top of page

SHY's pipeline

SHY’s drug development strategy leverages the essential roles of ATPases and GTPases by designing small molecules that specifically compete for their interactions with ATP or GTP, respectively, and then modulate their function. SHY’s portfolio includes its lead oncology candidate, SHY-ONC6, an oral proteasome inhibitor (IND filing in first half of 2026),

as well as non-covalent pan-KRas inhibitors (clinical candidate designation in first half of 2026) and programs targeting bacterial and fungal pathogens. SHY retains full rights and strong IP for its portfolio.

Program

Target

In Vitro

In Vivo

Lead Designation

IND Filing

Oncology
SHY-ONC6

ATPases in the 19S Proteasome Subunit

H1 2026

Oncology

pan-KRas

H1 2026

  • SHY-ONC6: a 19S proteasome ATPase regulatory particle cancer therapeutic candidate

     

    SHY has selected SHY-ONC6 as its first clinical candidate for testing as a cancer therapeutic and is now doing the pre-clinical work to file an IND for it in late 2025. 

     

    Mechanism of Action

     

    SHY-ONC6 is a selective inhibitor of a subset of AAA+ ATPases in the 19S regulatory proteasome particle. The 19S regulatory particle is responsible for recognizing, binding, deubiquitinating, and unfolding proteins before they are degraded in the 20S proteasome core particle. By interfering with this process, SHY-ONC6 induces apoptosis.

    Three FDA approved drugs (Bortezomib, Carfilzomib, and Ixazomib) have validated the clinical and commercial importance of the proteasome ubiquitin system as a therapeutic target but they operate through a mechanism distinct from SHY-ONC6. They are D-peptide like molecules that inhibit the proteolytic beta sites of the 20S proteasome core particle, whereas SHY-ONC6 is a small molecule inhibitor of ATPases in the 19S regulatory particle.

     

    Development Status

     

    SHY-ONC6 has been selected as a clinical candidate. It shows strong in vitro activity and in vivo efficacy across a variety of difficult-to-treat human tumors. Pre-clinical development is ongoing, with an Investigational New Drug (IND) application planned for the second half of 2025. SHY intends to initiate a Phase-I trial focusing on tumors that have shown positive pre-clinical results, with plans to refine target indications in subsequent trials.

     

    In Vitro Activity

     

    SHY-ONC6 demonstrates significant in vitro activity, with IC50 values of 100 nM or less in approximately 80% of the 869 human solid and blood-borne tumor cell lines tested in PRISM studies by the Broad Institute.

     

    In Vivo Efficacy

     

    In mouse xenograft models, SHY-ONC6 shows robust oral efficacy, achieving over 100% tumor growth inhibition (regression) in a range of human-derived tumor models. These include colon, gastric, lung (mesothelioma, small cell, and non-small cell), myeloma, and pancreatic cancers, all known for poor prognoses.

     

    SHY-ONC6 also has favorable pharmacokinetic characteristics and crosses the blood brain barrier.

     

    Safety and Tolerability

     

    Extensive in vivo studies in mice have shown that SHY-ONC6 is well-tolerated at effective doses, with no observed weight loss, behavioral changes, or adverse blood chemistry changes. 

     

    Comprehensive preclinical safety studies conducted prior to SHY’s IND filing.

Fungi

H1 2026

  • Non-Covalent Pan-KRAS Inhibitors

     

    Mutations in KRas, a member of the Ras superfamily of small GTPases, are associated with approximately 30% of human tumors, making KRas mutants long-time targets for cancer treatment. Most drugs bind non-covalently to their targets. Articles published in the 1980s and 1990s, however reported affinities for GTP and KRas in the range of 10 pM and led scientists to conclude that the KRas GTP binding site could not be targeted by non-covalent inhibitors – the chemical nature of most drug/target interactions. It was deemed undruggable. 

