Synthekine Presents Data at American Society of Hematology Annual Meeting Showing Orthogonal IL-2 Ligand Drives Deeper and More Durable Response of CD-19 CAR-T

On December 7, 2020 Synthekine Inc., an engineered cytokine therapeutics company, reported in vivo data presented at the American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting showing STK-009, its orthogonal IL-2 ligand, improved the efficacy and durability of SYNCAR-001, its CD-19 targeted CAR-T cell therapy modified with an orthogonal IL-2 receptor (Press release, Synthekine, DEC 7, 2020, View Source [SID1234572367]). The data were delivered in a poster presentation by Paul-Joseph Aspuria, Ph.D., of Synthekine.

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!

"While CAR-T therapies have proven to be very effective in treating hematological malignancies, the lack of sustained activity and CAR-T cell persistence limits their full therapeutic potential in lymphoma," said Martin Oft, M.D., chief development officer at Synthekine. "The presented data demonstrate our orthogonal IL-2 system is well positioned to overcome these shortcomings. STK-009 demonstrates the ability to expand CAR-Ts in vivo in pre-clinical models, allowing for a lower starting dose of CARs while achieving deep and durable efficacy with the possibility to re-expand resting CARs long after initial transfer. We look forward to advancing STK-009 and SYNCAR-001 and filing an IND for the program in 2021."

In both disseminated and subcutaneous RAJI lymphoma models in mice evaluating the efficacy of STK-009 with SYNCAR-001, STK-009 dosing drove increased levels of circulating SYNCAR-001 cells, resulting in complete responses. STK-009 expanded both CAR-T effector cells and stem cell memory cells, resulting in deep initial responses and long-term immune surveillance. Data on STK-009 from a multiple-ascending dose study in non-human primates were also presented, showing STK-009 was well tolerated, had long exposure, and had no effect on cells expressing only the wild type IL-2 receptor. A copy of the poster is available on Synthekine’s website.

"Selectivity of this orthogonal IL-2 ligand is fundamental, as more promiscuous activity on bystander cells could lead to systemic toxicity and limit the therapeutic potential of this combined treatment," said Dr. Aspuria. "Based on the data we presented, we are optimistic about the potential for STK-009 and SYNCAR-001 to overcome hurdles faced by CD-19 cell therapy today."

Data Published in Nature Cancer Highlight Preclinical Proof of Concept of Bolt Biotherapeutics’ Boltbody™ ISAC Platform

On December 7, 2020 Bolt Biotherapeutics, Inc., a clinical-stage immuno-oncology company developing tumor-targeted therapies that leverage the power of the innate and adaptive immune systems, reported the publication of a manuscript in Nature Cancer highlighting the development and use of Boltbody Immune Stimulating Antibody Conjugates (ISACs) for the treatment of HER2-expressing tumors in preclinical models (Press release, Bolt Biotherapeutics, DEC 7, 2020, View Source [SID1234572384]).

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 paper, titled "Immune-stimulating antibody conjugates elicit robust myeloid activation and durable anti-tumor immunity," describes that Boltbody ISACs initiated tumor killing and antigen presentation by myeloid cells with subsequent T cell-mediated anti-tumor immunity. The data indicate that Boltbody ISACs activate the innate and adaptive immune systems, ultimately resulting in complete tumor regressions in multiple tumor models and durable anti-tumor immunity. ISAC-mediated immunological memory was not limited to the target antigen as ISAC-treated mice were protected from rechallenge with the parental tumor lacking HER2 expression. These results provide a strong rationale for the currently ongoing Phase 1/2, multi-center, open-label study of the company’s lead candidate, BDC-1001, and the continued development of the Boltbody ISAC technology platform and Bolt’s immuno-oncology pipeline.

Michael Alonso, Ph.D., scientific co-founder and vice president of immunology and pharmacology at Bolt, stated, "This publication lays the framework for therapeutic strategies that harness Boltbody ISACs and the profound biological synergies that occur following covalent attachment of an immunostimulant to a tumor-targeting antibody – notably, the complete elimination of well-established tumors and mobilization of T cells that prevent tumor recurrence."

Shelley Ackerman, Ph.D, lead author and co-inventor of the platform, said, "The publication of this peer-reviewed manuscript is an important milestone for Bolt with the first demonstration of the potential of our Boltbody ISAC platform. The data demonstrate the advantages achieved through requiring tumor target engagement, Fc receptor-mediated phagocytosis and subsequent TLR activation to induce a potent and localized anti-tumor response. This process effectively bridges the innate and adaptive immune systems to launch a coordinated anti-tumor response."

