Medigene Presents Favorable Safety Profile of TCR-T Cells with Costimulatory Switch Protein at AACR Annual Meeting 2024

On April 8, 2024 Medigene AG (Medigene or the "Company", FSE: MDG1, Prime Standard), an immuno-oncology platform company focusing on the discovery and development of T cell immunotherapies for solid tumors, reported superior recombinant T cell receptor (rTCR) engineered T cell functionality as well as a favorable safety profile when rTCR-T cells are armored and enhanced with the PD1-41BB costimulatory switch protein (CSP) at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting 2024 taking place April 5-10, 2024, in San Diego, USA (Press release, MediGene, APR 8, 2024, View Source [SID1234641844]).
The poster presentation with the title "TCR-gated control of costimulatory switch protein (CSP) activation in rTCR-T cells expressing PD1-41BB" is available on Medigene’s website at View Source

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!

Targeting solid tumors with TCR-T therapies still faces significant challenges. Impaired T cell functionality and T cell exhaustion are driven by several factors within the hostile solid tumor microenvironment (TME). Programmed cell death ligand-1 (PD-L1), expressed by tumor cells in the TME engages the programmed cell death protein-1 (PD-1) expressed by activated T cells and induces T cell exhaustion and facilitates tumor immune escape. This is one major factor that allows cancer cells to proliferate and metastasize without being recognized by the host immune system. To counteract this inhibitory mechanism, a PD1-41BB CSP can be co-expressed in rTCR-T cells, turning an inhibitory signal mediated via the PD-1-PD-L1 axis into a costimulatory signal that improves TCR-T cell functionality.

"Medigene’s End-to-End Platform provides differentiated approaches to address the key challenges, including the immunosuppressive TME of solid tumors, in developing effective, safe and durable TCR-T therapies. The PD1-41BB CSP is our proprietary armoring and enhancement technology that improves immune function and persistence of TCR-T cells in the TME, resulting in better efficacy and sustained anti-cancer immune responses," said Dolores Schendel, Chief Scientific Officer of Medigene. "This latest data from our lead program MDG1015, a first-in-class, 3rd generation TCR-T therapy targeting NY-ESO-1/ LAGE-1a, armored and enhanced with our PD1-41BB CSP, shows the gating of the PD1-41BB effects through prior cancer antigen engagement with our 3S TCR. It represents a safe and effective approach to improve clinical outcomes in hard-to-treat solid tumor indications such as gastric cancer, ovarian cancer, myxoid/round cell liposarcoma, and synovial sarcoma. Based on our positive interactions for MDG1015 with regulatory authorities, we look forward to progressing our package for IND / CTA submission and expect to receive IND/CTA approval in the second half of 2024."

The data presented in the poster demonstrated that the CSP-mediated costimulatory signal is TCR-gated, such that costimulation only takes place when a specific peptide-human leukocyte antigen (pHLA) complex is present on a tumor cell and triggers a signal through the rTCR expressed by the TCR-T cells. Enhanced, gated T cell functionality of CSP-armored rTCR-T cells increased secretion of interferon-γ (IFNγ) only when tumor cells simultaneously expressed the pHLA target antigen and PD-L1. In addition, CSP-armored rTCR-T cells showed high sensitivity in recognition of diverse tumor cell lines of different tissue origin, such as melanoma, sarcoma, and gastric cancer which varied in levels of pHLA and PD-L1 in vitro. Rapid and sustained killing of 3D tumor cell-derived spheroids only occurred when PD-L1-positive tumor cells simultaneously expressed the specific pHLA target antigen.

Importantly, no recognition of healthy cells occurred if they lacked the pHLA target antigen, irrespective of PD-L1 expression, underpinning the safety of combining the CSP with a rTCR to generate rTCR-T cells that displayed no signs of toxicity for diverse healthy tissues in vitro.

Imugene’s oncolytic virotherapy VAXINIA and B cell immunotherapy HER-Vaxx featured at the AACR Annual Meeting 2024

On April 8, 2024 Imugene Limited (ASX: IMU), a clinical stage immuno-oncology company, reported poster presentations featuring its CF33 oncolytic virotherapy VAXINIA and B cell immunotherapy HER-Vaxx at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting 5-10 April 2024, in San Diego, CA.

