Sapience Therapeutics Presents Three Posters at the American Association for Cancer Research (AACR) Annual Meeting 2024

On April 8, 2024 Sapience Therapeutics, Inc., a clinical-stage biotechnology company focused on the discovery and development of peptide therapeutics to address oncogenic and immune dysregulation that drive cancer, reported the presentation of new data on its clinical and pipeline programs during three poster presentations at the 2024 American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting, taking place April 5-10, 2024, in San Diego, CA (Press release, Sapience Therapeutics, APR 8, 2024, View Source [SID1234641904]).

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!

"At AACR (Free AACR Whitepaper) 2024, we are thrilled to present foundational data that support our growing clinical and preclinical pipeline of peptide therapeutics against difficult-to-drug targets, such as β-catenin, C/EBPβ, and c-Jun (AP-1 complex)," said Jim Rotolo, Ph.D., SVP, Research and Translational Sciences. "These data demonstrate the ability of our lead SPEARs, ST316 and ST101, to positively impact the tumor immune microenvironment, providing further evidence of their therapeutic promise. We look forward to ST316 and ST101 continuing to advance through clinical development and moving our AP-1 program into IND-enabling studies."

Sapience scientists will present non-clinical immune-oncology results from both of its clinical programs, ST316, a first-in-class antagonist of β-catenin, and ST101, a first-in-class antagonist of C/EBPβ. Sapience will also present its first disclosure of pre-clinical data describing JunAP, its first-in-class AP-1 complex antagonist targeting the interaction of c-Jun with Fra1.

Poster presentation details and abstract highlights include:

Title: "ST316, a clinical peptide antagonist of beta-catenin, induces anti-tumor immune responses by multiple mechanisms of action"
Session Title: Inflammation in Tumor Initiation and Progression
Location: Poster Section 4
Abstract Number: 5332
Date and Time: Tuesday, April 9, 2024, 1:30 PM to 5:00 PM

ST316 is a first-in-class peptide antagonist of the interaction between β-catenin and its co-activator, BCL9, a complex that drives oncogenic gene expression in multiple cancers where aberrant Wnt/β-catenin pathway signaling is observed, and impacts the tumor immune microenvironment by promoting immunosuppressive myeloid-derived suppressor cell (MDSC) and tumor associated macrophage (TAM) populations. ST316 is currently being evaluated in a Phase 1-2 study (NCT05848739) enrolling patients with selected advanced solid tumors likely to harbor abnormalities of the Wnt/β-catenin signaling pathway.

In nonclinical studies, Sapience evaluated the ability of ST316 to enhance anti-tumor immune responses in combination with anti-PD-1 and anti-TIGIT therapies.
ST316 increases the ratio of proinflammatory M1 macrophages to immunosuppressive M2 macrophages, decreases PD-L1 expression on M2 macrophages and decreases PD1 expression on CD8+ T cells.
ST316 promotes cell surface expression of CD155/PVR to activate T cells in the presence of anti-TIGIT antibody.
In nonclinical in vivo studies, ST316 in combination with anti-PD-1 treatment displays significant anti-tumor activity with accompanied reduction in M2 macrophages.
These data support a paradigm in which ST316 promotes a shift to an immune-active tumor microenvironment via multiple mechanisms, including driving macrophage polarization toward an M1 immune-promoting phenotype, augmenting activity of cytotoxic T cells, and increasing expression of the checkpoint activator CD155/PVR on cancer cells.
Title: "ST101, a clinical C/EBPβ-antagonist peptide, promotes an immune-active tumor microenvironment by multiple cellular mechanisms"
Session Title: The Tumor Microenvironment as a Drug Target
Location: Poster Section 13
Abstract Number: 2909
Date and Time: Monday, April 8, 2024, 1:30 PM to 5:00 PM

ST101 is a peptide antagonist of C/EBPβ that is being evaluated in a Phase 2 clinical study in patients with recurrent and newly diagnosed glioblastoma (NCT04478279).

