Immunitas Therapeutics Presents Preclinical Data Supporting Combination of IMT-009 with Anti-PD1 Immunotherapy at the AACR 2024 Annual Meeting

On April 8, 2024 Immunitas Therapeutics ("Immunitas"), a clinical stage precision immunotherapy company committed to discovering and developing novel, differentiated therapeutics for patients with cancer, reported preclinical data supporting the combination potential of IMT-009 with anti-PD1 immunotherapy at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting (AACR 2024) in San Diego, California (Press release, Immunitas Therapeutics, APR 8, 2024, View Source [SID1234641891]).

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 need for new immuno-oncology approaches persists, with a significant proportion of patients unable to benefit from standard of care PD-1/PD-L1 checkpoint blockade treatment due to primary and acquired resistance. This need has fueled our work to develop IMT-009 as a differentiated cancer treatment option for patients," said Annalisa D’Andrea, Chief Scientific Officer at Immunitas. "The preclinical data presented at AACR (Free AACR Whitepaper) build upon this innovation, supporting potential for clinical benefit through combination approaches featuring IMT-009 and anti-PD-1 treatments."

Novel, anti-CD161 antibody IMT-009 has been shown to restore the anti-cancer activity of T and NK cells in preclinical studies by blocking interactions between CD161 and its ligand, CLEC2D, and is currently undergoing clinical evaluation for use in solid tumor and hematologic malignancies.

The data presented at AACR (Free AACR Whitepaper) 2024 build upon these preclinical data, reinforcing CD161 as a rational immunotherapy target with the potential to enhance T cell-mediated anti-tumor activity across a range of tumors. Single cell RNA sequencing analysis of published tumor datasets demonstrated the presence of CD161 expressing T cells in patients who have previously progressed on, or are refractory to, anti-PD-(L)1 therapy, indicating potential for benefit from treatment with IMT-009 as monotherapy or in combination with anti-PD-(L)1 therapy. In cancer cell models, combination of IMT-009 and anti-PD1 treatment significantly enhanced T cell mediated tumor killing. Transcriptomic changes upon treatment with IMT-009 also show robust T cell activation and cytotoxicity gene signatures which are further enriched in combination with anti-PD1 treatment. Together these data support clinical assessment of IMT-009 treatment in combination with anti-PD-(L)1 approaches in patients refractory to anti-PD-(L)1 treatment alone.

Presentation Details for AACR (Free AACR Whitepaper) 2024
Title: Abundance of KLRB1+ (CD161) T cells in anti-PD1 non responders coupled with enhanced tumor cytotoxicity of anti-CD161 (IMT-009) with anti-PD1 makes it a rational target for combination with anti-PD-(L)1 immunotherapy
Abstract Number: 1375
Date/Time: Monday, April 8, 2024, 9:00am – 12:30pm PT

About IMT-009
IMT-009 is a fully human, Fc-attenuated IgG1 monoclonal antibody that binds to CD161 and blocks its interaction with its ligand, CLEC2D. Preclinical data confirm that CD161 blockade with IMT-009 results in enhanced anti-tumor activity. IMT-009 is under evaluation in a Phase 1/2a clinical trial for use as a monotherapy and combination treatment for solid tumor and hematological malignancies. The Phase 1 study is designed to evaluate the safety, tolerability, pharmacodynamic biomarkers, and preliminary efficacy of IMT-009 as well as identify the Recommended Phase 2 Dose (RP2D).

Nested Therapeutics Announces Oral Presentation of Preclinical Data for NST-628, a Novel, Fully Brain-Penetrant, Pan-RAF/MEK Molecular Glue, at the 2024 AACR Annual Meeting

On April 8, 2024 Nested Therapeutics, a biotechnology company pioneering a next-generation precision medicine platform to address hard-to-treat cancers, reported that preclinical data for the company’s lead program, NST-628, were featured in an oral presentation in the "New Drugs on the Horizon" series at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting (Press release, Nested Therapeutics, APR 8, 2024, View Source [SID1234641890]). The presentation, titled "NST-628 is a Novel, Potent, Fully Brain-Penetrant MAPK Pathway Molecular Glue that Inhibits RAS- and RAF-Driven Cancers," was given by Klaus Hoeflich, Ph.D., chief scientific officer and co-founder of Nested. The data were published concurrently online in the journal Cancer Discovery.

