Study Establishes Molecular Link Between Tumor Metabolism and Drug Engagement in Cancer Cells

On December 9, 2025 Promega reported a new study published in Nature Communications reveals technological advances that accelerate breakthroughs in precision medicine. Conducted as a collaboration between Promega, the Center for Advanced Study of Drug Action at the State University of New York at Stony Brook, and the Centre for Medicines Discovery at the University of Oxford, the work leverages bioluminescent NanoBRET Target Engagement (TE) technology developed by Promega to characterize inhibitors that selectively target cancer cells without harming noncancerous cells. Their results demonstrate a connection between drug efficacy and tumor metabolic state, offering a mechanistic bridge between cancer metabolism and precision oncology.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"To our knowledge, this is the first time anyone has characterized this type of uncompetitive inhibitor mechanism directly in live cells." -Ani Michaud, Sr Research Scientist at Promega

Share
"The methods in this study enable us to characterize inhibitors that bind much more tightly in tumor cells with specific mutations," says Ani Michaud, Sr Research Scientist at Promega and co-first author of the Nature Communications paper. "To our knowledge, this is the first time anyone has characterized this type of uncompetitive inhibitor mechanism directly in live cells."

PRMT5: Top Target for Drug Discovery

The published study focuses on a gene-regulating protein called PRMT5, which has long been considered a top target for drug discovery. In normal cells, PRMT5 interacts with a molecule called SAM. However, in the tumor cells of approximately 10-15% of cancers, a deletion of the MTAP gene leads to PRMT5 interacting with the molecule MTA instead, reducing its function. This difference creates a key vulnerability for targeting cancer cells with a mutation to MTAP while leaving normal cells unaffected.

The University of Oxford team designed and developed CBH-002, a cell-permeable BRET probe that binds to a genetically encoded PRMT5-NanoLuc biosensor to report drug target engagement in live cells.

Dr Elisabeth Mira Rothweiler, Postdoctoral Researcher, Centre for Medicines Discovery, University of Oxford and co-first author, says: "CBH-002 could measure various PRMT5 inhibitor types in live cells, prompting us to test its sensitivity to the cofactor SAM. When we discovered the probe’s ability to sense metabolite levels, it established its utility as a metabolic biosensor. Through collaboration with Promega, we demonstrated how MTA influences drug selectivity, revealing why certain inhibitors are so effective in MTAP-deleted cancers."

Dr Rothweiler’s research further enables a strategy for developing molecules that exploit the metabolic vulnerabilities specific to MTAP-deleted cancers, potentially offering highly targeted treatments with minimal effects on healthy tissue.

Uncompetitive Binding in Live Cells

While past studies have characterized this mechanism-of-action (MoA) in biochemical assays, this is the first to use NanoBRET TE technology to characterize uncompetitive, or cooperative, binding in live cells. Biochemical assays can reveal uncompetitive MoAs, but there is often a discrepancy between biochemical data and functional assays like selective cell killing. The NanoBRET TE assay used in this study bridges the two modalities, showing binding MoA in a cellular context that aligns with functional assay results.

Professor Kilian Huber, Associate Professor, Centre for Medicines Discovery, University of Oxford and co-senior author of the study, says, "The biosensor lets us examine, in living cells, how different PRMT5 inhibitors behave under the specific metabolic conditions that make some tumors uniquely vulnerable. This provides unprecedented insight into why certain inhibitors are much more effective in cancers lacking MTAP and paves the way for highly targeted cancer treatment in the future. It’s like turning on the lights inside the cell so we can finally see which key actually fits the lock."

"Selectivity is one of the most critical challenges in cancer therapy, as most treatments also damage healthy cells, leading to dose-limiting toxicities and reduced therapeutic effectiveness," says Peter Tonge, distinguished professor of chemistry and director of the Center for Advanced Study of Drug Action at the State University of New York at Stony Brook and visiting professor at the University of Rochester. "A new class of tumor-specific drugs addresses this by acting uncompetitively with a metabolite that accumulates only in cancer cells, limiting activity to tumor tissue. We have now developed the first technology to quantify the activity of these drugs directly in live cells, providing a foundation for optimizing and advancing next-generation precision oncology therapeutics."

Collaboration Between Academia and Industry

This study was the result of collaboration between Promega, the Center for Advanced Study of Drug Action at the State University of New York at Stony Brook, and the Centre for Medicines Discovery at the University of Oxford, with additional contributions from researchers at Boston University and the Structural Genomics Consortium at the University of Toronto.