     

    Accordingly, KRas drug development efforts by pharmaceutical and biotechnology companies focused on alternative mechanisms for inhibiting mutant KRas signaling. Over time, opinions have shifted as to whether these compounds should specifically target just the different KRas mutants or not. Several covalently bound compounds targeting KRas G12C mutations (representing a meaningful but still relatively small number of patients) had impressive initial clinical effects, were approved by the FDA for sale, but, disappointingly, have proven in post-approval clinical use to have limited durations of effect. 

    Reasoning analogously based on his experience developing small molecule inhibitors of ATP binding to receptor tyrosine kinases, Dr. Hadari founded SHY with the expectations that:

    • previously reported affinity measurements for GTP/KRas binding were erroneous; 

    • small molecules could therefore be synthesized that compete non-covalently for GTP binding to KRas; 

    • such small molecules would be more therapeutically effective if they were not specific to just certain KRas mutants, but rather preferentially to KRas and also to other members of the protein superfamily of which KRas is one member; and, finally, 

    • notwithstanding the importance of KRas signaling to sustaining cellular functions, such non-covalently bound pan KRas inhibitors would not necessarily be toxic at therapeutically relevant doses.

    SHY has validated all these hypotheses and succeeded where others have not tried by utilizing state-of-the-art methods and combining proprietary high throughput screens with computationally tailored libraries and some inspired SAR.  

    • SHY conducted a series of measurements of GTP/KRas binding using more contemporary methods than the articles published in the 1980s. In 2020 we posted affinity measurements (Kd’s) for GTP/KRas and KRas mutant binding in the range of 250 nM on bioRxiv, Cold Spring Harbor Laboratory’s preprint server for biology. This Kd is approximately 10,000 times lower than the estimates published in the 1980s and 1990s. 

    • SHY has synthesized over 1,000 small molecules that compete non-covalently and differentiably for GTP for binding to KRas and other members of the KRas superfamily, and differentiably modulate KRas signaling. SHY employs a proprietary fluorescence-based cell-free assay to confirm competitive GTP binding. The inhibition of KRas signaling is further assessed in tumor-derived cell lines expressing both wild-type and mutant KRas.
       

    • Select molecules from this portfolio are undergoing further in vitro, pharmacokinetic, and in vivo xenograft studies to evaluate their potential as clinical candidates. To date, as anticipated, in animal studies they have shown anti-tumor efficacy but not systemic toxicity.

    A newly published study in Molecular Cancer Therapeutics, with research led by SHY scientists, demonstrates SHY’s differentiated approach to RAS inhibition, providing the first evidence that the GTP-binding site of RAS proteins can be competitively targeted by small molecules in a non-covalent manner—a mechanism not employed by current clinical agents.

    Additional supplementary data: Identification of Small-Molecule Inhibitors that Block the GTP-Binding Pocket of K-Ras and Other Members of the Ras Superfamily of Small GTPases

  • SHY has been awarded a Small Business Innovation Research grant from the National Institute of Allergy and Infectious Diseases of The National Institutes of Health. The funding will support SHY’s development of an early-stage anti-fungal program, which has already show promising in vitro activity against fungal strains of public health concern. 

     

    The research is supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under Award Number R43AI186577. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Bacterial

H1 2026

A portfolio of proprietary GTP and ATP mimetics

SHY Therapeutics retains full ownership of all pharmacophores and molecules developed in its programs.

19S Proteasome &
Pan K-Ras Programs

Our innovative programs target ATPase and GTPase proteins—pioneering new approaches with small molecules (ATP and GTP mimetics) that disrupt conventional pathways and offer unprecedented control over these targets.

Cutting-Edge
Technology

We continuously build our proprietary portfolio through novel computational analyses, machine learning from vast chemical libraries, and SAR-driven synthesis. This evolving platform is a powerhouse for discovering new targets and developing clinical candidates. Our proprietary assays and state-of-the-art techniques allow us to validate and refine our compounds, ensuring they have the greatest therapeutic potential.

Driven by Curiosity & Collaboration

We stay scientifically open-minded, technologically up-to-date, pragmatically focused, data-driven and patient focused. We value the insights and scrutiny from our scientific community to keep pushing the boundaries of what’s possible.