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!

"Dysregulation of RAS-MAPK pathway signaling is one of the most frequently occurring events in tumor development, impacting one in three newly diagnosed patients in the U.S. every year, the vast majority having no approved targeted treatment alternatives. While therapies have been developed for every node of the pathway, tolerability and durability of response continue to be challenging for patients with these difficult-to-treat cancers," said Dr. Hoeflich. "NST-628 was developed as a fully brain-penetrant, non-degrading molecular glue targeting the RAF and MEK nodes of the RAS-MAPK pathway. The preclinical data presented at AACR (Free AACR Whitepaper) show that NST-628 induces broad efficacy in tumor models and demonstrated the potential to overcome limitations of existing MEK and RAF inhibitors and RAS inhibitors in development. With a half-life and metabolic profile optimized to achieve a superior therapeutic index on a daily dosing schedule, as well as full intrinsic blood brain barrier penetrance, these data support NST-628’s potential as a best-in-class treatment for RAS- and RAF-driven cancers."

Preclinical data presented at AACR (Free AACR Whitepaper) and published in Cancer Discovery highlight the differentiated mechanism and drug-like properties of NST-628. Specifically:

In cellular and patient-derived tumor models harboring diverse KRAS, NRAS and BRAF alterations, NST-628 induced potent, deep and durable inhibition of the RAF-MEK signaling complex with broad efficacy without sacrificing tolerability at clinically achievable exposures over other MAPK-targeted compounds administered as either single agents or combinations.
With a predicted clinical half-life of 10-12 hours, the pharmacokinetic and metabolic profile of NST-628 has been optimized to broaden the therapeutic window and be clinically efficacious with once-daily oral dosing.
In mouse models with an intact blood brain barrier as well as central nervous system (CNS) models with RAS-MAPK alterations, NST-628 demonstrated full intrinsic CNS permeability. These data suggest NST-628 has the potential to treat brain metastases and primary CNS malignancies with MAPK pathway alterations. Approximately 40% of patients with metastatic cancer will develop symptomatic brain metastases, in particular in RAS-MAPK pathway dysregulated tumors such as lung, breast and melanoma.
Good laboratory practices (GLP) toxicology studies demonstrate significantly improved exposure margins when compared to other MEK inhibitors in non-clinical species.
Preclinical data support NST-628 as an ideal combination partner for upstream inhibitors including KRAS inhibitors by effectively preventing pathway reactivation.
In totality, the data validate NST-628’s potential to provide transformative clinical benefit as both a monotherapy or vertical combination anchor.
About the Phase 1 Study of NST-628
The ongoing Phase 1 open-label, single-arm, two-part study (NCT06326411) is intended to investigate the safety, pharmacokinetics (PK), pharmacodynamics (PD) and preliminary efficacy of single agent NST-628 in adult patients with RAS-MAPK pathway mutated/dependent advanced solid tumors, especially diverse KRAS, NRAS and BRAF alternations, who have exhausted standard treatment options. The study includes two parts: dose escalation (Part A) followed by dose expansion (Part B). The primary objectives for Part A, which recently initiated, are delineating NST-628’s safety profile and establishing the recommended dose for Part B. For more information, visit clinicaltrials.gov.