"This work underscores the value of research collaborations between academia and industry," says Matt Robers, Associate Director of R&D at Promega and co-senior author of the study. "By combining our complementary expertise in chemical biology and assay design, we were able to dissect how cooperativity can drive cancer cell selectivity. These findings have real potential to guide the development of future precision medicines."

Read the paper "A BRET biosensor for measuring uncompetitive engagement of PRMT5 complexes in cells" in Nature Communications here: View Source

(Press release, Promega, DEC 9, 2025, View Source [SID1234661337])

Natera Highlights New Findings in Lymphoma at the ASH Annual Meeting

On December 9, 2025 Natera, Inc. (NASDAQ: NTRA), a global leader in cell-free DNA and precision medicine, reported the results from two oral presentations that were presented at the American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

Real-world Signatera Analysis: Oral Presentation on December 6

A real-world analysis of personalized circulating tumor DNA (ctDNA) detection in lymphoma evaluated 144 patients across 14 lymphoma subtypes, including aggressive and indolent lymphomas and patients undergoing chimeric antigen receptor T-cell (CAR-T) therapy. Signatera was used clinically to assess baseline ctDNA detection, track molecular clearance during first-line (1L) therapy, and evaluate end-of-treatment (EOT) ctDNA status and post-CAR-T response. Key findings included:

Across 14 subtypes of lymphoma, 94% of patients had detectable ctDNA in a pre-treatment sample.
ctDNA clearance during treatment was highly predictive of CAR-T response (p = 0.0028).
Rapid ctDNA clearance after one cycle of chemotherapy was more predictive of positive outcomes vs. delayed clearance after two cycles of therapy (HR: 20.95 vs. 7.45).
Signatera status at the end of 1L therapy was highly prognostic of event-free survival (HR: 49.77, p<0.0001), outperforming standard of care PET-CT response assessment across lymphoma subtypes.
HOVON Study: Oral Presentation on December 7

The HOVON study was conducted by Foresight Diagnostics, a subsidiary of Natera, in collaboration with Amsterdam University Medical Centers, the Hemato-Oncology Foundation for Adults in the Netherlands (HOVON) and the Netherlands Comprehensive Cancer Organization (IKNL). The study evaluated longitudinal molecular residual disease (MRD) surveillance in 166 patients with diffuse large B-cell lymphoma. The study provided one of the most detailed evaluations to date of ctDNA-MRD dynamics over a two-year surveillance period using the CLARITY ctDNA assay. Key findings included:

The CLARITY ctDNA assay showed early molecular response was associated with improved clinical outcomes, demonstrating its utility as an early risk-stratification marker.
Following any negative ctDNA test during surveillance, the probability of remaining relapse-free was 99% at 6 months and 97% at 12 months.
Early on-treatment molecular response could serve as a dynamic marker for therapy de-escalation or escalation trials, enabling adaptive trial designs.
"These presentations highlight the value of ctDNA in assessing treatment response and long-term risk across lymphoma subtypes, including diffuse large B-cell lymphoma," said Minetta Liu, M.D., chief medical officer of oncology and early cancer detection at Natera. "The findings reinforce how the detection of disease at the molecular level can support more personalized treatment and surveillance strategies for patients with cancer."

(Press release, Natera, DEC 9, 2025, View Source [SID1234661336])

Tempus Announces Ten Abstracts Accepted for Presentation at the 2025 San Antonio Breast Cancer Symposium

On December 9, 2025 Tempus AI, Inc. (NASDAQ: TEM), a technology company leading the adoption of AI to advance precision medicine, reported that ten abstracts have been accepted for presentation at the 2025 San Antonio Breast Cancer Symposium (SABCS). The meeting takes place December 9–12 at the Henry B. González Convention Center in San Antonio, Texas.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"This year at SABCS, our research highlights the power of multimodal data to help unravel the complexity of breast cancer," said Ezra Cohen, MD, Chief Medical Officer of Oncology at Tempus. "By integrating genomic, transcriptomic, and real-world clinical data, we are moving beyond broad disease classifications to uncover specific molecular drivers of progression and resistance. These insights are essential for pinpointing patient populations who may benefit from novel therapies and for optimizing treatment strategies to ultimately improve outcomes for those with advanced disease."

