactions. Druggability,1 then, can be defined as the undruggability, including structural biology advances
property of a target for being known, or predicted, to (protein nuclear magnetic resonance and crystal
bind with high affinity to a drug and modulate its structures), three-dimensional annotated chemical li-
function with a therapeutic benefit. There are several braries, and novel computational methods to visualize
methods to predict target druggability, based on em- or predict aspects of molecular targets and in-
pirical analyses of libraries of compounds or based on teractions. Other advances in pharmacology have
structural analysis. “Drug likeness” refers to the contributed as well, including expanded reengineered
compounds that bind the target. The concept includes therapeutic antibodies and biologics, strategies to
enable the selective degradation of specific proteins,
the properties that enable pharmaceutical attributes of
and improved delivery systems for nucleic acid–based
Author affiliations a viable therapeutic entity, such as structural, physi-
compounds.2 In this article, we review the later de-
and support cochemical (e.g., chemical stability, solubility), phar-
information (if velopments, addressing how some new molecular
macokinetic (e.g., bioavailability, distribution), and
applicable) appear entities show encouraging signs that they will change
at the end of this
toxicity characteristics. Related to druggability (of the
the landscape of undruggability.
article. target) and drug likeness (drug candidate) is the
Accepted on March concept of “developability,” which considers the whole HISTORICAL PERSPECTIVE
24, 2021, and process, from target discovery and lead compound In the early 20th century, drug discovery was initially
published at development to its clinical entry. In addition to the based on random screening for active substances from
ascopubs.org on May
14, 2021: DOI https://
preclinical pharmacokinetic/pharmacodynamic prop- biologic extracts (e.g., plants, microorganisms). For
doi.org/10.1200/ erties, it encompasses factors, such as physiochemical cancer drug development, the attention initially was
EDBK_325885 properties, formulation, performance, manufacturability, focused on cytotoxic agents aiming at DNA replication,

2021 ASCO EDUCATIONAL BOOK | asco.org/edbook e145

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.
Coleman and Rodon

available. Biologics, in contrast, can interact with larger


surfaces; however, because of their larger molecular size,
PRACTICAL APPLICATIONS
they have limited delivery, requiring intravenous treatment
• Recent successes in targeting KRAS G12C and and targeting only extracellular targets (e.g., EGFR, HER2,
HRAS represent significant advances in drug-
VEGFR2, PD-1). In cancer, many validated drug targets are
ging “the undruggable” and provide proof of
druggable with small molecules or antibodies: serine/threonine/
concept.
tyrosine kinases, growth factor receptors, and receptor ligands.
• Pharmacologic targeting of intractable proteins Other classes of cancer molecular targets seem difficult to
is now a key challenge of modern drug devel-
address by either of these two classes of agents: the so-called
opment, requiring innovation and the devel-
undruggable targets.
opment of new technologies.
• Important advances that are likely to have New Technologies and Pharmacologic Entities Enable
a major impact on targeting undruggable tar- Drugging the Undruggable
gets include the advent of proteolysistargeting Along with tumor heterogeneity and drug-resistance
chimeras and protein-protein modulators,
mechanisms, developing therapeutics for apparently in-
which are leading to considerable excitement in
the field of drug development. tractable targets is one of the key challenges of targeted
therapeutics. Novel chemical entities are now being de-
veloped that may change the tide. Some examples of