Sapience evaluated the effect of ST101 on immunosuppressive cell populations and on activation of cytotoxic T cells in macrophage co-culture systems.
ST101 inhibits the immunosuppressive M2 macrophage program, resulting in polarization of macrophages to the proinflammatory M1 phenotype and increasing cytotoxic T cell activation.
In animal models, ST101 enhances the activity of anti-PD-1 treatment by increasing the M1/M2 ratio in the TAM population.
Gene expression analysis of clinical biopsies indicates that ST101 modulates the tumor immune microenvironment by suppressing genes required for M2 macrophage polarization, resulting in an enhanced CD8/Treg ratio.
ST101 represents a novel approach to enhance anti-tumor immune activity and these data suggest its utility for combination strategies in cancers with poor response to immune checkpoint inhibition.
Title: "JunAP, a peptide antagonist against the activator protein 1 transcription factor complex, demonstrates cancer cell cytotoxicity, reduced invasion in vitro and tumor regression in vivo in TNBC models"
Session Title: Oncogenic Transcription Factors
Location: Poster Section 18
Abstract Number: 3051
Date and Time: Monday, April 8, 2024, 1:30 PM to 5:00 PM

AP-1 complexes have been identified as novel therapeutic targets in cancer due to their role in tumor growth, invasion, metastasis, angiogenesis and chemoresistance.
AP-1 complexes consisting of c-Jun and Fra1 are implicated in the survival of a diverse set of tumors.
JunAP targets and blocks c-Jun and Fra1 dimerization, inhibits AP-1 transcriptional activity and inhibits AP-1-dependent cell survival and invasion in vitro.
JunAP demonstrates potent anti-tumor activity in vivo in mouse triple negative breast cancer and melanoma tumor models.
More information can be found on the 2024 AACR (Free AACR Whitepaper) website.

About ST316

ST316 is a first-in-class peptide antagonist of the interaction between β-catenin and its co-activator, BCL9, a complex that drives oncogene expression in multiple cancers where aberrant Wnt/β-catenin pathway signaling is observed. ST316-101 (NCT05848739) is a first-in-human, open-label, Phase 1-2 dose-escalation and expansion study designed to determine the safety, tolerability, PK, PD and early efficacy of ST316. The Phase 1 dose-escalation portion of the Phase 1-2 study is enrolling and dosing patients with select advanced solid tumors that are known to harbor abnormalities of the Wnt/β-catenin signaling pathway. The Company expects to complete the Phase 1 portion of the study in the first half of 2024. Following completion of the study’s Phase 1 portion, the recommended dose will advance to the Phase 2 dose expansion portion of the study in colorectal cancer patients.

About ST101

ST101, a first-in-class antagonist of C/EBPβ, is currently being evaluated in the Phase 2 portion of an ongoing Phase 1-2 clinical study in patients with recurrent GBM (rGBM) (NCT04478279). In an ongoing window-of-opportunity sub-study, ST101 is being evaluated as a monotherapy in rGBM and in combination with radiation and temozolomide in newly diagnosed GBM, with patients receiving ST101 before and after surgical resection. ST101 has been granted Fast Track designation for rGBM from the U.S. FDA and orphan designations for glioma from the U.S. FDA and the European Commission.

Hinge Bio Presents Preclinical Data from its GEM-DIMER Program Targeting B Cell Depletion at the American Association for Cancer Research (AACR) Annual Meeting 2024

On April 8, 2024 Hinge Bio, Inc., a privately-held biotechnology company, reported that Daniel Capon, Ph.D., Chief Scientific Officer, and Juha Punnonen, M.D., Ph.D., Chief Development Officer, are presenting data from the company’s GEM-DIMER program targeting B cell depletion at this year’s American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting 2024 in San Diego (Press release, Hinge Bio, APR 8, 2024, View Source [SID1234641903]).

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!

"Hinge Bio is addressing areas of significant unmet medical need with its revolutionary GEM-DIMER platform, which uses topological engineering to combine the most desirable properties of multiple clinically relevant antibodies into a single molecule with dramatically improved activity, while retaining the robust stability and manufacturability of conventional antibodies," said Dr. Capon. Dr. Punnonen added, "This encouraging preclinical data supports advancing its GEM-DIMER candidate targeting CD19, CD20, and Fc gamma receptors as quickly as possible to clinical investigation in order to treat the large number of patients with B cell mediated diseases who remain resistant to currently approved treatments."