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!

Daneng Li, M.D., a City of Hope associate professor in the Department of Medical Oncology & Therapeutics Research commented, "The results show that our novel oncolytic virus — both alone or in combination with immunotherapy — has the ability to control various cancer types previously resistant to other treatment options, and these early results provide patients with hope of a new treatment option for cancers refractory to standard treatment."

Imugene Managing Director & CEO Leslie Chong said: "Preliminary data from the MAST trial demonstrates encouraging anti-tumour activity with our oncolytic virus CF33-hNIS (VAXINIA). Notably, one patient with cholangiocarcinoma, or biliary tract cancer, achieved an immunological complete response (CR), meaning the disappearance of all signs of their cancer after treatment with VAXINIA, with no known recurrence after one year. These encouraging results warrant further investigation in patients will biliary tract cancer and other cancers. In addition, further analysis of the T cell repertoire reveals that T cell diversity may serve a predictive biomarker, which can be used to prospectively identify appropriate patients for treatment."

Details on the poster presentations are below:

Presentation Title: Oncolytic virus CF33-hNIS for the treatment of advanced cancer
Abstract Presentation Number: CT182
Session Date and Time: Tuesday April 9, 2024, 9:00 AM – 12:30 PM PT
Session Title: First-in-Human Phase I Clinical Trials 2
Presenter: Daneng Li, MD
Highlights include:

At the data cut-off of 19 December 2023, there were 31 efficacy-evaluable patients in the MAST study. In the intratumoural (IT) cohorts, 7 of 16 (44%) injected lesions had a reduction in tumour burden and 3 lesions were completely eradicated.Three of the IT treated patients had an objective response: 1 complete response by iRECIST in a patient with cholangiocarcinoma and 2 partial responses by RECIST inpatients with melanoma. In the intravenous (IV) cohorts, 9 of 17 (53%) patients achieved stable disease as their best response. Patients who received prior checkpoint blockade therapy derived clinical benefit with and without pembrolizumab. Viral replication, assessed by SPECT, was higher in patients who had a reduction in tumour burden.
Patients with a higher level of T cell diversity in peripheral blood (pre-treatment)respond better to VAXINIA therapy, consistent with the known mechanism of action of oncolytic virotherapies and their ability to promote an anti-tumour T cell response.
Both IT- and IV-treated patients have promising immunological changes in on-treatment tumour biopsies (including increases in cancer fighting CD8 T cells and PD-L1 expression) indicated that VAXINIA can transform the tumour microenvironment.
Several patients have had prior treatment with checkpoint blockade, including astable disease cholangiocarcinoma patient and two melanoma partial response patients. This suggests that VAXINIA +/- checkpoint inhibitor combination could be used in the checkpoint therapy refractory setting, which is seeing a growing and unmet market in oncology, by altering the tumour microenvironment.

Presentation Title: Frontline vaccination with the B-cell peptide compound HER-Vaxx (IMU-131), combined with standard-of-care chemotherapy induces high levels of HER2-specific antibodies mediating ADCC and intracellular phosphorylation inhibition resulting in overall survival benefit in patients with HER2+ metastatic or advanced gastric/GEJ adenocarcinoma – Final results from Phase II/HERIZON study
Poster Number: CT215
Session Date and Time: Tuesday April 9, 2024, 9:00 AM – 12:30 PM PT
Session Title: Phase II Clinical Trials 1
Presenter: Joshua Tobias Ph.D.
Highlights include:

HER-Vaxx treatment produced robust anti-HER2-IgG and IgG1 antibody response (p<0.001).
HER-Vaxx induced HER2-specific antibodies able to mediate antibody-dependent cell cytotoxicity (ADCC) and inhibit intracellular HER2 phosphorylation and correlated with tumour reduction.
The HER-Vaxx induced HER2-specific antibodies demonstrate a similar mechanism of action to HERCEPTIN validating B cell immunotherapy as an alternative anti-cancer agent to monoclonal antibodies.
About the MAST Trial
The multicenter Phase 1 MAST trial commenced by delivering a low dose of VAXINIA to patients with metastatic or advanced solid tumours who have had at least two prior lines of standard of care treatment. The City of Hope-developed oncolytic virus has been shown to shrink colon, lung, breast, ovarian and pancreatic cancer tumours in preclinical laboratory and animal models¹. Overall, the study aims to recruit cancer patients across approximately 10 trial sites in the United States and Australia.
The clinical trial is titled "A Phase I, Dose Escalation Safety and Tolerability Study of VAXINIA (CF33- hNIS), Administered Intratumorally or Intravenously as a Monotherapy or in Combination with Pembrolizumab in Adult Patients with Metastatic or Advanced Solid Tumours (MAST)." The trial commenced in May 2022 and is anticipated to run for approximately 24 months while being funded from existing budgets and resources. Full study details can also be found on clinicaltrials.gov under study ID: NCT05346484.

HiFiBiO Therapeutics Presents Two Posters at the 2024 American Association for Cancer Research Annual Meeting

On April 7, 2024 HiFiBiO Therapeutics, a leading clinical stage global biotechnology company committed to advancing patient outcomes through single-cell precision, reported two posters at the AACR (Free AACR Whitepaper) 2024 meeting held on April 5-10, 2024, in San Diego, CA (Press release, HiFiBiO Therapeutics, APR 7, 2024, View Source [SID1234641870]).

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!

"One of the major challenges cancer immunotherapies face is the difficulties in selecting patients with tumors that will respond. Single-cell technologies can provide valuable insights into disease biology and help discern tumors that are more likely to respond; however, the lack of integrated data across diverse single-cell platforms hinders the potential of these novel technologies to inform clinical decisions." Jack Russella-Pollard, Ph.D., Executive Director, Translational Data Science, commented, "HiFiBiO has built an AI/ML powered curation and data integration process within our Drug Intelligence Science (DIS) translational platform that integrates publicly available datasets with in-house generated datasets to predict tumor response and guide target as well as indication selection."

Jinping Gan, Ph.D., Vice President, Global Head of Research, remarked: "Currently, there is no suitable model to describe complex tumor-immune interactions that can effectively guide clinical decisions. HiFiBiO has successfully constructed semi-mechanistic PKPD models for an anti-TNFR2 agonist and an anti-OX40 agonist that advocate for pulsatile agonism to achieve optimal anti-tumor efficacy. This innovative scientific work serves as a valuable tool for guiding dose and dose regimen optimization for the humanized versions of these T cell co-stimulatory agonists in clinical development."

By harnessing these cutting-edge technologies and tools within its DIS platform, HiFiBiO Therapeutics is steering the development of its drug candidates, including three antibodies currently undergoing Phase 1 evaluation: a first-in-class TNFR2 agonist (HFB200301, NCT05238883), a next-generation OX40 agonist (HFB301001, NCT05229601), and a best-in-class BTLA antagonist (HFB200603, NCT05789069).

These presentations underscore HiFiBiO Therapeutics’ unwavering commitment to innovation and its relentless pursuit of novel therapeutic solutions to address unmet medical needs.

Details on the poster presentations are as follows. E-Posters will be posted on HiFiBiO’s website following the live presentations.

Abstract Number: 6202

Title: Integrating public single-cell transcriptomics and patient profiles to guide clinical development

Presenter: Jack Russella-Pollard, Ph.D., Executive Director, Translational Data Science
Session Category: Bioinformatics / Computational Biology / Systems Biology / Convergent Science

Session Title: Integrative Cancer Science

Session Date and Time: Tuesday Apr 9, 2024, 1:30 PM – 5:00 PM

Location: Poster Section 36

Poster Board Number: 5

View full abstract here.

Abstract Number: 7176

Title: Optimization of T cell co-stimulatory agonists: A semi-mechanistic PKPD model integrating drug properties and tumor-immune interactions

Presenter: Jinping Gan, Ph.D., Vice President, Global Head of Research

Session Category: Experimental and Molecular Therapeutics

Session Title: Pharmacology and Pharmacogenetics

Session Date and Time: Wednesday Apr 10, 2024, 9:00 AM – 12:30 PM

Location: Poster Section 24

Poster Board Number: 17

View full abstract here.