mitotic machinery, or DNA integrity, targeting, for example, chemical entities, beyond the classic inhibitors of kinases,
dihydrofolate reductase and topoisomerase. The advent of include protein-protein interaction (PPI) modulators, mul-
molecular biology fueled efforts to seek drugs directed tifunctional small molecules (e.g., proteolysis-targeting
against relevant molecular targets, such as growth factor chimeras [PROTACs]), peptides/peptidomimetics, nucleic
receptors, enzymes, or other biologic targets driving the acid–based therapeutics (e.g., oligonucleotides), and even
disease. Alongside the development of targeted therapies, cell therapies and T-cell receptor (TCR) mimetics. This
in the 1970s the pharmacologic industry moved toward major expansion of the “chemical matter” (chemical space
a rational drug discovery methodology of small molecules. used in drug discovery) used for therapeutics brings along
Marked advancements in the experimental and computa- new chemical, biologic, and pharmaceutical properties that
tional techniques used in the drug discovery process have could enable the modulation of difficult targets. Because of
changed the development of small molecules and anti- some of the characteristics of these compounds, most of
bodies, moving from an empirical screening toward these entities may require intravenous delivery; however,
a structured rational design. With the parallel development nanomedicine approaches could reduce dosing frequency,
of recombinant protein-based therapeutic agents in the while improving therapeutic windows.
1980s, rational development became the standard practice. Of these novel compound types, one notable recent success
Thus, the modern drug development paradigm was born.3 is venetoclax, a first-in-class BCL-2 inhibitor. It took 20 years
When considering the class of drugs approved by the U.S. of extensive chemistry efforts,4,5 several lead candidates,
Food and Drug Administration for management of cancer and many clinical trials, but its approval by the U.S. Food
under this paradigm, the vast majority fall into two cate- and Drug Administration in 2016 demonstrated the possi-
gories: small-molecule drugs (those with composite mo- bility of developing agents that impede PPIs with small
lecular mass , 1,000 daltons) and protein therapeutics or molecules. Since that time, BCL-2 is no longer on the list of
biologics (primarily monoclonal antibodies, some peptides, undruggables.
and a few vaccines). Until now, biologics have been The recent successes with targeting KRAS G12C and HRAS
designed to bind to specific target epitopes in extracellular are also important breakthroughs in targeting the undrug-
or cell surface targets, whereas small chemical entities are gable. The three RAS oncogene products, KRAS, NRAS,
intended to access and modulate intracellular proteins. and HRAS, have been among the most interesting, sought
Typically, small molecules bind to accessible hydrophobic after, and challenging targets, in part because of the fre-
invaginations in the target, more frequently in the catalytic quency of these lesions (approximately 30% of human
site (also known as catalytic inhibitors; e.g., erlotinib, cancers). In the case of KRAS G12C, the breakthrough
sunitinib, or copanlisib), as well as in other sites, “an al- came from structural insights provided by x-ray crystallog-
losteric site,” but altering the shape of the active site so that raphy and nuclear magnetic resonance that showed that
the target can no longer bind to its substrate (“allosteric this specific mutation creates a pocket that can be exploited
inhibitors” such as everolimus or trametinib). Although it therapeutically. Using state-of-the-art chemistry, pharma-
depends on their size and chemical properties, most small cologists have been able to synthesize covalent inhibitors
molecules developed as anticancer agents are orally that attach to Cys12, thereby inhibiting MAPK signaling.