David Dornan, Ph.D., senior vice president of research and manufacturing at Bolt, added, "The work described is a culmination of years of experimentation and collaboration supported by our dedicated and talented team. This novel technology can be applied across a diverse range of tumor targets and has the potential to enable cancer patients to generate immunological memory against their own tumors. Our published data highlight the fact that while our Boltbody ISACs are administered systemically, the immune activation takes place where it is needed – in the tumor microenvironment. We look forward to reporting data from our ongoing Phase 1/2 clinical study assessing intravenous administration of our lead clinical molecule, BDC-1001, for the treatment of HER2-expressing tumors."

About Bolt Biotherapeutics’ Immune-Stimulating Antibody Conjugate (ISAC) Platform Technology
The Boltbody ISAC platform technology harnesses the ability of innate immune agonists to convert cold tumors into immunologically hot tumors, thereby illuminating tumors to the immune system and allowing them to be invaded by tumor killing cells. Boltbody ISACs have demonstrated the ability to eliminate tumors following systemic administration as monotherapy in preclinical models and have also led to the development of immunological memory, which is predicted to translate into more durable clinical responses for patients.

About the Ongoing BDC-1001 Phase 1/2 Study in Patients with HER2-Expressing Solid Tumors
The Phase 1/2, multi-center, open-label study is evaluating the safety, pharmacokinetics, pharmacodynamics and proof of mechanism of BDC-1001 in patients with HER2-expressing solid tumors. The first portion of the study includes a monotherapy dose-escalation phase in which cohorts of patients will receive ascending intravenous doses of BDC-1001 to determine the maximum tolerated dose and/or the recommended dose to advance into expansion cohorts and Phase 2 based on safety and tolerability. The second portion of the study is a dose expansion phase in which patients will receive BDC-1001 monotherapy to further evaluate the safety, tolerability and clinical antitumor activity of the recommended Phase 2 dose. Please refer to clinicaltrials.gov NCT04278144 for additional clinical trial information.

Boehringer Ingelheim Collaborates with Proxygen to Explore Molecular Glue Degraders – a Novel Approach to Fight Cancer

On December 7, 2020 Boehringer Ingelheim and Proxygen reported they have entered into a collaboration and license agreement to enable the identification of molecular glue degraders against various oncogenic targets (Press release, Boehringer Ingelheim, DEC 7, 2020, View Source [SID1234572405]). The collaboration combines Proxygen’s unique molecular glue degrader discovery platform and its expertise in targeted protein degradation with Boehringer Ingelheim’s long-term strategy to provide first-in-class, breakthrough therapies for cancer patients.

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!

Molecular glue degraders and PROTACs harness the power of the cell’s recycling machinery to selectively eliminate disease-causing proteins. Molecular glue degraders achieve this by specifically bridging the distance between target proteins and ubiquitin ligases, which consequently flag the target proteins for rapid degradation. Molecular glues orchestrate this protein-protein proximity through highly cooperative binding. They circumvent the need for a defined binding pocket on the surface of the target protein, a requirement for conventional small molecule drugs, allowing pharmaceutical intervention on proteins that were previously considered undruggable.

"Proxygen’s molecular glue degrader platform has the potential to become a valuable component of Boehringer Ingelheim’s cancer research strategy to treat tumors driven by the most frequently mutated cancer-causing proteins that were previously considered undruggable and to further expand our efforts in the area of targeted protein degradation," says Norbert Kraut, Global Head of Cancer Research at Boehringer Ingelheim. "We are very pleased to partner with Proxygen, complementing our efforts in taking difficult-to-treat cancers on."

The identification of molecular glue degraders has been serendipitous up to this point. Proxygen’s highly scalable and broadly applicable molecular glue discovery platform promises a goal-oriented and comprehensive avenue towards an improved identification of molecular glue degrader candidates.

"We are very excited to join forces with Boehringer Ingelheim," says Bernhard Boidol, Chief Executive Officer of Proxygen. "Working together with a leader in innovative oncological research and development not only validates the broad applicability of our molecular glue degrader platform but also allows us to rapidly develop new therapies for the high unmet medical need of many patients with lung and colorectal cancers."