Edgewood Oncology Announces Positive Efficacy Data From Investigator-Sponsored Study of BTX-A51 in Preclinical Models of Liposarcoma

On April 7, 2024 Edgewood Oncology, a clinical-stage biotechnology company focused on delivering BTX-A51 to patients with hematologic malignancies and genetically-defined solid tumors, reported the publication of new preclinical data from a study of BTX-A51 in human liposarcoma (LPS) conducted by Dana-Farber Cancer Institute and Hebrew University-Hadassah Medical School to be presented at the AACR (Free AACR Whitepaper) Annual Meeting 2024 in San Diego (Press release, Edgewood Oncology, APR 7, 2024, View Source [SID1234641851]). BTX-A51 is a first-in-class, small molecule, multi-selective kinase inhibitor of casein kinase 1 alpha (CK1α) as well as the transcriptional regulators cyclin-dependent kinases 7 and 9 (CDK7 and CDK9) that synergistically co-targets master regulators of cancer to promote programmed cell death, or apoptosis.

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 presentation (Abstract 604), "Targeting casein kinase 1 alpha (CK1alpha) and transcriptional CDKs (CDK7/9) in human liposarcomas," highlighted findings for BTX-A51 in preclinical human models of LPS. The data demonstrate that BTX-A51 has preclinical efficacy in treating patient-derived LPS in cell lines and human xenograft models and provides insight into the synergy gained by inhibiting both CK1α and CDK9.

"Dedifferentiated liposarcomas (DDLPS) are rare tumors derived from precursors of fat cells which can occur anywhere in the body. Patients with metastatic DDLPS face a life-threatening disease in need of more effective therapeutic options, which we hope to develop by taking advantage of vulnerabilities in LPS that have been uncovered by recent research on the abnormal DNA and other molecular characteristics of LPS," said George Demetri, M.D., Professor of Medicine and director of the Sarcoma Center at Dana-Farber. "Based on these data, we believe that it is justified to move BTX-A51 into a clinical trial for patients with LPS, which we will plan to open soon for enrollment."

Liposarcomas are universally characterized by amplification of the mouse double minute 2 homolog (MDM2) gene, which leads to destruction of the normal (non-mutated, also known as "wild type") p53. The data demonstrate that BTX-A51 significantly reduces the abnormal overexpression of MDM2 in multiple patient-derived LPS cell lines, leading to upregulation of p53 expression and ultimately apoptosis. Finally, BTX-A51 was demonstrated to be well-tolerated and effective in an LPS patient-derived xenograft model.

"We’re pleased that investigators at Dana-Farber have taken an interest in BTX-A51, which is currently being evaluated in a Phase 2 combination study with azacitidine in patients with relapsed or refractory (R/R) AML," said David N. Cook, Ph.D., chief executive officer of Edgewood Oncology. "These data also suggest the potential for BTX-A51 in cancers with MDM2 amplifications, a group of genetically defined tumors that span multiple tissue types suggesting broader potential."

Additional Details about the Study

Using three different CDK9-selective inhibitors in human LPS cell lines, the authors showed that CDK9 inhibition suppressed cell growth and induced apoptosis by decreasing MDM2 levels while inducing expression of p53. It was also demonstrated that the potency of CDK9 inhibitors is significantly enhanced upon CK1α depletion. These data led to the investigation of BTX-A51, which targets CK1α and CDK9 as well as CDK7, in LPS cells and has previously been shown to inhibit these kinases with nanomolar efficacy in AML models. BTX-A51 potently reduced expression of MDM2 with marked induction of p53, resulting in apoptosis of LPS cells. BTX-A51 also reduces expression of MCL1 and primes LPS cell lines and primary LPS cells for BIM-induced apoptosis, as demonstrated by BH3 profiling. Importantly, preliminary in vivo data in an LPS patient-derived xenograft model reveal that BTX-A51 is well-tolerated under conditions that inhibit tumor growth.