e146 2021 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.
Taking Aim at the Undruggable

Sotorasib is one such small-molecule inhibitor, and its use utilizing endogenous biologic mechanisms, such as the
has resulted in confirmed objective response rates (com- ubiquitin-proteasome degradation system: the therapeutic
plete or partial response) of 32.2% in patients with KRAS effect is produced by modulating protein levels of the target.
G12C non–small cell lung cancer, as well as disease control PROTEOLYSIS-TARGETING CHIMERAS
rates (objective response or stable disease) of 88.1% in
non–small cell lung cancer and 73.8% in colorectal can- PROTACs are one class of bifunctional small molecules.
cer.6 Consequently, several additional compounds are in They simultaneously bind to a target protein and an
clinical development. E3 ubiquitin ligase, thereby causing ubiquitylation and
degradation of the target protein (Fig. 1).9 The ability of
In the case of HRAS, the breakthrough came from the PROTACs to degrade proteins, regardless of their function,
observation of the exquisite dependency of HRAS on far- makes this approach highly attractive, especially for those
nesyl transferase for prenylation, a critical step that is targets for which compounds can be developed that bind to
necessary for RAS to bind to the inner layer of the cell a given target without inhibiting its activity. Degradation of
membrane and function (other members of the RAS family the target protein by PROTACs is also suitable for targets
have redundant pathways). Tipifarnib, a farnesyl transferase that overcome the effect of an inhibitor by overexpression,
inhibitor, has been granted fast track designation by the which is often seen in cancer. As such, targeted protein
U.S. Food and Drug Administration for the treatment of degradation offers tremendous promise. In fact, PROTACs
patients with HRAS-mutant head and neck squamous cell have a number of advantages; because of their unique
carcinomas. Early clinical trials are showing an overall re- mechanism of action, a PROTAC molecule is capable of
sponse rate of 50.0% (95% CI, 26.0–74.0) and a median catalyzing the degradation of multiple molecules of the
duration of response of 14.7 months (95% CI, 2.1–not protein of interest. Consequently, compared with small-
reported) in patients with these diseases.7 molecule inhibitors, PROTACs may require significantly
Another recent success story related to the use of modern lower concentrations to elicit a desired pharmacologic ef-
crystallography and structure-based design is the targeting fect. PROTACs also have the potential to target so-called
of HIF-2α. von Hippel-Lindau disease and many clear cell undruggable proteins, such as transcription factors. For
renal cell carcinomas have inactivating mutations in VHL, example, STAT3 is a key factor for cell survival, proliferation,
resulting in constitutive activation of the HIF-2α transcrip- angiogenesis, metastasis, and chemotherapy resistance;
tion factor. The finding of a large cavity within the HIF-2α thus, blocking STAT3 activity is an attractive strategy that
PAS-B domain permitted the development of molecules has been pursued for many years, with little success.10
(e.g., PT2399, PT2385, and PT2977, later known as MK- However, PROTACs targeting STAT have been recently
6482) that bind to the allosteric site and block its di- reported,11,12 opening new possibilities.
merization with the HIF-1α/2α transcriptional dimerization PROTACs can be used to overcome drug resistance re-
partner ARNT/HIF-1β. Treatment of 61 patients with VHL- sulting from mutations of a protein of interest, such as
associated clear cell renal cell carcinoma using MK-6482 PROTACs targeting mutated forms of BCR-ABL,13 receptor
(NCT03401788) showed a confirmed response rate of tyrosine kinases,14 estrogen receptor-α,15 and Bruton ty-
27.9%, as well as 13.1% unconfirmed responses at the time rosine kinase.16 PROTACs can overcome resistance to
of reporting (86.9% of patients experienced some decrease small-molecule inhibitors resulting from target upregulation
in the size of their target lesions).8 This was the first story of by degrading the target; one such example is androgen
successful drug development against a transcription factor. receptor degraders, which have recently overcome re-
sistance developed to the androgen receptor antagonist
Some other programs are aiming to develop multispecific
enzalutamide during prostate cancer treatment.17
drugs. These compounds work through two or more
chemical entities (one effector and one target), with several Although the field of PROTAC research is still relatively
drug-target binding interfaces that work sequentially or new, these rapid exciting clinical developments, such as
concurrently. This multispecificity allows limiting drug ac- PROTAC-mediated targeting of androgen receptors, dem-
tivity to a specific location or anchors the target close to an onstrate proof of principle, proving that this approach is
endogenous effector, allowing the effector to modulate the broadly applicable. Numerous proteins are now reported to
target. Some of them are antibody/peptide-based, such as have been degraded in this manner, including Bruton ty-
heteroduplex immunoglobulin G, bispecific CD3 engagers, rosine kinase, BCR-Abl, FKBP12, BRD9, and CDK6.18
or antibody-toxin, antibody-drug, or antibody-cytokine fu- One of the most exciting and compelling aspects of
sion proteins. Others are small molecules called “match- unlocking the vast potential of protein degraders is the
makers,” which pull two entities together (the effector and opportunity to bring the wide variety of the historically
target), such that one (the effector) acts upon the other (the undruggable proteome into play for therapeutic benefit. In
target). These act as chemical inducers of degradation spite of these promising prospects, there are challenges in

2021 ASCO EDUCATIONAL BOOK | asco.org/edbook e147

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.
Coleman and Rodon

FIGURE 1. General Mechanism of Proteolysis-Targeting Chimera–Induced Degradation of the Protein of Interest


A proteolysis-targeting chimera molecule recruits an E3 ligase to a protein of interest, followed by polyubiquitin of the latter by an E2-conjugating enzyme.
The polyubiquitinated protein of interest is recognized by the proteasome for its degradation. The proteolysis-targeting chimera molecule is recycled to
induce the next round of degradation.

utilizing PROTAC molecules, such as attaining suitable also typically large, larger than that of the receptor-ligand
physicochemical properties; adequate absorption, distri- contact area,26,27 and is highly hydrophobic.24,26 In addition,
bution, metabolism, and excretion properties for oral dosing; the amino acid residues involved in PPIs result in high-
and/or even achieving adequate central nervous system affinity binding between the proteins, making it more
exposure.18 Therefore, overcoming some of these phar- challenging for small molecular compounds to inhibit.28
macologic obstacles will undoubtedly be essential in paving Compared with traditional drug target enzymes or re-
the way for the broader therapeutic potential of PROTAC ceptors, PPIs also lack endogenous ligands,28 and com-
molecules. pared with traditional small-molecule drugs, drugs acting on
Small Molecules Modulating Protein-Protein Interactions PPIs have a higher molecular weight (. 400 daltons).

