Marengo Therapeutics Presents Preclinical Data Supporting Clinical Investigation of its Lead Asset, STAR0602, at the 2022 Society for Immunotherapy of Cancer (SITC) Annual Meeting

On November 10, 2022 Marengo Therapeutics, Inc., a company pioneering novel therapeutics targeting the T cell receptor Vβ chain (TCR Vβ) to selectively activate the right T cell subsets to fight cancer, reported that it will be presenting preclinical data that characterizes the mechanism of action and confirms robust anti-tumor activity for its novel T cell-activating antibody STAR0602 at the Society for Immunotherapy of Cancer (SITC) (Free SITC Whitepaper) 37th Annual Meeting in Boston (Press release, Marengo Therapeutics, NOV 10, 2022, View Source [SID1234623867]). The data disclosed also includes extensive characterization pharmacokinetic (PK) and pharmacodynamic (PD) relationships of STAR0602 in NHP studies. These data support the modelling of human pharmacology and inform the design of START-001, a Phase 1/2 clinical trial of STAR0602 monotherapy in a biomarker enriched, tissue agnostic, PD-1 refractory cancer patient population.

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!

"STAR0602 has been shown to promote potent anti-tumor activity through a PD-1 independent mechanism and leads a new IO category of selective immune activators, potentially offering another important treatment option to reach more patients," said Zhen Su, M.D., MBA, Chief Executive Officer of Marengo. "We are pleased to present a compelling body of preclinical data that builds on the initial proof-of-concept data shared during our Plenary Oral presentation at the recent 34th EORTC-NCI-AACR (Free EORTC-NCI-AACR Whitepaper) (ENA) Symposium last month."

The data presented at SITC (Free SITC Whitepaper) by Marengo scientists and its academic collaborators highlight Marengo’s novel TCR Vβ repertoire-targeting antibody platform, which promotes the expansion of polyclonal Vβ T cells with a novel effector memory phenotype.

Presentation details are outlined below.

Title: A novel class of T cell-activating antibody that selectively targets the TCR β chain to promote antitumor activity through activation and expansion of a novel, polyclonal effector memory T cell subset
Abstract Number: 1316
Presenter: Andrew Bayliffe, Ph.D. (Marengo Therapeutics, Cambridge, Massachusetts USA)
Research Highlights: STAR0602 is a first-in-class bifunctional fusion molecule that selectively binds and activates subsets of the germline TCR repertoire. In vitro, STAR0602 promotes a novel T cell phenotype with hallmarks of both effector and central memory cells, and in vivo mSTAR0602 demonstrates potent and durable single-agent anti-tumor activity in several solid tumor models that is dependent on expanded Vβ T cells. The modulation of the tumor microenvironment (TME), striking increase in TCR diversity, and functional immune memory observed in murine models suggests that STAR0602 could remodel the adaptive immune response to solid tumors that are refractory to checkpoint inhibitor therapy, and thus represents a novel therapeutic strategy for patients.

Title: Preclinical evaluation of STAR0602, a novel, first-in-class anti-TCR Vβ targeted bispecific antibody with potent anti-tumor activity for PD-1 refractory solid tumors
Abstract Number: 1337
Presenter: James Gulley, M.D., Ph.D. (National Cancer Institute, Bethesda, Maryland USA)
Research Highlights: STAR0602 is a first-in-class T cell activator that targets subsets of the germline TCR repertoire that are enriched in TILs. STAR0602 potently expands both naive and antigen-specific human T cells. In PD1 refractory human organoid models with a high TMB, STAR0602 induced potent anti-tumor activity as monotherapy, mediated by selective expansion of Vβ CD8+ memory T cells. This pharmacology was translated into monkeys with IV dosed STAR0602 and supports the design of a novel Phase 1/2 precision-oncology trial with STAR0602 planned to commence in 2022.

