Veneno Technologies, a Novel DRP Drug Discovery Company, Announces Joint Research Agreement with Sumitomo Pharma Co. Ltd.

On April 18, 2023 Veneno Technologies Co. Ltd. is reported that the company has entered into a joint research agreement with Sumitomo Pharma Co. Ltd., headquartered in Osaka, Japan (Press release, Veneno Technologies, APR 18, 2023, View Source [SID1234630287]).

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 is the second such research agreement with a major Japanese research company we have completed this month. We are particularly pleased to be working with researchers at Sumitomo Pharma Co. Ltd." said Kazunori Yoshikawa, President and CEO at Veneno Technologies Co. Ltd.

Under the terms of the agreement, Veneno Technologies [View Source will conduct a program to obtain functional peptides (DRPs) for ion channels targeted by Sumitomo Pharma using Veneno Technologies’ next-generation peptide discovery technology, the PERISS method (intra-periplasm secretion and selection). Work on this project is expected to begin during the 2nd Quarter, 2023.

For inquiries regarding this release, please contact:
Veneno Technologies Co. Ltd.
[email protected]

Orum Therapeutics Unveils New Program at AACR 2023, Highlighting Cbl-b Inhibitor-PD-1 Conjugate, and Presents New Data Supporting Clinical Development of Two GSPT1 Programs

On April 18, 2023 Orum Therapeutics, a clinical-stage private biotechnology company pioneering cell-specific targeted protein degradation (TPD² ) and targeted protein stabilization (TPS² ), reported new preclinical data for three of Orum’s therapeutic programs: ORM-5029, which is a proprietary GSPT1 degrader conjugated to HER2-targeting antibody pertuzumab; ORM-6151, which delivers GSPT1 degrader to CD33+ tumors; and its new immuno-oncology program for a Cbl-b inhibitor (Cbl-bi) conjugated to anti-PD-1 antibody pembrolizumab (Press release, Orum Therapeutics, APR 18, 2023, View Source [SID1234630289]). The data were presented in three posters at the 2023 American Association for Cancer Research (AACR) (Free AACR 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!

"We are pleased to present new data supporting clinical development of two programs from our TPD² GSPT1 Degrader Platform, which was developed to improve the therapeutic window and realize the full potential of GSPT1 degraders through precision delivery to cancer cells via antibody drug conjugates"

Tweet this
"We are pleased to present new data supporting clinical development of two programs from our TPD² GSPT1 Degrader Platform, which was developed to improve the therapeutic window and realize the full potential of GSPT1 degraders through precision delivery to cancer cells via antibody drug conjugates," said Peter Park, Ph.D., Chief Scientific Officer of Orum Therapeutics. "We are also excited to unveil at AACR (Free AACR Whitepaper) our new TPS² platform, which has the potential to achieve the full promise of Cbl-b inhibitors through T cell-specific delivery. With this novel approach, we show that we can limit unwanted systemic exposure, including the risk of toxicity associated with c-Cbl inhibition, prolong the activation of exhausted PD-1+ T cells, and provide resistance to negative environmental cues such as TGF-β or regulatory T cells. We believe our approach of harnessing the power of protein degraders and stabilizers with the precision of antibody targeting will improve the treatment of cancer for more patients."

Details of the data presented are as follows:

Title: A novel antibody-enabled Dual-precision Targeted Protein Stabilization (TPS²) that augments anti-tumor immune response by targeting Cbl-b inhibitor to exhausted T cells while blocking checkpoint molecule, PD-1
Presenter: Joanne Lim, Ph.D.
Abstract Number: 4436
Summary: Cbl-b inhibition results in stabilization of intracellular proteins downstream of the T cell activation pathways. Using a murine model well-established to be refractory to anti-PD-1 antibody treatment, we demonstrated that anti-PD-1-Cbl-b inhibitor approach increased effector T cell markers in the tumor and delayed tumor growth.
Presented Data:

The delivery of Cbl-b inhibitor (Cbl-bi) conjugated to anti-PD-1 antibody enhances the saturating, maximum activation of anti-PD-1 antibody by 300% in PD-1/PD-L1 blockade assay.
Anti-PD-1-Cbl-bi conjugates enhance T cell responses of mixed lymphocyte reaction in a dose-dependent manner and overcome suppressive effects of TGF-β and Treg cells compared to antibody alone in vitro.
Anti-PD-1-Cbl-bi conjugates enhance ex vivo activation of tumor-associated T cells from human cancer patients compared to antibody alone or when treated with antibody and unconjugated Cbl-bi.
Anti-PD-1-Cbl-bi conjugates delay the growth of melanoma tumors in hPD-1 transgenic mice, with corresponding increase in intratumoral transcript levels of CD3e, granzyme B, and perforin in the tumor.
Title: ORM-6151: A first-in-class CD33-antibody enabled GSPT1 degrader for AML
Presenter: James Palacino, Ph.D.
Abstract Number: 2700
Summary: ORM-6151 shows robust single-dose efficacy, both in vitro and in vivo with superiority or comparability to standard of care (SOC) agents. In addition, in vitro testing demonstrated superior tolerability to healthy bone marrow progenitor cells, suggesting better tolerability. Responses in representative TP53 wild-type and mutant AML models demonstrated comparable activity, indicating the potential for responses in a large percentage of AML patients with poor treatment options.
Presented Data:

ORM-6151 shows robust in vitro activity in p53 wild-type and mutant cell models.
In ex-vivo AML blasts, ORM-6151 demonstrates efficacy superior to CC-90009, a small-molecule GSPT1 degrader, or Mylotarg, an FDA-approved treatment for AML.
Compared to CC-90009 or Mylotarg, ORM-6151 exhibits minimal cytotoxic activity to healthy hematopoietic progenitor cells in vitro.
A single treatment of ORM-6151 at 3 mg/kg regresses MV4-11 tumors in vivo mouse model with 9/9 complete response (CR), and a single dose of 1 mg/kg produced equivalent antitumor effect as SOC doublet given at optimal repeated dose.
Title: Development of RNAscope multiplex-based assay for exploratory pharmacodynamic biomarkers assessment in breast cancer patients from Phase I clinical trial of ORM-5029, a potent GSPT1 degrader
Presenter: Shikha Saini, Ph.D.
Abstract Number: 2118
Summary: To fulfill the need to develop pharmacodynamic biomarkers as predictors of efficacy to support clinical development of ORM-5029, Orum identified a novel GSPT1 antibody for detection of depletion following treatment with ORM-5029. GSPT1 depletion and subsequent activation of the integrated stress response are both seen following in vitro or in vivo treatment with ORM-5029. Degree of pharmacodynamic (PD) change correlates with depth and duration of in vivo response. Orum is currently developing a multiplexed (IF/ISH/ISH) assay to track PD responses in patients, with an aim to better predict responses and efficacious doses.
Presented Data:

Orum identified the depletion of GSPT1 as a target-proximal pharmacodynamic biomarker in ORM-5029 treated in vitro and in vivo samples.
Loss of GSPT1 drives activation of integrated stress response and upregulation of ATF3 and DDIT3 mRNA and these responses are also seen following treatment with ORM-5029, in both in vitro BT-474 cells and in vivo HCC1568 mouse model.
PD responses correlate with degree and duration of response in an in vivo model treated with ORM-5029.
The posters are available to download through the links below:
PD-1-Cbl-bi TPS² preclinical poster: View Source
ORM-6151 preclinical poster: View Source
ORM-5029 predictive biomarker poster: View Source

Repare Therapeutics Presents Initial Clinical Data from the Phase 1/2 TRESR and ATTACC Trials Evaluating Camonsertib in Combination with Three PARP Inhibitors at the 2023 AACR Annual Meeting