Critical biologic processes, such as DNA replication, tran- In spite of these obstacles, progress is being made. In recent
scription, translation, and transmembrane signal trans- years, some PPI modulators have entered clinical studies;
duction, rely on functional specific proteins that are some have been approved for marketing, indicating that the
regulated through protein complexes and controlled via modulators targeting PPIs may have broad prospects. In
PPIs19,20; aberrant PPIs are associated with many human comparison with peptides targeting PPIs, small-molecular
cancers.21 In contrast with small molecules targeting inhibitors of PPIs are increasingly appealing, because these
protein-ligand interactions (found in enzymes, ion channels, drugs tend to have lower research costs, oral preparations,
or receptors), targeting PPIs, previously regarded as and better tumor microenvironment penetration.
undruggable targets, is a more challenging and novel Inhibitors of MDM2/p53 interaction (small molecules,
therapeutic approach.22 peptides) p53 regulates the cell cycle and functions as
Designing a small molecule to bind to a PPI interface has a tumor suppressor. Almost 50% of human cancers have
proven to be difficult for a number of reasons. First, the alterations in the TP53 gene, which results in the in-
unique interface structure is a challenge for drug design: activation of p53 function or loss of p53 expression.29 The
compared with the binding pockets of conventional protein MDM2 protein is the p53 E3 ubiquitin ligase that, under
targets, the interface of PPIs tends to be flat and contains cellular stress conditions, ubiquitinates p53 and facilitates
few pockets, making it difficult for small-molecule com- p53 nuclear export and inhibition of transcription activity
pounds to bind.23-25 The interface area of the interaction is or ubiquitin-dependent proteasomal degradation of p53.

e148 2021 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.
Taking Aim at the Undruggable

Utilizing small molecules or peptides to block the p53/ with CDKN2A (cyclin-dependent kinase inhibitor). MTAP-
MDM2 interaction is one way to restore the p53 pathway in null cancer cells seem to have a dependency on PRMT5 (a
wild-type p53 tumors, thereby preventing the inactivation of methyltransferase); therefore, inhibition of PRMT5 could be
p53. Nutlins are the most well-characterized small mole- exploited for cancer treatment. In fact, PRMT5 inhibitors are
cules that disrupt this p53-MDM2 interaction, binding to the in clinical development.38 Based on this hypothesis, MTAP/
N-terminal pocket of MDM2 and stabilizing p53, which CDKN2A-null tumors could be sensitive to PRMT5 inhibitors
helps to retain tumor-suppressive functionality. There is while normal cells are spared, offering a wide therapeutic
extensive in vitro and in vivo evidence to demonstrate that window. Other examples of this paradigm include the 1p36
nutlins increase p53 levels and apoptosis, as well as de- deletion (which includes tumor-suppressor genes, such as
crease tumorigenicity.30 CAMTA1, mir34A, or KIF1B), the passengers enolase 1 or 6-
Building on this momentum, the nutlin derivative RG7112 phosphogluconate dehydrogenase, and the therapeutic
has advanced to clinical trials for leukemia and sarcomas inhibition of enolase 2 or components of the nonoxidative
along with a number of other small molecules with similar pentose phosphate shunt.39
mechanisms of action that are currently in phase I clinical Another way to modulate an undruggable protein with small
trials, including SAR405838, CGM097, HDM201, DS- molecules is to modulate its transcription levels: either in-
3032, MK-8242, and AMG232.31-33 Thus, there is consid- directly, by targeting chromatin regulation, or directly, by the
erable clinical interest and activity focused on inhibiting targeting of transcription factors or RNA that regulates its