Title: An atypical central-memory like phenotype can be induced in human T cells by Innate TCRαβ engagement
Abstract Number: 1392
Presenter: Pierre Vantourout, Ph.D. (Kings College London, London, UK)
Research Highlights: Engaging germline-encoded regions of human TCRVβ consistently activate primary human T cells toward an atypical central memory (TCM)-like phenotype distinct from those most commonly described for anti-CD3 antibody stimulation. The cells show myriad surface markers of chronic stimulation, but are not exhausted, being highly proliferative and strongly expressing cytolytic mediators and IFN-γ. This phenotype can be induced in cells previously driven toward effector memory (TEM) and TEMRA states. The use of TCRβ chain as an innate receptor offers new insight into T cell biology and ways in which such cells might be clinically manipulated.

HotSpot Therapeutics Presents Preclinical Data from CBL-B Program at 2022 Society for Immunotherapy of Cancer Annual Meeting

On November 10, 2022 HotSpot Therapeutics, Inc., a biotechnology company pioneering the discovery and development of orally delivered, small molecule allosteric therapies for the treatment of cancer and autoimmune diseases, reported the presentation of additional preclinical data on the Company’s Casitas B-lineage lymphoma proto-oncogene (CBL-B) program in two poster presentations at the 2022 Society for Immunotherapy of Cancer (SITC) (Free SITC Whitepaper) Annual Meeting (Press release, HotSpot Therapeutics, NOV 10, 2022, View Source [SID1234623866]).Due to its role as a gatekeeper in immune cell activation, CBL-B inhibition holds the potential to address several key mechanisms important in immuno-modulation. Translational data supports a role for CBL-B inhibition to address suboptimal response to current immunotherapies in certain cancers. Targeting CBL-B represents a novel therapeutic approach because inhibition of CBL-B has been shown to lower the threshold for T cell and NK cell activation, even in the absence of co-stimulatory signals, potentially bringing benefit to patients with suboptimal conditions in the tumor microenvironment.

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 HotSpot, we are generating preclinical data elucidating the central role of CBL-B inhibition to immune cell activation, providing strong biological rationale for this mechanism’s potential as a promising immuno-oncology treatment option," said Geraldine Harriman, Ph.D., Co-Founder and Chief Scientific Officer of HotSpot Therapeutics. "Our data in syngeneic mouse models underscore the broad range of immunostimulatory activity of CBL-B inhibition which, coupled with compelling data from the mixed lymphocyte reaction (MLR) assay, a clinical correlate for I-O agents, as well as clear synergistic activity with anti-PD-1, support the continued advancement of our CBL-B inhibitor program. We look forward to continuing to work toward advancing HST-1011, our lead CBL-B inhibitor development candidate, into the clinic."

The presentations describe compelling data for HotSpot compounds designed as novel, allosteric, small molecule inhibitors of CBL-B E3 ubiquitin ligase activity:

A HotSpot CBL-B inhibitor demonstrated immune-mediated tumor growth inhibition in multiple syngeneic mouse models. Gene expression profiling of the tumor microenvironment demonstrated upregulation of pro-inflammatory pathways in vivo. These effects were notable with CBL-B inhibition alone and further enhanced when combined with a PD-1 inhibitor.
In the MLR assay, a predictive correlate of the clinical activity of I-O therapies, the HotSpot CBL-B inhibitor demonstrated robust effects on cytokine release and T cell proliferation as monotherapy. Additionally, CBL-B inhibition demonstrated synergistic activity in the MLR assay when combined with anti-PD1.

Nektar Therapeutics Presents Preclinical Data from Novel PEGylated Interferon Gamma Program, NKTR-288, at the Society for Immunotherapy of Cancer (SITC) Annual Meeting

On November 10, 2022 Nektar Therapeutics (Nasdaq: NKTR) reported the first presentation of preclinical data for NKTR-288 at the 2022 Society for Immunotherapy of Cancer (SITC) (Free SITC Whitepaper) Annual Meeting (Press release, Nektar Therapeutics, NOV 10, 2022, View Source [SID1234623865]).