Demonstrating
Impact

Our ATP and GTP mimetics have already shown specific effects on disease targets like the mammalian proteasome ubiquitin system and KRas and additional members of the Ras superfamily of small GTPases. Our ATP and GTP mimetics have also demonstrated promise in models of also fibrosis, and infectious diseases.

  • SHY has been awarded a Small Business Innovation Research grant from the National Institute of Allergy and Infectious Diseases of The National Institutes of Health. The funding will support SHY’s development of an early-stage anti-fungal program, which has already show promising in vitro activity against fungal strains of public health concern. 

     

    The research is supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under Award Number R43AI186577. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

H1 2026

Target

In Vitro

In Vivo

Lead Designation

IND Filing

Bacterial

H1 2026

Target

In Vitro

In Vivo

Lead Designation

IND Filing

Fungi

We are excited by the number of opportunities we believe SHY has to develop first in class therapeutics to benefit patients with significant unmet medical needs. Borrowing an expression used by competitive rowers, we believe we have a lot of open water ahead.

Yaron Hadari & Michael Schmertzler

Founder and Chief Executive Officer & Founder and Executive Chair

sean-robertson-4SXtQktBKOk-unsplash.jpg

Don't be shy!

Partner with us

We welcome collaboration with researchers, advisors, and industry leaders.

Developing small molecule therapies targeting ATPases & GTPases

ATPases and GTPases are essential proteins involved in key cellular functions and disease. SHY’s compound portfolio targets these proteins with high target specificity and wide therapeutic windows, showing promise in models of unmet medical need.

Developing Small Molecules that Inhibit K-Ras/GTP Binding Based on New Affinity Measurements

Featured Publication

Untitled design_edited.jpg
Developing Small Molecules that Inhibit K-Ras_GTP Binding Based on New Affinity Measuremen

Publications

Publication

Heading 4

Jan 6, 2025

Learn more about SHY

  • Non-Covalent Pan-KRAS Inhibitors

     

    Mutations in KRas, a member of the Ras superfamily of small GTPases, are associated with approximately 30% of human tumors, making KRas mutants long-time targets for cancer treatment. Most drugs bind non-covalently to their targets. Articles published in the 1980s and 1990s, however reported affinities for GTP and KRas in the range of 10 pM and led scientists to conclude that the KRas GTP binding site could not be targeted by non-covalent inhibitors – the chemical nature of most drug/target interactions. It was deemed undruggable. 

     

    Accordingly, KRas drug development efforts by pharmaceutical and biotechnology companies focused on alternative mechanisms for inhibiting mutant KRas signaling. Over time, opinions have shifted as to whether these compounds should specifically target just the different KRas mutants or not. Several covalently bound compounds targeting KRas G12C mutations (representing a meaningful but still relatively small number of patients) had impressive initial clinical effects, were approved by the FDA for sale, but, disappointingly, have proven in post-approval clinical use to have limited durations of effect. 

    Reasoning analogously based on his experience developing small molecule inhibitors of ATP binding to receptor tyrosine kinases, Dr. Hadari founded SHY with the expectations that:

    • previously reported affinity measurements for GTP/KRas binding were erroneous; 

    • small molecules could therefore be synthesized that compete non-covalently for GTP binding to KRas; 

    • such small molecules would be more therapeutically effective if they were not specific to just certain KRas mutants, but rather preferentially to KRas and also to other members of the protein superfamily of which KRas is one member; and, finally, 

    • notwithstanding the importance of KRas signaling to sustaining cellular functions, such non-covalently bound pan KRas inhibitors would not necessarily be toxic at therapeutically relevant doses.

    SHY has validated all these hypotheses and succeeded where others have not tried by utilizing state-of-the-art methods and combining proprietary high throughput screens with computationally tailored libraries and some inspired SAR.  