p53-protein interactions using small molecules. Another expression. There are ongoing drug-discovery efforts fo-
target being modulated by the use of PPI is the interaction of cusing on small-molecule drugs targeting transcription
MLL1 and menin. Once the crystal structure of menin was factors, such as P53, MYC, and STAT3. These transcription
available, the binding site with MLL became clear; currently, factors, which are frequently altered in cancer, form protein
drugs such as KO-539 show promising clinical activity in complexes that are directed to specific sites on DNA and
refractory acute myeloid leukemia.34 regulate oncogenic events. Small molecules interacting with
Other Approaches Using Small Molecules MYC, STAT3, or P53 are in development. These agents
mostly aim to prevent the formation of these multiprotein
An indirect approach to targeting challenging and difficult complexes, rather than to disrupt the transcription factor–
targets is the search for “synthetic”35,36 and “collateral”37 DNA interaction.2
lethality scenarios. In synthetic lethality, cancer-specific
mutations that impair the function of one gene (e.g., Targeting chromatin regulation, the main goal of epige-
BRCA1) can be exploited with pharmacologic inhibition of netics, has also seen tremendous development with the
another key component of the pathway (e.g., inhibiting expansion of technologies that allow analysis of the epi-
PARP with olaparib), which would lead to the death of genome and drug discovery now aiming for new targets,
cancer cells with the genetic mutation but spare normal such as EZH2, LSD1, PRMT inhibitors, and others. How-
cells that lack the genetic vulnerability. In addition to the ever, the field of epigenetics drug development is beyond
approval of many PARP inhibitors for the management of the scope of this review and can be found elsewhere.40
BRCA1/2 ovarian and breast cancers, current clinical trials There is a conventional view that it is not possible to bind
are exploring this paradigm by using these agents in tumors RNAs with small molecules because they present unrelated
with alterations in other DNA-repair enzymes, such as structures and complex pockets for druggability (RNA
PALB2, ATM, and RAD51. Synthetic lethality is also ob- interference–based therapeutics are discussed in the next
served in tumors bearing mutations in the SWI/SNF chro- section). However, it has since been discovered that RNA
matin remodeling complexes (e.g., SMARCA2, SMARCA4, molecules actually adopt secondary and tertiary structures
ARID1A, and ARID1B); tazemetostat, an EZH2 inhibitor, that could be attractive targets for small molecules. In fact,
has recently been approved by the U.S. Food and Drug many drugs, including several antibiotics (e.g., amino-
Administration for the management of malignant rhabdoid glycosides) bind bacterial RNA in the ribosomes. This field
tumors, a disease driven by SMARCA4 mutations. changed in 2020, when the U.S. Food and Drug Admin-
The term “collateral lethality” is preferred to synthetic le- istration approved risdiplam, an SMN2-splicing modifier, to
thality when a “passenger” gene mutation (alteration not treat spinal muscular atrophy. This is the first drug designed
playing an active role in tumor progression) is lost along with specifically against a particular messenger RNA (it modifies
an adjacent oncogenic or “driver” alteration (in our case, an the splicing of SMN2 messenger RNA, resulting in an in-
undruggable tumor-suppressor gene). Using the lack of crease in the concentration of the functional SMN protein
function of passenger-deleted genes for a therapeutic ad- in vivo).41 The development of anticancer small molecules
vantage could exploit cancer-selective vulnerabilities. Such that specifically bind RNA, although still in its early stages,
is the case with MTAP, a gene that is frequently codeleted aims to design similar small-molecule RNA modulators that