On April 18, 2023 Repare Therapeutics Inc. ("Repare" or the "Company") (Nasdaq: RPTX), a leading clinical-stage precision oncology company, reported initial data from its ongoing Phase 1/2 TRESR clinical trial evaluating camonsertib (RP-3500/RG6526, partnered with Roche), a potent and selective oral small molecule inhibitor of ATR (Ataxia-Telangiectasia and Rad3-related protein kinase), in combination with a poly (ADP-ribose) polymerase inhibitor (PARPi), talazoparib, and initial data from its ongoing Phase 1b/2 ATTACC clinical trial, evaluating camonsertib in combination with two additional PARPis, niraparib or olaparib, in patients with advanced solid tumors (Press release, Repare Therapeutics, APR 18, 2023, View Source [SID1234630288]).

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 data involving novel combinations of low doses of camonsertib and three different PARPis are featured today at the 2023 AACR (Free AACR Whitepaper) Annual Meeting in a clinical plenary session titled, "Safety and efficacy of three PARP inhibitors (PARPi) combined with the ataxia telangiectasia- and Rad3-related kinase inhibitor (ATRi) camonsertib in patients (pts) with solid tumors harboring DNA damage response (DDR) alterations" (abstract presentation number CT018). This study population comprised patients with a broad range of historically difficult to treat tumors, including patients with platinum-resistant tumors, patients who had either recurred or progressed during or after treatment with PARPis, and patients who had developed known BRCA-reversion mutations.

"We see promise in the camonsertib-PARPi combinations when administered concomitantly, at low doses across tumor types, especially in recurrent ovarian cancer given that nearly all had recurred after prior PARPi treatment. We are particularly encouraged by the depth of response and duration of treatment," said Maria Koehler, MD, PhD, Chief Medical Officer of Repare. "Dose optimization to refine the combinatorial dose in additional tumor-specific expansions beyond ovarian within our ATTACC study is ongoing as part of our collaboration with Roche."

"We previously established a promising safety and early efficacy profile of camonsertib as a monotherapy and this year’s data at AACR (Free AACR Whitepaper) further support camonsertib as a partner for combinational regimens and provides a clear rationale for further development of this compound," said Lloyd M. Segal, President and Chief Executive Officer of Repare. "Notably, the circulating tumor DNA (ctDNA) data showed a strong correlation with the degree of tumor shrinkage and duration of disease control, and provide a mechanistic explanation for the observed durable clinical benefit in heavily pretreated patients, beyond the natural history of the disease. We look forward to refining our dose optimization efforts and efficacy assessment in tumor specific expansions. This data remains consistent with what we were anticipating at the time of entering our partnership with Roche and we are excited to continue this important clinical development together."

Key Initial Findings from the TRESR Phase 1/2 and ATTACC 1b/2 PARPi Combination Studies:

TRESR (NCT04497116) is a first-in-human, multi-center, open-label Phase 1/2 dose-escalation and expansion study, designed to establish the recommended Phase 2 dose and schedule, evaluate safety and pharmacokinetics and identify preliminary anti-tumor activity associated with camonsertib, given alone and in combination with talazoparib or in combination with gemcitabine.

ATTACC (NCT04972110) is a first-in-human, multi-center, open-label Phase 1b/2 dose-escalation and expansion study, designed to evaluate safety and pharmacokinetics and identify preliminary anti-tumor activity associated with camonsertib in combination with niraparib or olaparib.

The clinical plenary session described initial combination Phase 1/2 results from 107 patients, of which 90 patients were evaluable for efficacy treated at least 13 weeks prior to the data cutoff of February 27, 2023.