About NST-628
NST-628 is a fully brain-penetrant, mechanistically novel non-degrading molecular glue that targets multiple nodes in the RAS/MAPK pathway. NST-628 was developed based on Nested’s proprietary structural insights of how signaling complexes form and function in cancer and addresses common pitfalls of other MAPK-targeted compounds, which remain unable to circumvent the risk of intrinsic resistance via signaling pathway reactivation. Preclinical data evaluating oncology biomarkers relevant to RAS/MAPK-driven cell and patient-derived models collectively demonstrate superior anti-tumor activity, including in RAS and central nervous system-implanted tumor models, and tolerability of NST-628 compared to other MAPK-targeted compounds administered as either single agents or in combination. With a half-life and metabolic profile optimized to achieve a superior therapeutic index on a daily dosing schedule, as well as full intrinsic blood brain barrier penetrance, these data support NST-628’s potential as a best-in-class treatment especially for KRAS, NRAS and BRAF-driven cancers.

Phanes Therapeutics’ PT217 granted Fast Track Designation by the FDA

On April 8, 2024 Phanes Therapeutics, Inc. (Phanes), a clinical stage biotech company focused on innovative drug discovery and development in oncology, reported that the U.S. Food and Drug Administration (FDA) has granted Fast Track designation to PT217 for the treatment of patients with extensive-stage small cell lung cancer (ES-SCLC) with disease progression following platinum chemotherapy with or without a checkpoint inhibitor (Press release, Phanes Therapeutics, APR 8, 2024, View Source [SID1234641889]). PT217 was also granted orphan drug designation for the treatment of small cell lung cancer by the FDA in 2022.

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!

PT217, a first-in-class native IgG-like bispecific antibody (bsAb) targeting DLL3 and CD47, is being developed for the treatment of patients with small cell lung cancer (SCLC), large cell neuroendocrine carcinoma of the lung (LCNEC), and extrapulmonary neuroendocrine carcinomas (EP-NECs).

SCLC is an aggressive form of cancer characterized by high mortality rates and significant morbidities. The average survival for patients who present with ES-SCLC without treatment is 2-4 months after diagnosis; the median survival for patients with treatment is 7-11 months.

"PT217 has the potential to be a transformative treatment option for patients with SCLC, LCNEC and EP-NECs in various settings," said Ming Wang, Founder and CEO of Phanes Therapeutics. "Now we have two Fast Track designations in our pipeline. Last month, PT886 was granted Fast Track designation for the treatment of patients with metastatic claudin 18.2-positive pancreatic adenocarcinoma. Both assets are from our optimized anti-CD47 bispecific antibody franchise and currently in clinical studies. We believe the optimized anti-CD47 bispecific antibody approach can unlock the full potential of the innate immunity in targeting solid tumors."

The multi-center Phase I clinical trial of PT217 (NCT05652686), known as the SKYBRIDGE study, is currently evaluating the safety, tolerability, pharmacokinetics and preliminary efficacy of PT217 in patients with advanced or refractory cancers expressing DLL3.

Accent Therapeutics Presents Data Supporting Two Lead Programs at AACR 2024 Annual Meeting

On April 8, 2024 Accent Therapeutics, a biopharmaceutical company pioneering a novel class of small molecule precision cancer therapeutics reported advances in cancer targeting, including data on DHX9 and KIF18A inhibitor activity in multiple tumor types, at the 2024 American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting, taking place April 5-10 in San Diego, California (Press release, Accent Therapeutics, APR 8, 2024, View Source [SID1234641888]).

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!

"Accent has expanded the potential of our two lead programs in preclinical investigations, and we are excited for the promise they hold for addressing cancers with high unmet clinical need," said Robert A. Copeland, Ph.D., President, Founder, and Chief Scientific Officer of Accent Therapeutics. "Our results reveal that DHX9 and KIF18A inhibition are potent therapeutic strategies that stand to benefit large patient populations. These findings validate our systematic approach to tapping the rich therapeutic potential of our target space."

In the presentation entitled "RNA-Modifying Enzyme Inhibitors as Precision Cancer Therapeutics," Dr. Copeland delineates the promise of RNA-modifying enzyme (RME) inhibitors as precision cancer therapeutics that confer cancer-selective growth inhibition in vitro and in vivo. The potential of RME inhibitors is exemplified by inhibitors of the novel cancer target, DHX9, which display great promise as a novel treatment modality in BRCA1 and/or BRCA2 loss-of-function (LOF) cancers across multiple tumor types, including breast cancers.