    • SHY conducted a series of measurements of GTP/KRas binding using more contemporary methods than the articles published in the 1980s. In 2020 we posted affinity measurements (Kd’s) for GTP/KRas and KRas mutant binding in the range of 250 nM on bioRxiv, Cold Spring Harbor Laboratory’s preprint server for biology. This Kd is approximately 10,000 times lower than the estimates published in the 1980s and 1990s. 

    • SHY has synthesized over 1,000 small molecules that compete non-covalently and differentiably for GTP for binding to KRas and other members of the KRas superfamily, and differentiably modulate KRas signaling. SHY employs a proprietary fluorescence-based cell-free assay to confirm competitive GTP binding. The inhibition of KRas signaling is further assessed in tumor-derived cell lines expressing both wild-type and mutant KRas.
       

    • Select molecules from this portfolio are undergoing further in vitro, pharmacokinetic, and in vivo xenograft studies to evaluate their potential as clinical candidates. To date, as anticipated, in animal studies they have shown anti-tumor efficacy but not systemic toxicity.

    A newly published study in Molecular Cancer Therapeutics, with research led by SHY scientists, demonstrates SHY’s differentiated approach to RAS inhibition, providing the first evidence that the GTP-binding site of RAS proteins can be competitively targeted by small molecules in a non-covalent manner—a mechanism not employed by current clinical agents.

    Additional supplementary data: Identification of Small-Molecule Inhibitors that Block the GTP-Binding Pocket of K-Ras and Other Members of the Ras Superfamily of Small GTPases

pan-KRas

H1 2026

  • SHY-ONC6: a 19S proteasome ATPase regulatory particle cancer therapeutic candidate

     

    SHY has selected SHY-ONC6 as its first clinical candidate for testing as a cancer therapeutic and is now doing the pre-clinical work to file an IND for it in late 2025. 

     

    Mechanism of Action

     

    SHY-ONC6 is a selective inhibitor of a subset of AAA+ ATPases in the 19S regulatory proteasome particle. The 19S regulatory particle is responsible for recognizing, binding, deubiquitinating, and unfolding proteins before they are degraded in the 20S proteasome core particle. By interfering with this process, SHY-ONC6 induces apoptosis.

    Three FDA approved drugs (Bortezomib, Carfilzomib, and Ixazomib) have validated the clinical and commercial importance of the proteasome ubiquitin system as a therapeutic target but they operate through a mechanism distinct from SHY-ONC6. They are D-peptide like molecules that inhibit the proteolytic beta sites of the 20S proteasome core particle, whereas SHY-ONC6 is a small molecule inhibitor of ATPases in the 19S regulatory particle.

     

    Development Status

     

    SHY-ONC6 has been selected as a clinical candidate. It shows strong in vitro activity and in vivo efficacy across a variety of difficult-to-treat human tumors. Pre-clinical development is ongoing, with an Investigational New Drug (IND) application planned for the second half of 2025. SHY intends to initiate a Phase-I trial focusing on tumors that have shown positive pre-clinical results, with plans to refine target indications in subsequent trials.

     

    In Vitro Activity

     

    SHY-ONC6 demonstrates significant in vitro activity, with IC50 values of 100 nM or less in approximately 80% of the 869 human solid and blood-borne tumor cell lines tested in PRISM studies by the Broad Institute.

     

    In Vivo Efficacy

     

    In mouse xenograft models, SHY-ONC6 shows robust oral efficacy, achieving over 100% tumor growth inhibition (regression) in a range of human-derived tumor models. These include colon, gastric, lung (mesothelioma, small cell, and non-small cell), myeloma, and pancreatic cancers, all known for poor prognoses.

     

    SHY-ONC6 also has favorable pharmacokinetic characteristics and crosses the blood brain barrier.

     

    Safety and Tolerability

     

    Extensive in vivo studies in mice have shown that SHY-ONC6 is well-tolerated at effective doses, with no observed weight loss, behavioral changes, or adverse blood chemistry changes. 

     

    Comprehensive preclinical safety studies will, of course, be conducted prior to SHY’s IND filing.

ATPases

H1 2026

Target

In Vitro

In Vivo

Lead Designation

IND Filing

Oncology

bottom of page