Key highlights from the data presented at the 2023 AACR (Free AACR Whitepaper) Annual Meeting include:

Camonsertib combination resulted in durable clinical benefit across tumor types and different genomic alterations, regardless of choice of PARPi and presence of platinum resistance. Overall clinical benefit rate (CBR) for all patients was 48%. Patients with platinum-resistant tumors had an overall response rate (ORR) of 12% and CBR of 49%, and benefited similarly to non-platinum-resistant tumors (ORR 13%, CBR 46%).
Compelling results were observed particularly in patients with advanced ovarian cancer (n = 19). In these patients, overall response was 32%, CBR was 58% and median progression-free survival (mPFS) was approximately 7 months with treatment >16 weeks and ongoing in 9 patients.
Early ctDNA molecular responses in 66% (31/47) of evaluable patients confirms antitumor activity of low dose, intermittent PARPi + ATRi therapy. The molecular response rate (MRR) was significantly higher in patients with clinical benefit (83%) compared to those without (48%; p=0.015), confirming treatment effect. Molecular responses were observed in patients with prior PARPi exposure (57%) and platinum resistance (64%).
Camonsertib combinations appear to be well tolerated. Dose limiting toxicity (DLTs) in 68 patients treated with the proposed combination doses were related to myelotoxicity only (Grade 3+ anemia 3%, thrombocytopenia 6%, neutropenia 7%, and febrile neutropenia 3%). No prophylactic growth factors were required when administering the PARPis at evaluated doses.
Additional relevant presentations at AACR (Free AACR Whitepaper):

Title: Characterization of CCNE1 amplifications and associated genomic features in ovarian and uterine cancers
Session: Biomarkers of Therapeutic Benefit 5, Tuesday April 18, 2023, 1:30 PM – 5:00 PM
Abstract Number: 5469

Title: Tumor heterogeneity of CCNE1 copy number assessed by fluorescence in situ hybridization (FISH) in ovarian and uterine cancers and correlation with cyclin E protein expression
Session: Biomarkers of Therapeutic Benefit 2, Monday April 17, 2023, 9:00 AM – 12:30 PM
Abstract Number: 2132

Title: Targeting PKMYT1 kinase is an effective treatment strategy in triple negative breast cancers with low molecular weight cyclin E (LMW-E) expression
Session: Biomarkers of Therapeutic Benefit 1, Sunday April 16, 2023, 1:30 PM – 5:00 PM
Abstract Number: 950

Title: Investigating Wee1 and Myt1 combined inhibition as a potential cancer therapeutic strategy
Session: Combination Therapies for Cancer, Tuesday April 18, 2023, 1:30 PM – 5:00 PM
Abstract Number: 5511

Palleon Pharmaceuticals Presents Initial Phase 1 Results from the GLIMMER-01 Clinical Trial for E-602, the First Ever Glyco-Immune Checkpoint Inhibitor

On April 18, 2023 Palleon Pharmaceuticals, a clinical-stage company pioneering glyco-immunology drug development to treat cancer and inflammatory diseases, reported the initial Phase 1 results from the GLIMMER-01 trial of its lead EAGLE oncology platform program, E-602, at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting 2023 in Orlando, Florida (Press release, Palleon Pharmaceuticals, APR 18, 2023, View Source [SID1234630286]).

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 study demonstrated proof of mechanism for E-602, a first-in-class glyco-immune checkpoint inhibitor, including dose dependent desialylation and dose dependent immune system activation. Additionally, E-602 was found to be well tolerated across the entire dose range evaluated with no dose limiting toxicities. Having met pre-specified gating criteria based on safety and pharmacodynamic activity, Palleon announced plans to proceed with Phase 2 evaluation.

"E-602 represents a truly novel immuno-oncology candidate that builds on the promise of glycobiology, a field of study that offers unprecedented opportunities for drug development in cancer and other diseases," said David Feltquate, M.D., Ph.D., Chief Medical Officer of Palleon Pharmaceuticals. "Establishing proof of mechanism for this approach provides the validation necessary to initiate Phase 2 evaluation and take next steps towards our team’s shared goal of offering new options for patients in need."

"It is exciting to see the first clinical results generated from Palleon’s EAGLE glyco-immunology drug development platform," said Carolyn Bertozzi, Ph.D., Palleon Scientific Co-Founder and 2022 Nobel laureate. "Palleon is advancing an entirely new axis of immune modulation, defined by the interaction between sialoglycans and their receptors, which may benefit patients who do not respond to current medications."

