Additional product patent in the US for HyNap-Dasa

On February 12, 2020 Xspray Pharma (Nasdaq First North Growth Market: XSPRAY) reported that the United States Patent and Trademark Office (USPTO) has granted Xspray a new US patent for HyNap-Dasa (Press release, Xspray, FEB 12, 2020, View Source [SID1234649566]). The new patent, US 10,555,937, covers the pharmaceutical composition of the company’s primary product candidate, HyNap-Dasa. This is Xspray’s fourth product patent in the United States, which is the company’s main market. The patent is valid until January 11, 2033.
"This new patent on our most important market is yet another confirmation of our innovative development work. The formal clinical bioequivalence of the company’s primary product candidate HyNap-Dasa, which has been previously communicated, means that the patent further strengthens our position in negotiations with potential partners", says Per Andersson, CEO of Xspray.

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!

Xspray strives to obtain patents for both composition and technology and this new patent claims an amorphous solid dispersion (pharmaceutical composition) of dasatinib.

"Our strategic patent work will generate additional patents in the United States on our product candidates in the near future. The new HyNap-Dasa patent makes it significantly more difficult for other companies to launch a dasatinib product based on amorphous solid dispersion in the United States during the lifetime of the patent, i.e. up until January 2033", Per Andersson concludes.

Cue Biopharma to Present at the Biomarkers Series UK

On February 12, 2020 Cue Biopharma, Inc. (NASDAQ: CUE), a clinical-stage biopharmaceutical company engineering a novel class of injectable biologics to selectively engage and modulate targeted T cells within the body, reported Dr. Mary Simcox, vice president of translational biology and early development, will give a presentation highlighting the strategy and tactical details for testing CUE-101, the company’s lead biologic drug candidate, in a first-in-human Phase 1 trial (NCT03978689) at the Biomarkers Series UK taking place on Feb. 18-20, 2020 at the Manchester Central Convention Complex (Press release, Cue Biopharma, FEB 12, 2020, View Source [SID1234608305]). CUE-101 is designed to directly engage and activate T cells in the body to target HPV16-driven recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), enabled by the company’s proprietary therapeutic platform Immuno-STAT (Selective Targeting and Alteration of T cells).

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!

Presentation Details
Title: CUE-101 Phase 1 Trial in HNSCC Patients: Novel Immunotherapy Enabled by Patient Stratification & Pharmacodynamic Biomarkers
Session: Biomarker Congress Workshop, End-user Session, Designing/Applying a Biomarker Plan in Drug Development
Presenter: Mary Simcox, Ph.D., vice president of translational biology and early development
Date & Time: Feb. 18 at 4:15 p.m. GMT

"This presentation focuses on the importance of conducting early clinical immuno-oncology trials that are informed by detailed biomarker analyses, which characterize the mechanism of action and provide guidance on how the drug is used to maximize the potential for clinical benefit in cancer patients," said Ken Pienta, M.D., acting chief medical officer of Cue Biopharma. "The CUE-101 Phase 1 protocol includes implementation of multiple technologies and multiple industry and academic partners in the investigation of pharmacodynamic, response prediction, patient selection and response monitoring biomarkers. These critical biomarker data will be collected with the aim to discover correlative pharmacodynamic and response prediction markers to optimize the probability of clinical success and development of a promising and potentially new and transformative therapeutic."

About the CUE-100 Series
The CUE-100 series consists of Fc-fusion biologics that incorporate peptide-MHC (pMHC) molecules along with rationally engineered IL-2 molecules. This singular biologic is designed to selectively target, activate and expand a robust repertoire of tumor-specific T cells directly in the patient. The binding affinity of IL-2 for its receptor has been deliberately attenuated to achieve preferential selective activation of tumor-specific effector T cells while reducing potential for effects on regulatory T cells (Tregs) or broad systemic activation, potentially mitigating the dose-limiting toxicities associated with current IL-2-based therapies.

About Immuno-STAT
Immuno-STAT biologics are designed for targeted modulation of disease-associated T cells in the areas of immuno-oncology and autoimmune disease. Each of our biologic drugs is designed using our proprietary scaffold comprising: 1) a peptide-MHC complex (pMHC) to provide selectivity through interaction with the T cell receptor (TCR), and 2) a unique co-stimulatory signaling molecule to modulate the activity of the target T cells.

The simultaneous engagement of co-stimulatory molecules and pMHC binding mimics the signals delivered by antigen presenting cells (APCs) to T cells during a natural immune response. This design enables Immuno-STAT biologics to engage with the T cell population of interest, resulting in highly targeted T cell modulation. Because our drugs are delivered directly in the patient’s body (in vivo), they are fundamentally different from other T cell therapeutic approaches that require the patients’ T cells to be extracted, stimulated and expanded outside the body (ex vivo), and reinfused in an activated state.

NETRF funds $3.5 million in research to advance neuroendocrine tumor treatment

On February 12, 2020 The Neuroendocrine Tumor Research Foundation (NETRF), Boston, MA, reported $3.5 million in neuroendocrine tumor (NET) research grants to fund 12 projects around the world in pursuit of more precise treatments for this uncommon cancer affecting an estimated 171,000 Americans (Press release, NETRF, FEB 12, 2020, View Source [SID1234554385]).

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!

After investing $26 million in research during the past 15 years, NETRF has helped to establish the NET knowledge base needed to expand the exploration of improved treatments, according to Elyse Gellerman, NETRF chief executive officer. "We can see real momentum in this new round of grants. We hope the discoveries from these projects will lead to improved treatment options for patients."

NETRF is supporting a new pioneering approach to NET immunotherapy with a Petersen Accelerator Award to Steven Libutti, MD, Rutgers Cancer Institute of New Jersey, to characterize a novel immune regulator called B7x to determine whether it has a role in shutting off the body’s immune response to fight against pancreatic NETs.