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!

NKTR-288 is a novel polyethylene glycol (PEG)-conjugate of interferon gamma (IFN-γ), designed to modify binding of IFN-γ to its substrates and optimize the duration of IFN-γ signaling. IFN-γ is a cytokine that induces cellular antigen presentation and enhances tumor antigen-specific cytotoxic T cell response, and may have applications in a number of therapeutic areas, including oncology and infectious diseases, among others.

The presentation today showed the sustained pharmacological properties of NKTR-288 relative to native cytokine, including durable exposure and induction of the well characterized interferon gamma response. These data support further evaluation of NKTR-288 as a monotherapy or in combination with checkpoint inhibitors in the clinical setting.

"The results presented at SITC (Free SITC Whitepaper) clearly demonstrate that NKTR-288 has differentiated pharmacokinetic and pharmacodynamic properties compared to native IFN-γ, leading to durable in vivo anti-tumor activity in preclinical models," said Jonathan Zalevsky, Ph.D., Senior Vice President and Chief Research & Development Officer at Nektar. "Particularly promising were data that show NKTR-288 upregulates PD-L1 expression in tumor tissue, supporting potential combination therapy with checkpoint inhibitors by enhancing checkpoint blockade efficacy in low-MHC Class I ‘cold’ tumor models. It highlights the potential of NKTR-288 to augment currently available PD-1 or PD-L1 treatment strategies and possibly broaden the responsive patient population."

Also presented at SITC (Free SITC Whitepaper) by Nektar’s partner, Merck KGaA, is a trials in progress poster of the Phase 2 JAVELIN Bladder Medley study (NCT05327530). The ongoing clinical trial includes evaluation of BAVENCIO (avelumab), Merck’s anti-PD-L1 therapy, in combination with NKTR-255, Nektar’s IL-15 receptor agonist designed to increase the proliferation and survival of natural killer (NK) cells and memory CD8+ T cells to enhance anti-tumor immunity. Merck KGaA is running this study and is on track to generate comparative progression-free survival (PFS) data for this study by the end of 2024.

Key details and takeaways from the presentation are as follows:

Abstract 1086: "NKTR-288, a PEGylated Interferon Gamma Drug Candidate for the Treatment of Cancer", Hamel, D., et al.

NKTR-288 demonstrated reduced affinity to both IFN-γ receptor and heparin, decreasing target mediated drug disposition (TMDD) and increasing bioavailability compared to native IFN-γ.
Induced sustained upregulation of major histocompatibility complex class I (MHC Class I) expression in fast-growing, low-MHC Class I "cold" syngeneic tumor models, as well as T cell-mediated anti-tumor activity, alone and in combination with checkpoint inhibitors.
NKTR-288 also upregulated PD-L1 expression in tumor tissue as a monotherapy, while significant anti-tumor activity was observed when combined with anti-PD-1 or anti-PD-L1 antibodies.
Well tolerated at pharmacologically active doses in preclinical models well below maximum tolerated dose (1 mg/kg).
These results support the development of NKTR-288, a novel, optimized IFN-γ pathway agonist, to treat cancer as a monotherapy or in combination with PD-1/PD-L1 therapy.
Details of the Trials in Progress poster presentation are as follows:

Abstract 665: "JAVELIN Bladder Medley: a phase 2 trial of avelumab in combination with other antitumor drugs as first-line maintenance therapy for advanced urothelial carcinoma", Hoffman-Censits, J., et al.