Proxygen’s new method has recently been recognized by Boehringer Ingelheim through its Grass Roots ‘Innovation Prize’ in Vienna, Austria. The ‘Innovation Prize’ recognizes the innovation power of young life-science companies and bio-entrepreneurs to create and sustain pipelines for the next generation of medicines. Launched in 2015, the Grass Roots programs comprise of ‘Office Hours,’ ‘Academy’ and the ‘Innovation Prize.’ As a company dedicated to improving health and quality of life, these programs give Boehringer Ingelheim the opportunity to lend expertise to the early-stage innovation community and offer guidance around the science to help enable ideas to deliver the next breakthroughs.

Proxygen is entitled to receive up-front payments, research and development support and milestone gated development payments, as well as tiered royalties based on future commercial sales of developed products.

Arch Oncology Announces First Patient Dosed in Phase 1/2 Clinical Trial of Anti-CD47 Antibody AO-176 in Multiple Myeloma

On December 7, 2020 Arch Oncology, Inc., a clinical-stage immuno-oncology company focused on the discovery and development of anti-CD47 antibody therapies, reported that the first patient has been dosed in a new Phase 1/2 clinical trial for AO-176 in relapsed/refractory multiple myeloma (Press release, Arch Oncology, DEC 7, 2020, View Source;utm_medium=rss&utm_campaign=arch-oncology-announces-first-patient-dosed-in-phase-1-2-clinical-trial-of-anti-cd47-antibody-ao-176-in-multiple-myeloma [SID1234572317]). AO-176 is an anti-CD47 antibody with a potential best-in-class profile that works by blocking the "don’t eat me" signal and also by directly killing tumor cells, with preferential binding to tumor versus normal cells. In preclinical myeloma models involving large tumors, AO-176 has demonstrated promising activity as a single agent as well as in combination with standard approved therapies used to treat this disease.

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 begin dosing patients in our second clinical trial for AO-176," said Julie Hambleton, M.D., Interim President and Chief Executive Officer of Arch Oncology. "This is an important milestone for AO-176 and for patients as this is the first dedicated trial of an anti-CD47 antibody exclusively in patients with multiple myeloma. In our first clinical trial of AO-176, we saw encouraging anti-tumor activity as a single agent in patients with solid tumors and we are excited to evaluate our therapy for patients with multiple myeloma. With this second clinical trial initiated, we are making progress advancing AO-176 for both solid tumors and hematologic malignancies as we aim to deliver new cancer treatments to broader groups of patients."

Paul Richardson, M.D., Director of Clinical Research at the Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute and Co-Principal Investigator for this clinical trial, commented, "Research shows AO-176 has a highly-differentiated mechanism among this promising new class of anti-CD47 agents. We look forward to evaluating AO-176 as a monotherapy and in combination with standard therapies for patients with relapsed and refractory multiple myeloma. Patients who have progressed despite multiple lines of prior treatment urgently need new treatment options, and we are eager to assess the safety and preliminary efficacy profile of this exciting novel agent."

This open-label, multi-center, dose-escalation Phase 1/2 trial is evaluating the safety, tolerability, pharmacokinetics/pharmacodynamics, and preliminary efficacy of AO-176 in patients with relapsed/refractory multiple myeloma. Up to 100 patients whose disease has progressed following at least three prior lines of treatment will be enrolled. In Phase 1, patients enrolled in up to four dose-escalation cohorts will receive AO-176 monotherapy. Next, patients will receive AO-176 in combination with dexamethasone and bortezomib. Then, in Phase 2, patients will receive a recommended dose of AO-176 in combination with dexamethasone and bortezomib to evaluate the safety and preliminary efficacy of this combination.

Parameswaran Hari, MD, MRCP, MS, Chief of the Division of Hematology and Oncology in the Department of Medicine and Professor at Medical College of Wisconsin and Principal Investigator for this clinical trial, added, "We are pleased to dose the first patient in this new multiple myeloma clinical trial. As we treat patients with multiple myeloma, an incurable disease for many patients, we look forward to evaluating AO-176 as a potential new treatment option for patients living with this disease. Given the mechanism and profile of AO-176, we envision additional future combinations with AO-176 to evaluate as future treatments for patients."

