Nature Paper Identifies “Avidity Escape” as Solid Tumor Evasion Mechanism of CAR T Cell Cancer Therapies

On May 2, 2022 LUMICKS, a leading life science tools company developing instruments and consumables for dynamic single-molecule and cell avidity analysis, reported the publication in Nature of a landmark study, led by researchers at Massachusetts General Hospital, Harvard Medical School and the Broad Institute of MIT and Harvard (Press release, LUMICKS, MAY 2, 2022, View Source;utm_medium=rss&utm_campaign=nature-paper-study-marcela-maus-solid-tumor-evasion [SID1234613282]).

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 demonstrates the power of LUMICKS’ z-Movi Cell Avidity Analyzer to measure cell avidity, a crucial biomarker for understanding immune cell function and for providing superior predictive information of therapeutic efficacy. The Nature paper (April 16, 2022), entitled "CAR T cell killing requires the IFNγR pathway in solid but not liquid tumors" was published by a pioneering team of chimeric antigen receptor (CAR) T cell researchers led by Dr. Marcela V. Maus, Associate Professor of Medicine at Harvard Medical School and Director of the Cellular Immunotherapy Program at Massachusetts General Hospital.

While CAR T cell therapies have shown great potential in combating blood-related cancers, the more prevalent solid tumors have been far less responsive to this therapeutic approach. This study exposes a groundbreaking discovery about the mechanism solid tumors use to evade CAR T cell therapies. Solid tumor cells avoid CAR T cell killing by down-regulating the adhesion strength or "cell avidity" between the effector and its target, which is an essential step in successfully clearing a solid tumor. Measuring cell avidity should now be an integral part of CAR T cell development programs, to help reduce failure rates of solid tumor clinical trials.

The LUMICKS z-Movi Cell Avidity Analyzer showed that loss of interferon-γ receptor (IFNγR) signaling decreased cell binding avidity between CAR T cells and their target solid tumor cells. The insufficient cell avidity was determined to be the mechanism underpinning poor in vivo therapeutic behavior of CAR T cells, identifying "avidity escape" as a new avoidance mechanism used by solid tumors.

"We are excited about the publication of this seminal paper in Nature that clearly demonstrates the technological and scientific power of measuring cell avidity with the z-Movi Cell Avidity Analyzer, opening the door to new innovations in cell therapy development," said Andrea Candelli, Ph.D., Chief Scientific Officer of LUMICKS. "We are at a watershed moment in the CAR T cell field, and we are excited to empower therapeutic developers with novel insights for successfully harnessing the power of the immune system to treat diseases."

The z-Movi Cell Avidity Analyzer measures cell avidity, or level of binding, between immune cells and their targets, enabling researchers to identify the most potent immunotherapeutic effector cells. This unique technology provides predictive, reproducible, and fast results at single-cell resolution. LUMICKS’ cell avidity solutions use acoustics to measure forces and interactions between cells, with the goal of such research enabling the shortening the drug development cycle of immunotherapies and reducing failure rates in clinical trials. First introduced in 2020, the z-Movi Cell Avidity Analyzer is being rapidly adopted by academic and biopharma laboratories around the world.

About the Study Findings

This study found that, as opposed to blood-based cancers, the loss of genes in the interferon-γ receptor (IFNγR) signaling pathway rendered glioblastoma and other solid tumors more resistant to killing by CAR T cells both in vitro and in vivo. Mechanistically, the loss of IFNγR1 in glioblastoma cells reduced overall CAR T duration of cell binding and cell avidity, as measured using LUMICKS z-Movi Cell Avidity Analyzer. IFNγR signaling was found to regulate critical surface proteins responsible for sufficient adhesion of CAR T cells to mediate therapeutic response through productive cytotoxicity.

This work demonstrates that blood-based tumors and solid tumors differ in their interaction strength with CAR T cells. The study suggests that enhancing binding interactions between T cells and tumor cells may yield improved responses for next generation drugs, especially through new molecular designs that optimize cell avidity strength. This highlights the power of cell avidity measurements as a crucial biomarker for understanding immune cell function and mechanistic guide for successful CAR T engineering.

Luxeptinib Preclinical Data Extend Potential Applications from Oncology to Inflammation

On May 2, 2022 Aptose Biosciences Inc. ("Aptose" or the "Company") (NASDAQ: APTO, TSX: APS), a clinical-stage precision oncology company developing highly differentiated oral kinase inhibitors to treat hematologic malignancies, reported recent publications of preclinical data for luxeptinib (CG-806) in three peer-reviewed scientific journals (Press release, Aptose Biosciences, MAY 2, 2022, View Source [SID1234613298]). Luxeptinib, Aptose’s oral, dual lymphoid and myeloid kinome inhibitor, is an investigational drug currently in two Phase 1 a/b trials: one in patients with relapsed or refractory B cell malignancies, and separately in patients with relapsed or refractory acute myeloid leukemias (AML) or high-risk myelodysplastic syndromes (MDS) .