Studies in breast cancer models demonstrate the potential impact of DHX9 inhibitor treatment for patients with defective DNA repair. These data are highlighted in a presentation entitled "DHX9 inhibition as a novel therapeutic for cancer with loss-of-function mutations in DNA damage repair genes BRCA1 and BRCA2." Additional key takeaways from this talk are:

DHX9 inhibition shows selective growth inhibition in breast cancer cell lines that exhibit BRCA LOF
DHX9 inhibition results in DNA damage and catastrophic increase in replication stress in BRCA LOF cell lines, causing cell cycle arrest prior to onset of apoptosis
In vivo, DHX9 inhibitors were well tolerated and demonstrate robust tumor growth inhibition in multiple BRCA LOF cell line xenograft models
Accent scientists further reinforce KIF18A – a kinesin that plays a critical role in facilitating mitosis – as a compelling therapeutic target in chromosomally instable tumors. In a poster entitled "Inhibition of KIF18A leads to mitotic arrest and robust anti-tumor activity in chromosomally instable tumors," they show data that Accent’s novel, potent and selective small molecule inhibitors of KIF18A:

Upregulate biomarker proteins, indicating specific induction of mitotic arrest, DNA damage, and apoptosis
Exhibit robust anti-proliferative activity, and result in fragmented nuclei and malformed mitotic spindles in chromosomally instable ovarian cancer cell lines
Induce cell cycle arrest in G2/M for sensitive cell lines, consistent with the role of KIF18A in facilitating mitosis
Lead to dose-dependent tumor growth inhibition in an OVCAR-3 cancer cell line-derived xenograft model, while sparing tumors that are insensitive to KIF18A loss
Data describing a DHX9-specific pharmacodynamic biomarker of inhibition are presented in a poster entitled "circBRIP1 RNA as a non-invasive target engagement pharmacodynamic biomarker for DHX9 inhibition." Key takeaways include:

Positive correlation of circBRIP1 induction with DHX9 biochemical activity, dose, and exposure for DHX9 inhibitors
Similar potency of dose-dependent induction of circBRIP1 across cell lines that are sensitive or insensitive to DHX9 inhibition, indicating that circBRIP1 is a unique target engagement PD biomarker, but not a predictor of DHX9 sensitivity
circBRIP1 induction correlates with DHX9 inhibitor concentration within individual tumors in mouse xenografts, providing strong evidence of a predictable PK/PD relationship
Select posters and presentations will be shared on Accent’s website following the meeting.

About DHX9
Accent’s lead program is a first-in-class DHX9 inhibitor with the potential to address high unmet need indications not adequately served by existing therapies, including tumors with BRCA loss of function (breast, ovarian, and others), mismatch repair deficient (dMMR) or microsatellite instability-high (MSI-H) cancers (colorectal, endometrial, gastric) and additional undisclosed cancer types representing large patient populations. DHX9 is a DNA/RNA helicase that has been reported to play important roles in replication, transcription, translation, RNA splicing, RNA processing, and maintenance of genomic stability. Hence, this enzyme represents a compelling novel oncology target as inhibition of DHX9 exploits key tumor vulnerabilities, resulting in cancer-specific death. Accent is currently conducting IND-enabling studies for this program.

About KIF18A
Accent’s second lead program is a potential best-in-class inhibitor for KIF18A which may address a large patient population across several cancer indications, including ovarian and triple negative breast cancer (TNBC). KIF18A is a mitotic kinesin motor protein critical for cell division in select tumors with chromosomal instability. A subset of tumor cells with an abnormal number of chromosomes (aneuploid) are reliant on KIF18A and show rapid cell killing in vitro and in vivo upon KIF18A inhibitor treatment, while cells with normal numbers of chromosomes (euploid) are unaffected. Accent is planning to initiate IND-enabling studies for KIF18A in the first half of 2024.