In the initial results from the GLIMMER-01 trial presented by lead investigator, Jason Luke, M.D., of UPMC Hillman Cancer Center, 40 patients were treated with at least one dose of E-602, the most common tumors treated being colorectal (n=21) and pancreatic (n=10). Doses up to 30 mg/kg were tolerated with no dose-limiting toxicities. Both dose-dependent desialylation and immune system activation, as measured by increases in activated (CD69+) immune cells in circulation and elevation of circulating cytokines and chemokines including IP-10/CXCL10, were observed. Phase 2 studies will evaluate clinical activity of E-602 monotherapy in patients with lung cancer and melanoma.

The presentation can be found in the Palleon Publications section of the Education Hub page of our website.

Exai Bio Presents New Non-Small Cell Lung Cancer Early Detection Data at the American Association for Cancer Research (AACR) 2023 Annual Meeting

On April 18, 2023 Exai Bio reported new data demonstrating that its novel RNA- and AI-based platform detected non-small cell lung cancer with high accuracy, at the earliest stages and for the smallest tumors, in both training and validation cohorts (Press release, Exai Bio, APR 18, 2023, View Source [SID1234630285]). In the independent validation cohort, stage I sensitivity was 96% and tumor size T1a-b sensitivity was 94%, at 90% specificity. These findings continue to highlight the power of Exai’s proprietary technology to detect cancer at its earliest stages from a blood draw. This study will be presented today at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) 2023 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!

"These results demonstrate the unique advantages of our RNA-based liquid biopsy platform in early detection," said Patrick Arensdorf, Chief Executive Officer of Exai. "We set the bar high in this study, emphasizing small and early stage tumors, in order to address head-on the unmet need for earlier lung cancer detection. Exai’s platform reveals actionable insights into novel cancer biology in order to achieve our ultimate goal of improving patient outcomes."

Today’s non-small cell lung cancer results at AACR (Free AACR Whitepaper) expand a growing body of evidence that Exai’s platform can be used across multiple tumor types and clinical applications, using standard blood samples. Previously, Exai has presented breast cancer early detection and screening data at the San Antonio Breast Cancer Symposium (SABCS) 2022, monitoring and molecular residual disease detection data in breast cancer at SABCS 2021, and colorectal cancer early detection and screening data at the 2022 European Society for Medical Oncology (ESMO) (Free ESMO Whitepaper) meeting.

Exai’s platform uses RNA sequencing to identify a novel category of cancer-associated, small non-coding RNAs, termed orphan non-coding RNAs (oncRNAs). OncRNAs are actively secreted from living cancer cells and are stable and abundant in the blood of cancer patients. Exai has created a catalog of hundreds of thousands of oncRNAs and thousands of patient oncRNA profiles, spanning all major cancer types. When combined with Exai’s proprietary artificial intelligence, this unique platform has several scientific and practical advantages over tests that focus on circulating tumor DNA including sensitivity, specificity and informative properties for active cancer biology. Exai’s universal platform can be used across multiple cancer care settings such as screening and early detection, monitoring, molecular residual disease and therapy selection.

AACR Presentation Details

Presentation Title: Blood-based Early Detection of Non Small Cell Lung Cancer Using Orphan Noncoding RNAs

Session Title: Increasing the Clinical Utility of Cell Free DNA Testing

Date: Tuesday, 4/18/23, 2:30 – 4:30 PM EDT

Authors: Mehran Karimzadeh, Jeffrey Wang, Aiden Sababi, Dare Afolabi, Ti Lam, Alice Huang, Diana Corti, Kristle Garcia, Seda Kilinc, Allen Zhao, Jeff J Wang, Taylor Cavazos, Patrick Arensdorf, Kimberly Chau, Helen Li, Hani Goodarzi, Lisa Fish, Fereydoun Hormozdiari, Babak Alipanahi