The objective of the phase 2, multicenter, randomized, open-label parallel-arm JAVELIN Bladder Medley umbrella trial (NCT05327530) is to assess the safety and efficacy of avelumab in combination with other antitumor agents, including NKTR-255, as first-line (1L) maintenance treatment for patients with advanced urothelial carcinoma (UC).
When completed, the ongoing JAVELIN Bladder Medley study will show whether 1L maintenance treatment with avelumab-based combinations can improve PFS vs avelumab alone and report the safety and tolerability of the combination regimens.
Details of the data presentations at SITC (Free SITC Whitepaper) are available on the Science section of Nektar’s website at View Source

About NKTR-288

NKTR-288 is a PEG-conjugate of the protein, IFN-γ, a cytokine that induces cellular antigen presentation and has great potential in cancer treatment through enhancement of tumor antigen specific cytotoxic T cell response. NKTR-288 was designed utilizing a site-specific conjugation approach to modify binding of IFN-γ with one of its substrates, and overall, to optimize the duration of IFN-γ signaling. This program has applications in a number of therapeutic indications including oncology as well as in infectious disease and others.

About NKTR-255

NKTR-255 is a biologic that targets the IL-15 pathway in order to activate the body’s innate and adaptive immunity. Through optimal engagement of the IL-15 receptor complex, NKTR-255 is designed to enhance functional NK cell populations and formation of long-term immunological memory, which may lead to sustained and durable anti-tumor immune response.

Preclinical findings suggest NKTR-255 has the potential to synergistically combine with antibody-dependent cellular cytotoxicity molecules as well as to enhance CAR-T therapies. Nektar has initiated a Phase 1 dose escalation and expansion clinical study of NKTR-255 in adults with relapsed or refractory non-Hodgkin lymphoma or multiple myeloma, as well as a Phase 1/2 clinical study of NKTR-255 in patients with relapsed or refractory head and neck squamous cell carcinoma or colorectal cancer. Nektar is also continuing its oncology clinical collaboration with Merck KGaA and Pfizer Inc. to evaluate the maintenance regimen of NKTR-255 in combination with avelumab, a PD-L1 inhibitor, in patients with locally advanced or metastatic urothelial carcinoma in the Phase 2 JAVELIN Bladder Medley study. Nektar is also currently designing a Nektar-sponsored Phase 2/3 study combining NKTR-255 with approved CAR-T cell therapies in diffuse large B-cell lymphoma, which it aims to initiate in the first quarter of 2023.

Corbus Presents Latest Preclinical Data for CRB-601 at the Society for Immunotherapy of Cancer (SITC) 37th Annual Meeting

On November 10, 2022 Corbus Pharmaceuticals Holdings, Inc. (NASDAQ: CRBP) ("Corbus" or the "Company"), an oncology company, reported that it is presenting new preclinical data for CRB-601 in a poster presentation at the Society for Immunotherapy of Cancer (SITC) (Free SITC Whitepaper) Annual Meeting in Boston (Press release, Corbus Pharmaceuticals, NOV 10, 2022, View Source [SID1234623864]).

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 latest preclinical data demonstrate significant tumor growth inhibition by CRB-601 as a single agent and in combination with anti-PD-1 treatment in the syngeneic murine tumor models MC38 and EMT6. In addition, treatment with CRB-601 appears to restore sensitivity to checkpoint inhibitors in the Pan02 and 4T1 syngeneic tumor models, regarded as "desert" tumors that are non-responsive to current CPIs. CRB-601 is the only anti-αvβ8 targeting agent known to demonstrate such an effect in these models to date.

The anti-tumor effects in the immune excluded EMT6 model correlated with changes in the immune cell populations in the tumor microenvironment. There were increases in proliferating CD4+ and CD8+ T-cells and Natural Killer cells, and a shift in macrophage polarization to the inflammatory M1 phenotype. Collectively these data suggest that treatment with CRB-601 could address tumors with an immune excluded phenotype, as represented by this model, and facilitate sensitization to anti-PD-1 therapy.

Additionally, tumor protection is associated with a durable T-cell memory response. Tumor-bearing mice initially rendered tumor-free by treatment with the combination of CRB-601 + anti-PD-1 were subsequently reinoculated with the same tumor cells (MC38) and monitored for tumor establishment and growth. After 30 days no tumors had formed in this cohort of animals, whereas implantation and eventual death occurred in 100% of the treatment naïve control animals. T-cell depletion studies indicated that protection against MC38 tumor growth in these rechallenged animals was dependent upon CD8+ T-cells.