Tempus will highlight its latest scientific and clinical research findings via ten poster presentations:

Integrative Modeling of Multimodal Real-World Data for Improved Risk Stratification of First-Line CDK4/6 Inhibitor Outcomes in Patients with Estrogen Receptor (ER)-Positive/Human Epidermal Growth Factor Receptor 2 (HER2)-Negative Metastatic Breast Cancer
Date/Time: Wednesday, December 10, 2025, 12:30 p.m. – 2:30 p.m. CDT

Presentation Number: PS1-11-08

Summary: We developed a machine learning model that used clinical, genomic, and transcriptomic features to stratify patients with ER-positive/HER2-negative metastatic breast cancer, based on response to first-line CDK4/6 inhibitor plus endocrine treatment and identify predictors of response. This study demonstrated that multimodal real-world data collected during routine care can provide valuable insights into the biology of response to CDK4/6 inhibitors in patients with metastatic breast cancer and help improve patient stratification.
Distinct Transcriptional and Immunosuppressive Microenvironment Signatures in PIK3CA- and ESR1-Mutant Hormone Receptor Positive (HR+)/HER2- Metastatic Breast Cancer (MBC)
Date/Time: Wednesday, December 10, 2025, 12:30 p.m. – 2:30 p.m. CDT

Presentation Number: PS1-11-22

Summary: This study compared transcriptomic and immune profiles in HR+/HER2- metastatic breast cancer across wild-type, PIK3CA-mutant, ESR1-mutant, and co-mutant groups. SFRP2 downregulation was specific to ESR1-mutant tumors, while SCGB2A2 was robustly upregulated in PIK3CA-mutant and co-mutant tumors, suggesting its potential as a diagnostic and therapeutic target. Immune analysis revealed increased M2 macrophages and regulatory T cells in PIK3CA-mutant and co-mutant tumors, with the most pronounced immunosuppressive microenvironment in PIK3CA-mutant cases.

Clinical and Molecular Landscape of ESR1 and PIK3CA Co-Mutated Hormone Receptor–Positive Metastatic Breast Cancer (HR+ MBC): Insights from 8,626 Patients Including Polyclonality and TP53 Alterations
Date/Time: Wednesday, December 10, 2025, 12:30 p.m. – 2:30 p.m. CDT

Presentation Number: PS1-11-16

Summary: Distinct mutation groups—no-MUT, PIK3CA-MUT, ESR1-MUT, and co-MUT (PIK3CA + ESR1)—exhibited unique clinical and genomic features; co-MUT was linked to more bone metastases, elevated tumor mutational burden, increased polyclonality, and shorter real-world overall survival. Notably, the frequency of ESR1 and co-mutations was higher in patients who underwent late molecular testing, indicating these alterations often emerge after extended endocrine therapy and highlighting the importance of timing in molecular assessment for guiding treatment strategies.
Clinicogenomic and Immune Profiles of Male Breast Cancer by Race: Insights from a Large Real-World Cohort
Date/Time: Wednesday, December 10, 2025, 5:00 p.m. – 6:30 p.m. CDT

Presentation Number: PS2-06-28

Summary: Real-world analysis of male breast cancer revealed broadly similar clinicogenomic profiles between Black and White patients, with HR+/HER2-negative and luminal subtypes predominating. Notable immune microenvironment differences—lower M2 macrophage infiltration and higher PD-L1 negativity in Black patients—may impact immunotherapy response and warrant further research into racial variation.
Cathepsin Protease Expression and Therapeutic Outcomes to Trastuzumab Deruxtecan (T-DXd) in Metastatic Breast Cancer
Date/Time: Wednesday, December 10, 2025, 5:00 p.m. – 6:30 p.m. CDT

Presentation Number: PS2-08-18

Summary: This study characterized pre-treatment expression of cathepsin proteases prior to T-DXd (cleavable linker) vs. T-DM1 (non-cleavable linker) treatment to understand their impact on clinical outcomes in metastatic breast cancer using the Tempus real-world database. This study demonstrated that among T-DXd treated patients, high cathepsin B and L protease expression was associated with improved outcomes in patients with HR+/HER2- disease, whereas in HER2+ patients, high expression of these proteases was associated with worse survival. In contrast, in the T-DM1 cohort there were no significant associations between protease expression and OS in the overall cohort as well as in the HER2+ cohort. This highlights the potential role of proteases as biomarkers of response to T-DXd in HER2 low/ultralow, but not HER2+ metastatic breast cancer.
Treatment Sequencing in HR+ HER2- Metastatic Breast Cancer and Associated Real World Outcomes
Date/Time: Wednesday, December 10, 2025, 5:00 p.m. – 6:30 p.m. CDT