Oral Poster details are as follows:

Beyond antibodies and CAR-T: Topologically-engineered, super-dimeric antibody-like molecules with dual Fc domains for trispecific, bivalent targeting of CD19, CD20, and Fc gamma receptors

Abstract #2730, Session PO.IM01.06 – Single Target and Bispecific Antibodies

Session Date/Time: April 8, 2024, 1:30 PM – 5:00 PM, Section 6

Presenting Authors: Daniel Capon, PhD, Chief Scientific Officer;
Juha Punnonen, MD, PhD, Chief Development Officer

Summary: Hinge Bio’s CD19/CD20-targeting GEM-DIMER molecules are promising candidates to provide efficient depletion of both CD19+ and CD20+ cells, providing potential for broad and deep depletion of B cells with reduced risk of emergence of antigen escape variants. These data support the advancement of these CD19/CD20-targeting GEM-DIMER molecules in multiple indications where depletion of CD19+ and/or CD20+ B cells is needed. Preparations for clinical investigation are ongoing.

Adela Presents Data Demonstrating Ability of Tissue-Agnostic MRD Assay to Predict Recurrence in Head & Neck Cancer at the American Association for Cancer Research Annual Meeting 2024

On April 8, 2024 Adela, Inc., an innovator in blood testing for minimal residual disease monitoring and early cancer detection through a proprietary genome-wide methylome enrichment technology, reported initial results demonstrating the ability of its MRD assay to predict recurrence in head & neck cancer, at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting from April 5-10, 2024 (Press release, Adela, APR 8, 2024, View Source;neck-cancer-at-the-american-association-for-cancer-research-annual-meeting-2024-302109070.html [SID1234641902]).

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!

"In patients with head & neck cancer, determining the most appropriate course of action following curative intent treatment can be challenging due to the need to balance the potential side effects of treatment with the ramifications of recurrence. Furthermore, detection of recurrence is complicated by inaccuracies of medical imaging and a lack of standardized surveillance procedures", said Scott Bratman MD, PhD, Chief Innovation Officer, Adela and Radiation Oncologist and Clinician-Scientist at Princess Margaret Cancer Centre, University Health Network. "We are highly encouraged by these initial results and the potential for a novel approach to improve care of patients with head & neck cancer."

The ability of Adela’s assay to quantify cfDNA cancer signal and predict recurrence was evaluated in individuals diagnosed with stage I-IVB human papillomavirus (HPV)-negative and HPV-positive head and neck cancer treated at Princess Margaret Cancer Centre, University Health Network. This training analysis included 249 plasma samples collected from 75 patients (a subset of the full cohort of >1,100 plasma samples from >300 individuals). Blood draws occurred before and after curative intent treatment, and in a subset of patients, at 12 and 24 months post-curative intent treatment. Adela’s MRD assay demonstrated the ability to identify patients more likely to recur than not, based on the blood draw following curative intent treatment (i.e., landmark), and also at the longitudinal timepoints. Significant differences in recurrence-free survival (RFS) were observed, with a hazard ratio (HR) of 10.97 (P<0.001) at the landmark timepoint, and an HR of 22.83 (P<0.001) longitudinally, when patients were stratified by MRD positivity.

"By identifying patients with head & neck cancer who are more likely to recur, our MRD test has the potential to provide clinicians with useful information that may aid in their decisions regarding the use of adjuvant therapy as well as enable recurrences to be detected and treated earlier," said Dr. Anne-Renee Hartman, Chief Medical Officer of Adela. "These initial results give us confidence that our tissue-agnostic approach to MRD detection for both HPV-positive and HPV-negative head & neck cancer can address the strong unmet need in this particular indication."

Validation results from a held-out test set from the cohort will be presented at a future meeting. Adela plans to commercialize its tissue agnostic test for minimal residual disease (MRD) monitoring in 2025.

Data will also be presented at AACR (Free AACR Whitepaper) on the analytical performance of Adela’s genome-wide methylome enrichment platform on April 9, 2024.

Presentation Details

Abstract # 2427: The development of a tissue-agnostic genome-wide methylome enrichment MRD assay for applications across the cancer care continuum for head and neck malignancies
Geoffrey Liu1
Mon Apr 8, 2024 9 am-12:30 pm PT
Section 40 Poster Board Number 23

Abstract # 5024: Analytical performance of a genome-wide methylome enrichment platform to detect minimal residual disease from plasma-derived cell-free DNA
Hestia Mellert2
Tues Apr 9, 2024 9am – 12:30pm PT
Section 40 Poster Board Number 11

Nuvalent Presents New Preclinical Data Supporting Profiles of HER2-Selective Inhibitor, NVL-330, and ROS1-Selective Inhibitor, Zidesamtinib, at AACR Annual Meeting 2024

On April 8, 2024 Nuvalent, Inc. (Nasdaq: NUVL), a clinical-stage biopharmaceutical company focused on creating precisely targeted therapies for clinically proven kinase targets in cancer, reported the presentation of new preclinical data for its novel HER2-selective inhibitor, NVL-330, and novel ROS1-selective inhibitor, zidesamtinib (NVL-520) (Press release, Nuvalent, APR 8, 2024, View Source [SID1234641901]). The two posters will be presented at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting taking place from April 5 – 10 in San Diego. The posters will also be available on the Nuvalent website at www.nuvalent.com following the presentations.