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!

Recent Journal Articles:

Luxeptinib disables NLRP3 inflammasome-mediated IL-1β release and pathways required for secretion of inflammatory cytokines IL-6 and TNFα Biochemical Pharmacology (2022)195 114861 (link)

Luxeptinib is an orally bioavailable kinase inhibitor with potency against select kinases including BTK. Aberrant activation of inflammasomes act as drivers of pathological complications observed during autoimmune and inflammatory disorders, metabolic syndromes, and cancer; and inhibiting the inflammasome-induced activation of pro-inflammatory cytokines has shown beneficial effects in human disease models. BTK and certain other kinases serve as an integral components or influence functions of the NLRP3 inflammasome complex. The aim of this study was to determine if luxeptinib interferes with the release of IL-1β, IL-6 and TNFα from THP-1 monocytes and bone marrow-derived macrophages following endotoxin exposure and priming of the NLRP3 inflammasome.

Key findings:

Luxeptinib inhibits NLRP3 inflammasome function in THP-1 monocytes and bone marrow-derived macrophages.
Mechanistically, luxeptinib prevents the release of cleaved IL-1β from THP-1 cells by inhibiting the ability of the NLRP3 inflammasome to proteolytically cleave caspase-1 to its active form.
Luxeptinib does not impede the assembly of the NLRP3-ASC complex but does potently inhibit three kinases phosphorylated in response to endotoxin. Phosphorylation and nuclear translocation of transcription factor NF-κBp65 was inhibited by luxeptinib.
Luxeptinib also inhibits the release of TNFα and IL-6 in response to activation of the TLR pathway without affecting the TLR/IRAK/MyD88 proteins.
These studies provide novel insights into the mechanisms by which luxeptinib interferes with the NLRP3 inflammasome and endotoxin-induced inflammatory signaling pathways, along with its protective effects against inflammation-induced toxicity in murine models.
The ability of luxeptinib to inhibit inflammatory pathways at concentrations which are well-tolerated in patients makes it a potential clinical candidate for the treatment of inflammatory diseases and inflammation-associated resistance in cancer.
Dual BTK/SYK inhibition with CG-806 (luxeptinib) disrupts B-cell receptor and Bcl-2 signaling networks in mantle cell lymphoma Cell Death & Disease – Nature (2022)13:246 (link)

Small molecules BTK inhibitors like ibrutinib are approved for the treatment of mantle cell lymphoma, or MCL, a rare subtype of non-Hodgkin’s lymphoma (NHL). Nevertheless, median duration of response is less than two years, and MCL patients who develop therapeutic resistance have poor outcomes. Resistance to BTK inhibitors is not clearly understood and a number of alternative mechanisms have been implicated. Luxeptinib, previously known as CG-806, inhibits LYN, SYK, and BTK activation, potently inhibiting both wildtype and C481S mutant BTK, and is expected to have activity in settings where resistance to BTK inhibitors is driven by these mutations. In a Phase 1 trial in patient with chronic lymphocytic leukemia (CLL) and NHL, treatment with luxeptinib resulted in decreased phosphorylation of SYK and BTK in the circulating malignant cells within eight hours of administration. This current pre-clinical study investigates mechanism and efficacy of luxeptinib in MCL.

Key findings:

Luxeptinib induced apoptosis in ibrutinib-resistant MCL cell lines, and in primary MCL cells luxeptinib downmodulated anti-apoptotic proteins Mcl-1 and Bcl-xL and reversed stromal cell survival effects.
Luxeptinib, but not ibrutinib, blocked SYK and BTK signaling and inhibited ERK phosphorylation in MCL cell lines and primary MCL cells.
Dual suppression of BTK/SYK activation with luxeptinib demonstrated efficacy in a PDX MCL model, where efficacy was accompanied by downmodulation of Bcl-2 family proteins and NFκB.
Luxeptinib induced metabolic reprogramming toward a glycolytic shift in MCL cells, accompanied by mitochondrial depolarization and induction of mitophagy that eliminates dysfunctional mitochondria and leads to cell death.
Luxeptinib Inhibits BTK/SYK/ERK signaling and is a potentially promising new therapy in MCL and NHL.
Luxeptinib (CG-806) targets FLT3 and clusters of kinases operative in acute myeloid leukemia Molecular Cancer Therapeutics (2022), Mol. Cancer Ther. molcanther.0832.2021 (link)

AML cells survive via dysregulation of multiple pathways, including FLT3 mutations that occur in approximately 30% of AML patients and are associated with increased risk of relapse and poor survival. Luxeptinib, currently in a Phase 1a/b clinical trial for the treatment of AML, potently inhibits both FLT3 and many of the kinases that participate in rescue pathways that contribute to relapsed and refractory disease. In this study, researchers investigated the range of kinases it inhibits, its antiproliferative landscape ex vivo with AML patient samples, and its in vivo efficacy in xenograft models.