Presentation Number: PS2-04-11

Summary: This study investigates the impact of first-line and second-line therapy selections on real-world progression-free survival (rwPFS) and real-world overall survival (rwOS) in patients with HR+ HER2- mBC. CDK4/6i significantly outperforms taxane in first-line. Patients who received second-line CDK4/6i following first-line taxane had longer second-line rwPFS than those who received CDK4/6i first-line treatment. The findings suggest that taxane may cause sensitization to CDK4/6i, potentially conferring better survival outcomes in a subset of patients where first-line taxane is advised, or a potential advantage to chemo before CDK4/6i re-challenge.
Real-World Second-Line Treatment Use and Clinical Outcomes in Patients With HR-Positive/HER2-Negative Metastatic Breast Cancer and an ESR1 Mutation
Date/Time: Wednesday, December 10, 2025, 5:00 p.m. – 6:30 p.m. CDT

Presentation Number: PS2-06-18

Summary: The study objective was to characterize real-world second-line treatment use and clinical outcomes (rwPFS and rwOS) in a metastatic breast cancer patient population with positive ESR1m test after first-line treatment with AI+CDK4/6i. Findings show that this cohort describes a complex subgroup of patients to treat, with rwPFS outcomes highlighting the need for more effective strategies that address resistance to AI+CDK4/6i and improve patient outcomes. Results from Flatiron Health EHR and Tempus both indicated consistent findings.
Comparative Analysis of the Tumor Immune Microenvironment (TIME) and Primary and Metastatic Tissue in HR+/HER2- and Triple-Negative Breast Cancer (TNBC)
Date/Time: Thursday, December 11, 2025, 12:30 p.m. – 2:00 p.m. CDT

Presentation Number: PS3-12-27

Summary: Tempus Lens was leveraged to assess the Tumor Immune Microenvironment (TIME) across metastatic sites of disease in HR+/HER2- breast cancer compared to TNBC, finding significant differences by site in both subtypes, with HR+/HER2- cancer being less immunogenic overall. Lung, pleura, and peritoneum metastases, along with breast and lymph nodes, showed the highest CD8+ T cell proportions, suggesting a subset of HR+/HER2- patients with these sites may potentially respond to Immune Checkpoint Inhibitor (ICI) therapy. This high variability of TIME profiles across metastatic sites warrants further study in prospective trials to guide patient selection for ICI.
The Molecular and Immune Landscape of HER2 Positive Breast Cancer
Date/Time: Thursday, December 11, 2025, 5:00 p.m. – 6:30 p.m. CDT

Presentation Number: PS4-05-21

Summary: Tempus Lens was employed to evaluate the tumor immune microenvironment of HER2+ breast cancer, assessing immune cell infiltration, TMB, PD-L1 status, and HLA allele prevalence, to uncover biomarkers for treatment guidance. A notable percentage of patients with localized and de novo metastatic disease displayed TMB high status and/or PD-L1 positivity. Additionally, TMB-high and PD-L1 positive patients with de novo metastatic disease treated with first-line chemotherapy or anti-HER2 therapy had significantly worse real-world overall survival (rwOS), suggesting a potential therapeutic benefit of incorporating immunotherapy into the treatment paradigm, in both localized and metastatic disease settings. Furthermore, observed ethnic HLA polymorphisms in the cohort may contribute to differences in outcomes and could potentially guide the development of population-specific immunotherapeutic strategies.

Real-World Data (RWD) Outcome Analysis of ESR1 Mutation Emergence in HR+/HER2- Metastatic Breast Cancer Through the Continuum of Standard of Care Hormonal Therapy
Date/Time: Friday, December 12, 2025, 12:30 p.m. – 2:00 p.m. CDT
Presentation Number: PS5-05-02
Summary: This large multimodal RWD outcome analysis from longitudinal molecular surveillance testing (xF) in HR+/HER2- mBC pts treated with AI+CDK4/6i sheds light on the continuum of ESR1m emergence and patient outcomes from first-line and beyond outside of clinical trial data. Analyses show higher ESR1m incidence is associated with reduced survival regardless of line of therapy.

(Press release, Tempus, DEC 9, 2025, View Source [SID1234661335])

Artera Debuts ArteraAI Prostate Test (Post‑RP), Expanding its Oncology Portfolio

On December 9, 2025 Artera, a developer of multimodal AI-based prognostic and predictive cancer tests, reported the commercial launch of the ArteraAI Prostate Test (Post‑RP) for patients experiencing biochemical recurrence (BCR) following radical prostatectomy.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The test builds on the established ArteraAI platform and delivers prognostic results and insights into short-term ADT benefit to optimize decision-making in the salvage therapy setting, where additional treatment is given if cancer returns after surgery.

After radical prostatectomy, PSA should be undetectable. A rising PSA after surgery can indicate biochemical recurrence, suggesting residual cancer cells may remain. With an estimated 20% to 40% of patients affected, the test now delivers personalized, AI-driven data to this previously underserved population, expanding access to advanced precision medicine.