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!

"Today’s presentations continue to reinforce the differentiated profiles of our drug candidates," said Henry Pelish, Ph.D., Senior Vice President of Drug Discovery at Nuvalent. "In comparative in vitro and in vivo analyses of NVL-330 with currently approved and investigational HER2-targeting agents, NVL-330 demonstrated a differentiated preclinical profile by achieving higher CNS penetration and deeper intracranial response. Importantly, in these preclinical studies, NVL-330 also demonstrated potency against a broad range of HER2 oncogenic alterations and selectivity over wild-type EGFR, in line with our goal of designing molecules that can thread the needle between multiple competing challenges."

Dr. Pelish continued, "In our ongoing ARROS-1 clinical trial of zidesamtinib, preliminary Phase 1 data has demonstrated a differentiated profile combining activity against ROS1 resistance mutations, CNS penetrance, and TRK avoidance which we believe has the potential to translate to deep, durable responses for patients with ROS1-driven cancers. A new preclinical mutagenesis screen reinforces this potential, showing that on-target resistance is unlikely following treatment with zidesamtinib at its average observed clinical concentration."

In 2024, the company expects to initiate a Phase 1 trial for its HER2 program and to share updated data from the ARROS-1 trial at a medical meeting.

AACR Presentation Overviews:

Title: Preclinical characterization of NVL-330, a selective and brain penetrant HER2 tyrosine kinase inhibitor with broad activity on HER2 oncogenic alterations
Authors: Yuting Sun*1, Kristin L. Andrews1, Anupong Tangpeerachaikul1, Tuan M. Nguyen1, Baudouin Gerard1, Nancy E. Kohl2, Joshua C. Horan1, Henry E. Pelish1
Abstract Number: 1979
Session Category: Experimental and Molecular Therapeutics
Session Title: Kinase and Phosphatase Inhibitors 2
Session Date and Time: Monday April 8, 2024, from 9:00 – 12:30 p.m. PT
Location: Poster Section 25

Presentation summary:

NVL-330 had broad preclinical activity on HER2 oncogenic alterations, including HER2 exon20ins, HER2 activating point mutations, and amplified wild-type HER2.
In a preclinical comparison with the selective tyrosine kinase inhibitor, zongertinib, NVL-330 demonstrated:
Similar potency and selectivity over wild-type EGFR; and,
Higher CNS penetrance.
In a preclinical comparison with antibody drug conjugate, T-DXd (Enhertu), NVL-330 demonstrated:
Deeper response and higher CNS penetrance in an intracranial tumor model; and,
Activity in cells with acquired resistance to T-DXd.
Title: Mutagenesis screens support potential best-in-class profile for selective, brain-penetrant, and TRK-sparing ROS1 inhibitor zidesamtinib (NVL-520)
Authors: Anupong Tangpeerachaikul*1, Franklin Gu1, Henry E. Pelish1
Abstract Number: LB182
Session Title: Late-Breaking Research: Experimental and Molecular Therapies 2
Session Date and Time: Monday April 8, 2024, from 1:30 – 5:00 p.m. PT
Location: Poster Section 52

Presentation summary:

Comparison of the clinical concentration of zidesamtinib to its efficacious in vitro concentration suggests a potential for a deep and sustained inhibition of ROS1 and ROS1 G2032R fusions in humans, including in the CNS.
Zidesamtinib effectively suppressed on-target resistance in ENU mutagenesis screens with both ROS1 and ROS1 G2032R fusions, predicting that on-target resistance is unlikely when used as either a first-line or a later-line therapy.
On-target resistance is predicted to be more likely for earlier-generation ROS1 inhibitors crizotinib, entrectinib, and potentially repotrectinib as a first-line therapy.
These mutagenesis screens provide additional preclinical support for zidesamtinib’s potential to drive deep and durable responses for patients with ROS1-driven cancers.
*Presenter, corresponding author; 1Nuvalent, Inc., Cambridge, MA, USA; 2Kohl Consulting, Wellesley, MA, USA

About NVL-330
NVL-330 is a novel brain-penetrant HER2-selective tyrosine kinase inhibitor designed to address the combined medical need of treating HER2-mutant tumors, including those with HER2 exon 20 insertion mutations, avoiding treatment related adverse events due to off-target inhibition of wild-type EGFR, and treating brain metastases.