Key findings:

Luxeptinib, an oral non-covalent kinase inhibitor, potently suppresses wild type and mutant forms of FLT3 and select clusters of kinases that can participate in rescue pathways.
Oral luxeptinib demonstrated strong antitumor activity in preclinical in vivo AML xenograft models but no myelosuppression or evidence of tissue damage in acute toxicology studies.
Importantly, luxeptinib does not inhibit the TEC, EGFR, or ERBB2 kinases that can be associated with cardiac, skin and bleeding adverse events.
Ex vivo profiling of luxeptinib against 186 AML fresh patient samples demonstrated greater potency relative to other FLT3 inhibitors, including cases with mutations in FLT3, IDH1/2, ASXL1, NPM1, SRSF2, TP53 or RAS.
A combination of venetoclax and luxeptinib enhanced cell killing of the majority of AML samples relative to either drug alone.
Luxeptinib retained activity against FLT3 mutants that render cells resistant to quizartinib, gilteritinib, and crenolanib FLT3 inhibitors.
"These publications contribute to the wealth of preclinical data demonstrating luxeptinib’ s unique activity as a lymphoid and myeloid kinome inhibitor, and now as an inflammation kinome inhibitor, and support its continued clinical development in several therapeutic areas," said William G. Rice, Ph.D., Chairman, President, and Chief Executive Officer. "Luxeptinib is a clinical-stage compound, currently in Phase 1 a/b studies in AML and B-cell malignancies. We look forward to reporting on our progress in the upcoming months."

Deciphera Pharmaceuticals to Announce First Quarter 2022 Financial Results and Host Conference Call and Webcast on May 4, 2022

On May 2, 2022 Deciphera Pharmaceuticals, Inc. (NASDAQ: DCPH) reported that it will report its first quarter 2022 financial results on Wednesday, May 4, 2022 (Press release, Deciphera Pharmaceuticals, MAY 2, 2022, View Source [SID1234613314]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

In connection with the earnings release, Deciphera’s management team will host a live conference call and webcast at 8:00 AM ET on Wednesday, May 4, 2022, to discuss the Company’s financial results and provide a corporate update.

The conference call may be accessed by dialing (877) 270-2148 (domestic) or (412) 902-6510 (international). A webcast of the conference call will be available in the "Events and Presentations" page in the "Investors" section of the Company’s website at View Source The archived webcast will be available on the Company’s website approximately two hours after the conference call and will be available for 30 days following the call.

Agendia to Deliver Robust Suite of Data Advancing Breast Cancer Care at ASCO 2022

On May 2, 2022 Agendia, Inc., a commercial stage company focused on enabling optimized decision-making by providing physicians with next-generation diagnostic and information solutions that can be used to help improve outcomes for breast cancer patients worldwide, reported it will present new data from ongoing clinical research evaluating its comprehensive genomic tests at the upcoming American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting (ASCO) (Free ASCO Whitepaper), taking place June 3-7, 2022 in Chicago, Illinois (Press release, Agendia, MAY 2, 2022, View Source [SID1234613330]).

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!

Data will highlight Agendia’s clinical focus on whole transcriptome analysis as well as breast cancer care for underserved populations, in addition to several sub-studies derived from the company’s FLEX Registry, the real-world, multicenter, prospective, observational breast cancer study. Two abstracts selected by ASCO (Free ASCO Whitepaper) for oral discussion will feature an investigation of the ImPrint genomic test, currently for research use only, from the I-SPY trial series and an analysis from FLEX of hormone receptor-positive breast cancer in Black women classified by BluePrint.

The company believes the FLEX Registry’s approach to cancer research is accelerating impactful data generation, aimed at redefining cancer care. Its patient-centric design and national network of participating sites is backed by Agendia’s infrastructure, allowing its investigator-initiated sub-studies to produce important results that have the potential to drive science forward, like those being shared at ASCO (Free ASCO Whitepaper) 2022.