2021 ASCO EDUCATIONAL BOOK | asco.org/edbook e149

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.
Coleman and Rodon

bind RNA molecules and change protein synthesis by leveraged for this approach include those arising from
modifying RNA processing (RNA splicing) or impeding cancer mutations (e.g., in otherwise undruggable proteins),
protein production.42 known as neoantigens, and tumor-associated antigens,
which result from overexpressed genes. This principle,
NEW BIOLOGICS CAN ALSO ENABLE THE IMPOSSIBLE
which is the basis for modern cell therapy (e.g., CAR T cells,
Despite efforts to modulate RNA and protein expression with engineered TCR therapy) is now also being replicated by
small molecules, described above, biologic molecules, such different types of peptides (TCR “mimetics”)43: TCR-like
as antisense technologies, have remained the most com- antibodies and TCR fusion proteins. These TCR mimetics
monly deployed method to target disease-associated RNAs. are advantageous in that they can have more drug-like
First-generation oligonucleotide structures had poor cell properties and avoid some of the limitations of traditional
permeability and distribution and triggered immunoreac- cell therapies (e.g., prolonged and expensive production
tivity; as a result of their poor pharmacology, their devel- times, cost). In addition to CD19, which was the first target
opment languished for years. Now, these initial problems of for leukemias (tisagenlecleucel, the first CAR T-cell therapy
RNA therapeutics have been addressed with improvements was approved for acute lymphoblastic leukemia in 201744),
in trigger design, sequence selection (to avoid unintended in solid tumors, initial research with cell therapy and TCR
off-target RNA interference effects), chemical formulation mimetics initially focused on tumor-associated antigens,
(to avoid degradation by endonucleases and exonucleases), such as GP100, MAGEA4, NYESO1, and PRAME; the first
and delivery mechanisms (lipid nanoparticles and others), generation of studies is ongoing. Theoretically, neoantigen-
enabling a second generation of drugs. Consequently, in specific CAR T cells or TCR mimetics (antibodies or soluble
2018, the U.S. Food and Drug Administration approved TCRs) that are specific for the peptide–major histocom-
patisiran, a small interfering RNA for the treatment of he- patibility complex could be a promising strategy to tackle
reditary transthyretin amyloidosis with polyneuropathy. repetitive mutated variants in undruggables, such as fusion
Since then, two more drugs of this kind, lumasiran and peptides in sarcoma (breakpoint-specific neoantigens have
givosiran, have been approved. Patisiran, which primarily been predicted and validated, in part, for EWSR1-FLI1,
acts on the liver, is not a metabolically stabilized small PAX3-FOXO1, and SS18-SSX1)45,46 or KRAS in pancreatic
interfering RNA, but it uses a lipid nanoparticle delivery cancer.47
formulation. More recently, small interfering RNA thera-
The clinical proof of concept for cell therapy came from
peutics have been metabolically stabilized to improve their
targeting CD19.48 For TCR mimetics, this has likely been
stability.
achieved with the recently presented data on tebentafusp
However, not all RNA transcripts transmit a message for (IMCgp100) that showed clinical benefit, including target
protein coding and translation, but there is a diverse range of lesion reduction.49 Tebentafusp is a solution of soluble TCRs
noncoding RNA molecules (e.g., long noncoding RNAs, stabilized by a disulfide bond and fused to an anti-CD3 scFv
microRNAs, small nuclear and small nucleolar RNAs) with (a so-called “ImmTAC molecule”). Early data from a phase II
broad roles in modulating gene expression, including trial in patients with HLA-A*0201+ gp100+ metastatic uveal
cancer deregulation. Because of recent developments in melanoma showed that, although the overall response rate
the field, RNA therapeutics are not limited to antisense by response evaluation criteria in solid tumors was low (5%),
oligonucleotides; investigators are exploring small in- it was accompanied by a reduction in target lesion in 44% of
terfering RNA, microRNA, messenger RNA, RNA aptamers, patients. The median overall survival was 16.8 months
short activating RNA, and single guide RNA for CRISPR/ (95% CI, 12.9–21.3), which seems promising compared
Cas9 systems as potential therapeutic platforms. Some of with historical controls.50
these novel RNA therapeutics are also aiming for some
undruggable targets that are relevant in cancer, such as CONCLUSION
EWS/FLI1 (Ewing sarcoma), KRAS G12D (pancreatic can- There is considerable excitement surrounding the devel-
cer), miR-155 (lymphoma and leukemias), and miR-16 opment of cancer targets considered undruggable, with
(malignant pleural mesothelioma and non–small cell lung classic classes of agents, including transcription factors,
cancer). such as STAT3, TP53, MYC. Others, like RAS (KRAS G12C
One final approach using biologics to target complicated and HRAS), HIF-2α, BCL-2, MDM2, and MLL, are now
intracellular targets involves the TCR-HLA (human leuko- validated druggable targets.
cyte antigen) system. The target, processed in the immu- Because of the possibility that many other pharmacologic
noproteasome, is broken into different peptides that are barriers might be overcome by the new entities described
presented by the tumor cell’s HLA system to the immune here, the concept of undruggable is being challenged, and
system via the TCR; this interaction activates the immune some now consider it an inappropriate or démodé term. In
system against the tumor cell. Antigens that can be the presence of novel molecular entities that promise to

e150 2021 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.
Taking Aim at the Undruggable

become game changers, some advocate for using terms ACKNOWLEDGMENT


such as “undrugged,” “difficult-to-drug targets,” or “targets Editorial assistance was provided by Dr. Paul Fletcher and
with developability challenges” rather than undruggable. Daley Drucker (Moffitt Cancer Center’s Scientific Editing
Time, and the efforts of a new generation of drug de- Department). No compensation was provided beyond their
velopers, will tell. regular salaries.

AFFILIATION Holcombe Blvd., FC8.3052, Houston, TX 77030; email: jrodon@


Division of Cancer Medicine, Department of Investigational Cancer mdanderson.org.
Therapeutics, The University of Texas MD Anderson Cancer Center,
Houston, TX
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
AND DATA AVAILABILITY STATEMENT
Disclosures provided by the authors and data availability statement (if
CORRESPONDING AUTHOR applicable) are available with this article at DOI https://doi.org/10.1200/
Jordi Rodon, MD, PhD, Department of Investigational Cancer EDBK_325885.
Therapeutics, The University of Texas MD Anderson Cancer, 1400