About zidesamtinib (NVL-520)
Zidesamtinib is a novel brain-penetrant ROS1-selective inhibitor created with the aim to overcome limitations observed with currently available ROS1 inhibitors. Zidesamtinib is designed to remain active in tumors that have developed resistance to currently available ROS1 inhibitors, including tumors with treatment-emergent ROS1 mutations such as G2032R. In addition, zidesamtinib is designed for central nervous system (CNS) penetrance to improve treatment options for patients with brain metastases, and to avoid inhibition of the structurally related tropomyosin receptor kinase (TRK) family. Together, these characteristics have the potential to avoid TRK-related CNS adverse events seen with dual TRK/ROS1 inhibitors and to drive deep, durable responses for patients across all lines of therapy. Zidesamtinib has received breakthrough therapy designation for the treatment of patients with ROS1-positive metastatic non-small cell lung cancer (NSCLC) who have been previously treated with 2 or more ROS1 tyrosine kinase inhibitors and orphan drug designation for ROS1-positive NSCLC. Zidesamtinib is currently being investigated in the ARROS-1 trial (NCT05118789), a first-in-human Phase 1/2 clinical trial for patients with advanced ROS1-positive NSCLC and other solid tumors.

GV20 Therapeutics Presents Preclinical Data on GV20-0251 for Solid Tumors Targeting IGSF8 at AACR Annual Meeting 2024

On April 8, 2024 GV20 Therapeutics, a clinical-stage biotechnology company integrating AI, genomics, and disease biology to create next-generation antibody therapeutics for cancer, reported preclinical data on it GV20-0251 program during an oral presentation at the American Association of Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting 2024 in San Diego, CA (Press release, GV20 Therapeutics, APR 8, 2024, View Source [SID1234641900]).

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!

"I am honored to showcase our novel technology platform and present the previously unknown immune function of IGSF8 at AACR (Free AACR Whitepaper) 2024. GV20-0251 is the first ever AI-designed antibody against an AI-predicated target to advance into the clinic and can potentially bring significant benefits to patients with solid tumors," said Shirley Liu, CEO of GV20 Therapeutics. "We have made great progress on GV20-0251 Phase I dose escalation and look forward to advancing into monotherapy cohort expansion and combination studies later this year."

The oral presentation at AACR (Free AACR Whitepaper) included an overview on the company’s novel discovery and development process of its lead antibody, GV20-0251, which antagonizes a novel cancer immunotherapy target, IGSF8. Many cancer patients’ immune systems make anti-PD1/anti-PDL1 antibodies, however the patients do not make enough and have the wrong IgA Fc to completely kill the tumors. GV20 uses tens of thousands of tumors as experimental systems and leverages artificial intelligence (AI) to mine big data from large tumor profiles to predict the antibodies in patient tumors that can be developed into cancer drugs. Through this modeling, GV20 discovered that IGSF8 is a novel innate immune checkpoint highly expressed on malignant cells with antigen presentation defects. Anti-IGSF8 enhances NK cell killing, antigen presentation, and turns immune-cold tumors hot. In addition, anti-IGSF8 alone or in combination with anti-PD1 inhibits tumor growth in multiple models, including some known to resist anti-PD1 therapy.

GV20’s in-house STEAD platform (Simultaneous Target Evaluation and Antibody Discovery), which integrates AI, genomics, and disease biology, can not only identify novel drug targets, but also create next-generation therapies against these targets, like GV20-0251. The ongoing Phase I study in solid tumors (NCT05669430) is advancing and GV20-0251 is demonstrating promising early clinical activities.

Details of the oral presentation at AACR (Free AACR Whitepaper) are below:

Title: "IGSF8 is a novel innate immune checkpoint and cancer immunotherapy target"

Session Type: Minisymposium
Session Category: Immunology
Session Title: Immune Targets and Therapies
Session Date/Time: Monday Apr 8, 2024, 2:30 PM – 4:30 PM
Abstract Presentation Number: 3914