Following are details of the nine Agendia abstracts that have been accepted for poster discussion or poster sessions at the ASCO (Free ASCO Whitepaper) 2022 Annual Meeting:

Poster discussion sessions

The ImPrint immune signature to identify patients with high-risk early breast cancer who may benefit from PD1 checkpoint inhibition in I-SPY2
Authors: Kuilman, MM., et al.
Presenter: Lorenza Mittempergher, PhD | Research and Development, Agendia NV
Session: Breast Cancer – Local/Regional/Adjuvant
Poster Discussion: Monday, June 6, 2022 | 1:15 PM-2:45 PM CDT
Abstract #: 514
Whole transcriptomic analysis of HR+ breast cancer in Black women classified as basal-type by BluePrint
Authors: Reid, S., et al.
Presenter: Sonya A. Reid, MD, MPH | Vanderbilt University Medical Center
Session: Breast Cancer – Local/Regional/Adjuvant
Poster Discussion: Monday, June 6, 2022 | 1:15 PM-2:45 PM CDT
Abstract #: 517
Poster sessions

Whole transcriptome analysis of tumors with discordant oncotype and MammaPrint results in the FLEX trial
Authors: Socoteanu, M., et al.
Session: Breast Cancer – Local/Regional/Adjuvant
Session Date & Time: Monday, June 6, 2022 | 8:00 AM-11:00 AM CDT
Abstract #: 556
Clinical implications for patients with discordant oncotype and MammaPrint results
Authors: Socoteanu, M., et al.
Session: Breast Cancer – Local/Regional/Adjuvant
Session Date & Time: Monday, June 6, 2022 | 8:00 AM-11:00 AM CDT
Abstract #: 560
Investigation of a genomic signature for transcription factor MAF gene amplification and lack of bisphosphonate benefit in early breast cancer
Authors: Nasrazadani, A., et al.
Session: Breast Cancer – Local/Regional/Adjuvant
Session Date & Time: Monday, June 6, 2022 | 8:00 AM-11:00 AM CDT
Abstract #: 559
Identification of transcriptional changes with MammaPrint and BluePrint in early-stage breast cancer after neoadjuvant chemotherapy
Authors: Chung, A., et al.
Session: Breast Cancer – Local/Regional/Adjuvant
Session Date & Time: Monday, June 6, 2022 | 8:00 AM-11:00 AM CDT
Abstract #: 585
Distribution of breast cancer molecular subtypes within receptor classifications: Lessons from the I-SPY2 trial and FLEX Registry
Authors: Cha, J., et al.
Session: Breast Cancer – Local/Regional/Adjuvant
Session Date & Time: Monday, June 6, 2022 | 8:00 AM-11:00 AM CDT
Abstract #: 592
FLEX, the 30,000 breast cancer transcriptome project: A platform for early breast cancer research using full-genome arrays paired with clinical data
Authors: Ma, C., et al.
Session: TBC
Session Date & Time: Monday, June 6, 2022 | 8:00 AM-11:00 AM CDT
Abstract #: TPS612
Defining transcriptomic profiles of early-stage mucinous breast cancers: A FLEX sub study
Authors: Sivapiragasam, A., et al.
Session: Developmental Therapeutics – Molecularly Targeted Agents and Tumor Biology
Session Date & Time: Sunday, June 5, 2022 | 8:00 AM-11:00 AM CDT
Abstract #: 3134
Agendia will be sharing important updates on its Twitter, Facebook and LinkedIn pages throughout the conference. The event program can be found at the ASCO (Free ASCO Whitepaper) 2022 website.

Poseida Therapeutics to Present at American Society of Gene and Cell Therapy 25th Annual Meeting

On May 2, 2022 Poseida Therapeutics, Inc. (Nasdaq: PSTX), a clinical-stage biopharmaceutical company utilizing proprietary genetic engineering platform technologies to create cell and gene therapeutics with the capacity to cure, reported that preclinical data highlighting the use of anti-c-kit CAR-T cells, P-ckit-ALLO1 as a preconditioning agent to enable hematopoietic stem cell (HSC) transplants, will be presented at the American Society of Gene and Cell Therapy (ASGCT) (Free ASGCT Whitepaper) 25th Annual Meeting, being held in Washington, D.C. and virtually on May 16-19, 2022 (Press release, Poseida Therapeutics, MAY 2, 2022, View Source [SID1234613412]).

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 Company’s anti-c-kit CAR-T program leverages its proprietary piggyBac Gene Delivery System and Cas-CLOVER Site-specific Gene Editing System to develop fully allogeneic CAR-T cells targeting human c-kit which is highly expressed on HSCs, as well as on myeloid malignancies such as acute myeloid leukemia (AML), meaning the treatment can be used for either HSC transplant conditioning or as a treatment for AML. In addition to the CAR gene, the piggyBac transposon includes a selection marker for generation of a pure CAR+ product and a proprietary fast-acting safety switch enabling rapid clearance of the reactive CAR-T cells prior to donor HSC transplant.

Presentation details:

Poster Presentation: Anti-c-kit CAR-T Cells Enable HSC Engraftment in a Humanized Model of Stem Cell Transplant Conditioning
Session Title: Cell Therapies II
Session Date/Time: Tuesday, May 17, 2022, 5:30 – 6:30 PM ET
Poster Board Number: Tu-239
Location: Walter E. Washington Convention Center, Hall D
Abstract Number: 734