"The launch of the ArteraAI Prostate Test (Post-RP) is a significant milestone in Artera’s continued growth," said Andre Esteva, CEO of Artera. "This expansion broadens our portfolio, extends our reach to an underserved patient population, and strengthens our partnerships across the healthcare ecosystem. As demand for responsible AI in oncology accelerates, we remain focused on scaling access, delivering measurable value to clinicians, and ensuring our technology reaches the patients who need it most."

This test is based on the robust data presented at the 2024 AUA Annual Meeting, which showed that ArteraAI’s MMAI-based prostate biomarker can accurately estimate the risk of metastasis and help predict which men may benefit from intensified salvage therapy, including hormone therapy plus radiation.

"There is a continued need to optimize treatment for men with post-prostatectomy BCR, especially in understanding which patients benefit from ADT when undergoing salvage radiation," said Todd Morgan, urological surgeon and Chief of Urologic Oncology at Michigan Medicine. "This test has substantial promise for helping individualize these treatment decisions."

As with all Artera products, the ArteraAI Prostate Test (Post‑RP) integrates seamlessly into standard pathology workflows, enabling rapid turnaround and broad accessibility. The test relies on the digitized image of the surgical specimen, preserving the resected tissue while delivering actionable insights to guide personalized treatment decisions.

The launch of the ArteraAI Prostate Test (Post-RP) reinforces Artera’s mission to advance the frontier of precision oncology, offering a timely, cost-effective tool that complements clinical assessments for patients with recurrent prostate cancer.

The ArteraAI Prostate Test (Post‑RP) is now available for commercial ordering nationwide. For more information, visit artera.ai.

(Press release, Artera, DEC 9, 2025, View Source [SID1234661334])

CytoAgents to Present New CTO1681 Data at the ESMO Immuno-Oncology Congress 2025

On December 9, 2025 CytoAgents, Inc., a clinical-stage biotechnology company developing CTO1681, a novel, steroid-sparing inhibitor of prostaglandin-mediated inflammation reported that data from two of its Investigational New Drug (IND) application enabling studies will be presented at the European Society for Medical Oncolgy (ESMO) (Free ESMO Whitepaper) Immuno-Oncology Congress 2025, taking place December 10-12, 2025 in London, United Kingdom.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

CTO1681 is an orally administered immune modulator that targets inflammation in the tumor microenvironment and mitigates cell therapy induced toxicities frequently seen in lymphoma patients receiving CAR T-Cell Therapy. CytoAgents is currently enrolling patients in a Phase 1b/2a clinical trial evaluating CTO1681 in lymphoma patients receiving CAR T-Cell Therapy at risk for inflammatory driven toxicities.

At ESMO (Free ESMO Whitepaper), the company will present data from two non-clinical studies demonstrating that CTO1681 attenuates key cytokines known to drive Cytokine Release Syndrome (CRS) without compromising the CAR T-Cell mediated tumor killing in vitro and in vivo. These studies demonstrate that CTO1681 does not interfere with CD19 CAR T-Cell expansion and anti-tumor activity in vivo while reducing the inflammatory cytokines associated with CRS and ICANS.

"These data suggest CTO1681 could enable safer CAR T-Cell Therapy administration, support outpatient treatment paradigms and broaden patient access without compromising anti-tumor efficacy," said Teresa Whalen, Chief Executive Officer at CytoAgents. "We look forward to sharing these data at ESMO (Free ESMO Whitepaper) and to continuing our advancement of CTO1681."

Poster Presentation Details:

Title: CTO1681 Does Not Interfere with CAR T-Cell Anti-Tumor Efficacy In Vivo

Presenter: Michael Howell, PhD, Founder and President Mountaineer Biosciences, San Diego, CA, USA

Final Poster Number (FPN): 109P

Session Date and Time: Wednesday, December 10, 2025, 5:15 PM – 6:30 PM GMT

Location: Queen Elizabeth II Centre – Churchill Room

Title: CTO1681 Attenuates CRS-Driving Cytokines Without Compromising CAR T-Cell-Mediated Tumor Killing In Vitro

Presenter: Michael Howell, PhD, Founder and President Mountaineer Biosciences, San Diego, CA, USA

Final Poster Number (FPN): 113P

Session Date and Time: Wednesday, December 10, 2025, 5:15 PM – 6:30 PM GMT

Location: Queen Elizabeth II Centre – Churchill Room

(Press release, CytoAgents, DEC 9, 2025, View Source [SID1234661333])