"CRB-601 demonstrates robust pre-clinical anti-tumor activity alone and in combination with anti-PD-1 therapy in tumors exhibiting a range of sensitivities to anti-PD-1 therapy. The ability of CRB-601 to significantly reduce the growth of tumors that are non-responsive to CPIs and to sensitize these tumors to CPI therapy continues to support our hypothesis that effective blockade of latent TGFβ activation should lead to enhanced immune cell invasion in the tumor microenvironment and augment the effects of anti-PD-1 therapy. The increased activity of CRB-601 in tumors expressing αvβ8 provides a promising biomarker for patient selection and stratification," commented Rachael Brake, Ph.D., Chief Scientific Officer of Corbus.

Corbus is currently developing CRB-601 as a potential treatment for solid tumor cancers, and the program is on schedule for an IND submission in mid-2023.

Dr. Daqing Wang, PhD will be presenting the new data today in Poster #815 in Hall C from 11:40 am – 1:10 pm and again from 7:30 pm – 9 pm. The SITC (Free SITC Whitepaper) poster is available on the company website at: View Source

Regen BioPharma Developing a Non-Exhaustible CAR-T Cell Therapeutic

On November 10, 2022 Regen BioPharma, Inc. (OTC PINK: RGBP) and (OTC PINK: RGBPP) reported that it has recently discussed the use of CAR-T therapies in cancer treatment (View Source;explores-the-car-t-therapeutic-space-301660078.html) and its development of a novel approach to creating CAR-T cells that can potentially be used to treat solid tumors (View Source;develops-novel-dedifferentiation-approach-for-increasing-efficacy-of-car-t-cells-to-treat-solid-tumors-301604599.html).

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 tumor microenvironment contains molecules and cells which can suppress T cells and prevent them from attacking and killing the tumor. "T cell exhaustion" is a term that describes T cells that are initially recruited to a tumor to kill it but end up rapidly losing their effectiveness. Eventually the tumor is surrounded by T cells that are exhausted and ineffective.

The Company has focused on NR2F6, an immune checkpoint which has been convincingly shown to be a very important transcription factor that keeps T cells in check1-4. When this transcription factor is blocked, T cells become very active and can kill tumors much more effectively. The Company has coupled its proprietary shRNA technology, which inhibits NR2F6, with existing CAR-T cell technology to create a potentially durable CAR-T cell which cannot be exhausted by the solid tumor microenvironment. The Company calls this novel CAR-T cell DuraCAR.

"We believe DuraCAR has the characteristics to overcome several of the major hurdles in getting CAR-T cells to be effective in treating solid tumors," says Dr. David Koos Chairman and CEO of the company. "We will know more in the coming months from our experimental work how long the road will be to get our technology fine-tuned to the point where we have a proof-of-concept drug. It is very exciting to see our shRNA technology applied in a way that can potentially help so many people with cancer."

1Klepsch et al. Targeting the orphan nuclear receptor NR2F6 in T cells primes tumors for immune checkpoint therapy Cell Communication and Signaling (2020) 18:8. View Source

2Klepsch, V., Hermann-Kleiter, N., Do-Dinh, P. et al. Nuclear receptor NR2F6 inhibition potentiates responses to PD-L1/PD-1 cancer immune checkpoint blockade. Nature Communications (2018) 9, 1538. View Source

3Klepsch et. al. Beyond CTLA-4 and PD-1: Orphan nuclear receptor NR2F6 as T cell signaling switch and emerging target in cancer immunotherapy. Immunology Letters (2016) 178, 31-36. View Source

4Hermann-Kleiter, et. al. The Nuclear Orphan Receptor NR2F6 Is a Central Checkpoint for Cancer Immune Surveillance. Cell Reports (2015) 12:12, 2072-2085. View Source