Recent Preclinical Data Presentations on AO-176 in Multiple Myeloma

Event: ASH (Free ASH Whitepaper) Annual Meeting & Exposition 2020

Date: December 5, 2020 7:00 am – 3:30 pm PT
Abstract Title: Pre-clinical Combination of AO-176, a Highly Differentiated Clinical Stage CD47 Antibody, with Either Azacitidine or Venetoclax Significantly Enhances DAMP Induction and Phagocytosis of Acute Myeloid Leukemia

Date: December 6, 2020 7:00 am – 3:30 pm PT
Abstract Title: AO-176, a Highly Differentiated Clinical Stage Anti-CD47 Antibody, Exerts Potent Anti-Tumor Activity in Preclinical Models of Multiple Myeloma as a Single Agent and in Combination With Approved Therapeutics

About AO-176

AO-176 is a humanized anti-CD47 IgG2 antibody with a potential best-in-class profile. AO-176 is highly differentiated, with the potential to improve upon the safety and efficacy profile relative to other agents in this class of innate checkpoint inhibitors. AO-176 works by blocking the "don’t eat me" signal, the standard mechanism of anti-CD47 antibodies. Beyond blocking this signal, AO-176 has additional mechanisms, including directly killing tumor cells and inducing DAMPs (Damage Associated Molecular Patterns), resulting in Immunogenic Cell Death. Importantly, AO-176 binds preferentially to tumor cells, instead of to normal cells, and binds even more potently to tumors in their acidic microenvironment (low pH). Publications and presentations on AO-176 can be found at View Source

AO-176 is being evaluated in Phase 1/2 clinical trials for the treatment of patients with select solid tumors and multiple myeloma, both as monotherapy and in combination with standard therapies. In a Phase 1 trial in solid tumors, AO-176 demonstrated encouraging safety and evidence of anti-tumor activity when administered as a single agent. Additional information about these trials may be found at www.clinicaltrials.gov using the trial identification number NCT03834948 (solid tumors) or NCT04445701 (multiple myeloma).

KaliVir Immunotherapeutics and Astellas Enter Worldwide Exclusive Licensing Agreement for Development and Commercialization of VET2-L2 Novel Oncolytic Virus

On December 7, 2020 KaliVir Immunotherapeutics LLC (CEO: Helena Chaye, Ph.D., J.D., "KaliVir") and Astellas Pharma Inc. (TSE: 4503, President and CEO: Kenji Yasukawa, Ph.D., "Astellas") reported that they entered into a worldwide licensing agreement for the research, development, and commercialization of VET2-L2, an intravenously administered oncolytic virus for Immuno-Oncology, as well as a research collaboration to generate a Second Product, a follow-on virus (Press release, Astellas, DEC 7, 2020, View Source [SID1234572335]).

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!

KaliVir holds its unique technology platform based on genetically modified vaccinia virus, and is developing VET2-L2, an oncolytic vaccinia virus as their lead program. VET2-L2, which is delivered by intravenous administration, reaches and destroys cancer cells and activates anti-cancer immunity through expression of therapeutic transgenes. KaliVir’s vaccinia virus-based oncolytic viral immunotherapies can be delivered intravenously to cancer patients, eliminating the need for complicated procedures of the direct intra-tumoral administration and enables access for a broader patient population. VET2-L2 is in preclinical stage.

This collaboration, which combines KaliVir’s expertise in the development of oncolytic viruses with Astellas’ capabilities in advanced drug development and its global business experience, will enable both parties to develop new Immuno-Oncology therapies.

Under the terms of the agreement, Astellas will pay to KaliVir up to US$56 million in the form of an upfront payment and other payments to support research and preclinical activities related to VET2-L2 and the Second Product. Additionally, Astellas may pay up to US$307 million and up to US$271 million for development, regulatory and commercialization of VET2-L2 and Second Product, respectively. Astellas also may make royalty payments on net sales of each licensed product.

"We are thrilled that Astellas has chosen KaliVir, and specifically VET2-L2, our lead product candidate, to add to their oncology program. VET2-L2, a multi-mechanistic, intravenously-delivered oncolytic vaccinia virus, has demonstrated strong precinical data prompting us to plan our initial clinical trial for VET2-L2," said KaliVir CEO Helena Chaye, Ph.D., J.D. "With Astellas’ excellent track record for drug development and commercialization, we believe that their commitment to collaborating with KaliVir represents strong third-party validation for our VET platform. We are commited to bringing this exciting product to cancer patients and believe that our collaboration with Astellas will expedite our ability to do this."

Naoki Okamura, Representative Director, Corporate Executive Vice President, Chief Strategy Officer and Chief Financial Officer, at Astellas said, "We, at Astellas, have positioned Immuno-Oncology as one of the Primary Focuses of our R&D strategy, and we are committed to developing the next generation of Immuno-Oncology therapies through new modalities and technologies. Oncolytic viruses are one of the therapies in which we are particularly focused as we strive to provide new options for patients who have no effective treatment options. We expect this KaliVir collaboration to enhance our pipeline and further expand our cancer treatment options as we work to develop innovative medical solutions that turn innovative science into VALUE for patients."