Affini-T Therapeutics Presents Preclinical Data from its Oncogenic Driver Programs Targeting KRAS G12D and p53 R175H at the American Association for Cancer Research (AACR) Annual Meeting 2024

On April 7, 2024 Affini-T Therapeutics, Inc., a precision immunotherapy company unlocking the power of T cells against oncogenic driver mutations, reported that management is presenting data from the company’s oncogenic driver programs targeting HLA-A*11:01 KRAS G12D and HLA-A*02:01 p53 R175H at this year’s American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting 2024 in San Diego (Press release, Affini-T Therapeutics, APR 7, 2024, View Source [SID1234641850]). In addition, the team is presenting two trial-in-progress posters for Affini-T’s Phase 1 clinical-stage programs targeting KRAS G12V, the company-sponsored AFNT-211 study and the Fred Hutchinson Cancer Center investigator-initiated AFNT-111 study at AACR (Free AACR Whitepaper).

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 excited to be at AACR (Free AACR Whitepaper) unveiling new preclinical data, which includes a closer look at the promise of our novel non-viral TRAC-knocked-in T cell therapy for the treatment of p53 R175H-mutant solid tumors," said Loïc Vincent, Ph.D., Chief Scientific Officer, Affini-T Therapeutics. "We are also proud to present preclinical data from our cell therapy targeting KRAS G12D, which highlights the promising potential of our proprietary non-viral knock-in THRIVE platform. These findings continue to motivate our team as we advance programs leveraging TCR-engineered T cells as potential paradigm-shifting treatments for patients with solid tumor cancers."

Poster presentation details are as follows:

AFNT-212: A TRAC-knocked-in KRAS G12D-specific TCR-T cell product enhanced with CD8αβ and a chimeric cytokine receptor for treatment of solid cancers

Abstract #5973, Session: Adoptive Cell Therapies 1: Tumor Antigen-Specific T-cells and TCR-T
Session Date/Time: Sunday, April 7, 2024, 1:30 PM – 5:00 PM
Presenting Author: Loïc Vincent, Ph.D., Chief Scientific Officer, Affini-T Therapeutics
Summary: AFNT-212-engineered TCR-T cells showed high functional avidity and in vitro cytotoxicity against KRAS G12D-positive tumor cell lines, including CL40 (colon), PANC-1 and HPAF-II (pancreas), SK-LU-1 (lung), HuCCT1 (cholangiocarcinoma), etc. Additionally, engineered TCR-T cells demonstrated robust and durable anti-tumor activity in vivo and low risk of off-target/off-tumor toxicity. Affini-T’s proprietary non-viral knock-in (KI) THRIVE platform achieved high transgene integration efficiency and cell growth to yield relevant numbers of engineered TCR-T cells for clinical application. The study supports the planned clinical investigation of the novel KRAS G12D mutant TCR-engineered CD4+ and CD8+ T cell therapy in 2024.
Non-viral engineered T cell therapy specific for the hotspot mutation p53 R175H that integrates signal 1 (TCR), signal 2 (co-stimulation) and signal 3 (cytokine) and co-opts FasL-dependent apoptosis to achieve a coordinated antitumor CD4/8 T cell response

Abstract #7242, Session: Adoptive Cell Therapies 1: Tumor Antigen-Specific T-cells and TCR-T
Session Date/Time: Sunday, April 7, 2024, 1:30 PM – 5:00 PM
Presenting Author: Gary Shapiro, Ph.D., VP Biology Discovery, Affini-T Therapeutics
Summary: The tumor suppressor TP53 is the most frequently mutated gene across human cancers, with a highly recurrent arginine to histidine hotspot alteration in codon 175 leading to novel tumor-dependent functions. In this study, we reported the use of a novel CRISPR-Cas nuclease system to knock in a six-gene multi-cistronic cassette into the TRAC locus with high efficiency. We employed several strategies to maximize the potency and durability of a TCR-T cell product targeting the p53 R175H oncogenic driver, TCR, co-stimulation and cytokine signaling – which delivered full stimulation of both CD8+ and CD4+ T cells. These data support the planned clinical development of a novel non-viral TRAC-knocked-in T cell therapy for the treatment of p53 R175H-mutant solid tumors.