REFERENCES
1. Abi Hussein H, Geneix C, Petitjean M, et al. Global vision of druggability issues: applications and perspectives. Drug Discov Today. 2017;22:404-415.
2. Lazo JS, Sharlow ER. Drugging undruggable molecular cancer targets. Annu Rev Pharmacol Toxicol. 2016;56:23-40.
3. Martin-Liberal J, Hierro C, Ochoa de Olza M, et al. Immuno-oncology: the third paradigm in early drug development. Target Oncol. 2017;12:125-138.
4. DeGoey DA, Chen HJ, Cox PB, et al. Beyond the rule of 5: lessons learned from AbbVie’s Drugs and Compound Collection. J Med Chem. 2018;61:2636-2651.
5. Mullard A. Pioneering apoptosis-targeted cancer drug poised for FDA approval [published correction appears in Nat Rev Drug Discov. 2016;15:292]. Nat Rev
Drug Discov. 2016;15:147-149.
6. Hong DS, Fakih MG, Strickler JH, et al. KRASG12C inhibition with sotorasib in advanced solid tumors. N Engl J Med. 2020;383:1207-1217.
7. Ho AL, Hanna GJ, Scholz CR, et al. Preliminary activity of tipifarnib in tumors of the head and neck, salivary gland and urothelial tract with HRAS mutations. J Clin
Oncol. 2020;38(15_suppl):6504.
8. Jonasch E, Donskov F, Iliopoulos O, et al. Phase II study of the oral HIF-2α inhibitor MK-6482 for von Hippel-Lindau disease–associated renal cell carcinoma.
J Clin Oncol. 2020;38(15_suppl):5003.
9. Lai AC, Crews CM. Induced protein degradation: an emerging drug discovery paradigm. Nat Rev Drug Discov. 2017;16:101-114.
10. Redell MS. A STAT3 decoy lures AML out of hiding. Blood. 2016;127:1628-1629.
11. Bai L, Zhou H, Xu R, et al. A potent and selective small-molecule degrader of STAT3 achieves complete tumor regression in vivo. Cancer Cell. 2019;36:498-511.
e17.
12. Zhou H, Bai L, Xu R, et al. Structure-based discovery of SD-36 as a potent, selective, and efficacious PROTAC degrader of STAT3 protein. J Med Chem. 2019;
62:11280-11300.
13. Zhao Q, Ren C, Liu L, et al. Discovery of SIAIS178 as an effective BCR-ABL degrader by recruiting von Hippel-Lindau (VHL) E3 ubiquitin ligase. J Med Chem.
2019;62:9281-9298.
14. Deng J. How to unleash mitochondrial apoptotic blockades to kill cancers? Acta Pharm Sin B. 2017;7:18-26.
15. Gonzalez TL, Hancock M, Sun S, et al. Targeted degradation of activating estrogen receptor α ligand-binding domain mutations in human breast cancer
[published correction appears in Breast Cancer Res Treat. 2020;180:623]. Breast Cancer Res Treat. 2020;180:611-622.
16. Buhimschi AD, Armstrong HA, Toure M, et al. Targeting the C481S ibrutinib-resistance mutation in Bruton’s tyrosine kinase using PROTAC-mediated deg-
radation. Biochemistry. 2018;57:3564-3575.
17. Kregel S, Wang C, Han X, et al. Androgen receptor degraders overcome common resistance mechanisms developed during prostate cancer treatment. Neoplasia.
2020;22:111-119.
18. Chamberlain PP, Hamann LG. Development of targeted protein degradation therapeutics. Nat Chem Biol. 2019;15:937-944.
19. Rual JF, Venkatesan K, Hao T, et al. Towards a proteome-scale map of the human protein-protein interaction network. Nature. 2005;437:1173-1178.
20. Stelzl U, Worm U, Lalowski M, et al. A human protein-protein interaction network: a resource for annotating the proteome. Cell. 2005;122:957-968.
21. Nero TL, Morton CJ, Holien JK, et al. Oncogenic protein interfaces: small molecules, big challenges. Nat Rev Cancer. 2014;14:248-262.
22. Santos R, Ursu O, Gaulton A, et al. A comprehensive map of molecular drug targets. Nat Rev Drug Discov. 2017;16:19-34.