Transcenta Announces Collaboration with Agilent to Develop a Claudin18.2 Companion Diagnostic to Support Osemitamab (TST001) Global Phase III Trial

On April 8, 2024 Transcenta Holding Limited ("Transcenta") (HKEX: 06628), a clinical stage biopharmaceutical company with fully-integrated capabilities in discovery, research, development and manufacturing of antibody-based therapeutics, and Agilent Technologies, reported a collaboration to develop a Claudin18.2 (CLDN18.2) companion diagnostic to support TranStar301 global Phase III pivotal trial of Osemitamab (TST001) in combination with Nivolumab and chemotherapy as first-line treatment in patients with CLDN18.2 expressing locally advanced or metastatic gastric or gastroesophageal (G/GEJ) adenocarcinoma (Press release, Transcenta, APR 8, 2024, View Source [SID1234641887]).

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!

Transcenta has developed a mouse anti-CLDN18.2 monoclonal antibody, clone 14G11 which specifically binds to CLDN18.2 but not CLDN18.1. This antibody, generated against a linear epitope located on the extracellular domain of loop 1, has a binding site that overlaps with the binding site of therapeutic antibody Osemitamab (TST001).

Transcenta has been collaborating with Agilent, a world leader in CDx development, to further develop this antibody for use in a companion diagnostic assay. Agilent is developing Claudin18.2 IHC 14G11 pharmDx, an immunohistochemistry (IHC) assay for the detection of CLDN18.2 protein in gastric and gastroesophageal junction (GEJ) adenocarcinoma with the potential for other indications. Agilent and Transcenta presented the early results of the Claudin18.2 IHC 14G11 pharmDx assay at AACR (Free AACR Whitepaper) Annual Meeting. Claudin18.2 IHC 14G11 pharmDx for Investigational Use Only/for Performance Evaluation Only will be used for patient selection in the phase III trial of gastric/GEJ adenocarcinoma where applicable ethics committee and regulatory approvals have been granted.

"Agilent’s expertise in the development of companion diagnostics is impressive, as is their strong track record of developing companion diagnostics across the precision oncology sector," said Dr. Caroline Germa, Transcenta’s Executive Vice President, Global Medicine Development and Chief Medical Officer. "We are excited about the collaboration and look forward to working together to pave the way for enhanced patient health outcomes."

"We are excited to be working with Transcenta on the development of the Claudin18.2 IHC 14G11 pharmDx companion diagnostic assay," said Dr. Paul Beresford, VP/GM of CDx, Agilent. "This partnership with Transcenta will further pave the way for enhanced precision medicine products for those with gastric and gastroesophageal junction adenocarcinoma, and continue transforming diagnostics, treatments, and patient health outcomes."

About Osemitamab (TST001)

Osemitamab (TST001) is a high affinity humanized anti-CLDN18.2 monoclonal antibody with enhanced antibody-dependent cellular cytotoxicity ("ADCC"). It has shown potent anti-tumor activities in tumor xenograft models. Osemitamab (TST001) is the second most advanced CLDN18.2 targeting antibody being developed globally. Osemitamab (TST001) was generated using Transcenta’s Immune Tolerance Breaking Technology (IMTB) platform. Osemitamab (TST001) kills CLDN18.2 expressing tumor cells by mechanisms of ADCC. Leveraging advanced bioprocessing technology, the fucose content of Osemitamab (TST001) was significantly reduced during the production, which further enhanced NK cells mediated ADCC activity of Osemitamab (TST001). Clinical trials for Osemitamab (TST001) are ongoing in the U.S. and China (NCT05190575, NCT04396821, NCT04495296, NCT05608785/CTR20201281). Osemitamab (TST001) has been granted Orphan Drug Designation in the U.S. by FDA for the treatment of patients with gastric or gastroesophageal junction (G/GEJ) and pancreatic cancer.