2021 ASCO EDUCATIONAL BOOK | asco.org/edbook e151

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.
Coleman and Rodon

23. Buchwald P. Small-molecule protein-protein interaction inhibitors: therapeutic potential in light of molecular size, chemical space, and ligand binding efficiency
considerations. IUBMB Life. 2010;62:724-731.
24. Arkin MR, Wells JA. Small-molecule inhibitors of protein-protein interactions: progressing towards the dream. Nat Rev Drug Discov. 2004;3:301-317.
25. Dı́az-Eufracio BI, Naveja JJ, Medina-Franco JL. Protein–protein Interaction Modulators for Epigenetic Therapies. In Donev R, (ed). Advances in Protein Chemistry
and Structural Biology. Cambridge, MA: Academic Press; 2018;65-84.
26. Smith MC, Gestwicki JE. Features of protein-protein interactions that translate into potent inhibitors: topology, surface area and affinity. Expert Rev Mol Med.
2012;14:e16.
27. Cheng AC, Coleman RG, Smyth KT, et al. Structure-based maximal affinity model predicts small-molecule druggability. Nat Biotechnol. 2007;25:71-75.
28. Ivanov AA, Khuri FR, Fu H. Targeting protein-protein interactions as an anticancer strategy. Trends Pharmacol Sci. 2013;34:393-400.
29. Feki A, Irminger-Finger I. Mutational spectrum of p53 mutations in primary breast and ovarian tumors. Crit Rev Oncol Hematol. 2004;52:103-116.
30. Vassilev LT, Vu BT, Graves B, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004;303:844-848.
31. Sun D, Li Z, Rew Y, et al. Discovery of AMG 232, a potent, selective, and orally bioavailable MDM2-p53 inhibitor in clinical development. J Med Chem. 2014;
57:1454-1472.
32. Zhao Y, Bernard D, Wang S. Small molecule inhibitors of MDM2-p53 and MDMX-p53 interactions as new cancer therapeutics. BioDiscovery. 2013;8:4.
33. Wang S, Sun W, Zhao Y, et al. SAR405838: an optimized inhibitor of MDM2-p53 interaction that induces complete and durable tumor regression. Cancer Res.
2014;74:5855-5865.
34. Wang ES, Altmann JK, Pettit K, et al. Preliminary data on a phase 1/2A first in human study of the menin-KMT2A (MLL) inhibitor KO-539 in patients with relapsed
or refractory acute myeloid leukemia. Blood. 2020;136(suppl 1):7-8.
35. Hartwell LH, Szankasi P, Roberts CJ, et al. Integrating genetic approaches into the discovery of anticancer drugs. Science. 1997;278:1064-1068.
36. Kaelin WG Jr. Choosing anticancer drug targets in the postgenomic era. J Clin Invest. 1999;104:1503-1506.
37. Muller FL, Aquilanti EA, DePinho RA. Collateral lethality: a new therapeutic strategy in oncology. Trends Cancer. 2015;1:161-173.
38. Rugo HS, Jacobs I, Sharma S, et al. The promise for histone methyltransferase inhibitors for epigenetic therapy in clinical oncology: a narrative review. Adv Ther.
2020;37:3059-3082.
39. Garbers C, Hermanns HM, Schaper F, et al. Plasticity and cross-talk of interleukin 6-type cytokines. Cytokine Growth Factor Rev. 2012;23:85-97.
40. Cheng Y, He C, Wang M, et al. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther.
2019;4:62.
41. Ratni H, Ebeling M, Baird J, et al. Discovery of risdiplam, a selective survival of motor neuron-2 (SMN2) gene splicing modifier for the treatment of spinal muscular
atrophy (SMA). J Med Chem. 2018;61:6501-6517.
42. Costales MG, Hoch DG, Abegg D, et al. A designed small molecule inhibitor of a non-coding RNA sensitizes HER2 negative cancers to herceptin. J Am Chem Soc.
2019;141:2960-2974.
43. Chang AY, Dao T, Gejman RS, et al. A therapeutic T cell receptor mimic antibody targets tumor-associated PRAME peptide/HLA-I antigens. J Clin Invest. 2017;
127:2705-2718.
44. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439-448.
45. Worley BS, van den Broeke LT, Goletz TJ, et al. Antigenicity of fusion proteins from sarcoma-associated chromosomal translocations. Cancer Res. 2001;
61:6868-6875.
46. Chang TC, Carter RA, Li Y, et al. The neoepitope landscape in pediatric cancers. Genome Med. 2017;9:78.
47. Sim MJW, Lu J, Spencer M, et al. High-affinity oligoclonal TCRs define effective adoptive T cell therapy targeting mutant KRAS-G12D [published correction
appears in Proc Natl Acad Sci USA. 2020;117:27743-27744]. Proc Natl Acad Sci USA. 2020;117:12826-12835.
48. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507-1517.
49. Sacco JJ, Carvajal R, Butler MO, et al. 64MO A phase (ph) II, multi-center study of the safety and efficacy of tebentafusp (tebe) (IMCgp100) in patients (pts) with
metastatic uveal melanoma (mUM). Ann Oncol. 2020;31:S1442-S1443.
50. Sato T, Nathan PD, Hernandez-Aya L, et al. Redirected T cell lysis in patients with metastatic uveal melanoma with gp100-directed TCR IMCgp100: overall
survival findings. J Clin Oncol. 2018;36(15_suppl):9521.

e152 2021 ASCO EDUCATIONAL BOOK | asco.org/edbook

Downloaded from ascopubs.org by 103.161.152.134 on May 1, 2023 from 103.161.152.134


Copyright © 2023 American Society of Clinical Oncology. All rights reserved.

You might also like