This round of funding features multiple new fronts for NETRF. To help grow the NET scientific workforce, the Foundation granted two inaugural Mentored Awards for early career researchers, one of which was funded by an educational grant from Ipsen. There were also new areas of NET inquiry. For the first time, NETRF is funding pheochromocytoma and paraganglioma research, including an evaluation of a novel radiotracer for imaging adrenal NETs.

NETRF also funded four research projects in lung NETs, an area that has not previously received the attention of other NET sites. These lung studies include:

Conducting single-cell genomic analyses to understand how lung NETs form, grow, and spread.
Mapping the cellular networks of typical and atypical lung NETs to find biomarkers that help predict a tumor’s aggressiveness.
Characterizing the molecular makeup of a newly identified, aggressive lung NET called "supra-carcinoid."
Determining the sociodemographic and geographic patterns of lung NETs in California.
"Advances in NET research have been hampered by the lack of effective laboratory disease models, and a limited understanding of the molecular and genetic profiles of NETs," said John Kanki, PhD, NETRF director of research. "Now that we are making strong headway along these lines, we can finally begin to drill deeper with greater specificity, to identify and explore new strategies for treating NETs."

NET tumors require the expansion of new blood vessels in order to grow and spread and two new grants explore new therapies that target developing tumor blood vessels. Researchers at Vanderbilt University will explore the potential efficacy of a combination therapy testing a drug known to affect the formation of new blood vessels (cabozantinib) together with an experimental drug called CB-839. At Columbia University Medical School, scientists will conduct preclinical laboratory experiments to test whether turning off two complementary blood vessel-forming processes together can improve therapeutic efficacy in pancreatic NETs.

NETRF also approved grants to study a potential cause of small intestinal multifocal tumors and to evaluate the role of the gut microbiome in carcinoid syndrome.

Seventy-five percent of the grants were awarded to research institutions that are new to NETRF including five grants funding international research at Erasmus MC, University Medical Center Rotterdam, Netherlands; Hebrew University of Jerusalem, Israel; International Agency for Research on Cancer (IARC-WHO), Lyon, France; Istituto Auxologico Italiano – Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy; and Weizmann Institute of Science, Rehovot, Israel.

Seven of the 12 grants fund innovative NET research at American academic institutions, including Columbia University Medical Center, New York, NY; Dana-Farber Cancer Institute, Boston, MA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ; University of California, San Francisco, CA; Tufts Medical Center, Boston, MA; University of Michigan, Ann Arbor, MI; and Vanderbilt University Medical Center, Nashville, TN.

The NETRF grant process is a competitive and structured peer-reviewed process. The Foundation is currently accepting applications for its next grant cycle. Applications must be received by March 9, 2020. View Source

Gellerman thanked the many individuals and foundations whose gifts to the 501(c)(3) nonprofit organization support NETRF’s research and educational activities. A generous gift from the Margie and Robert E. Petersen Foundation will fund several of the new projects. Additional support has been provided by the Goldhirsh-Yellin Foundation of Los Angeles and Advanced Accelerator Applications.

Designer probiotic treatment for cancer immunotherapy

On February 12, 2020 Columbia Engineering reported that have engineered probiotics to safely deliver immunotherapies within tumors (Press release, Columbia University, FEB 12, 2020, View Source [SID1234554384]). These include nanobodies against two proven therapeutic targets–PD-L1 and CTLA-4. The drugs are continuously released by bacteria and continue to attack the tumor after just one dose, facilitating an immune response that ultimately results in tumor regression. The versatile probiotic platform can also be used to deliver multiple immunotherapies simultaneously, enabling the release of effective therapeutic combinations within the tumor for more difficult-to-treat cancers like colorectal cancer. The study is published today in Science Translational Medicine.

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!

Antibodies that target immune checkpoints, PD-L1 and CTLA-4, have revolutionized cancer immunotherapy treatments, achieving success in a subset of cancers. However, systemic delivery of these antibodies can also cause substantial side effects with high percentages of patients reporting adverse reactions. Furthermore, although combinations of these therapies are more effective than single therapy regimens, they also produce severe toxicities, sometimes leading to drug discontinuation. The team, led by Tal Danino View Source, assistant professor of biomedical engineering View Source, aimed to address these challenges.

"We wanted to engineer a safe probiotic vehicle capable of delivering immune checkpoint therapies locally to minimize side effects," says Danino, who is also a member of the Herbert Irving Comprehensive Cancer Center View Source and Data Science Institute. "We also wanted to broaden the versatility of the system by producing a range of immunotherapeutic combinations, including cytokines that could further elicit antitumor immunity, but are otherwise difficult to systemically deliver because of toxicity concerns."

Bacterial cancer therapy is not a new idea: in the 1890s, William Coley, a New York City surgeon, demonstrated that injection of live streptococcal organisms into cancer patients could shrink tumors. While his method was never widely adopted because radiotherapy was discovered around the same time and antibiotics were not widely available, physicians have been using a tuberculosis vaccine, BCG, as a therapy for bladder cancer for decades.

The Danino lab has pioneered engineered bacteria for cancer therapy, developing methods to characterize different strains of bacteria, therapeutics, and genetic control circuits to effectively release cancer drugs. In this most recent study, led by PhD student Candice Gurbatri, they sought to engineer a translational therapeutic platform that improved upon a previous lysis circuit.

Using computational modeling, they first scanned multiple parameters to find the optimal circuit variants to maximize drug release within the tumor. This led to the integration of the circuit into the genome of a widely-used probiotic strain–E.coli Nissle 1917–resulting in a strain they call "SLIC," or the synchronized lysing integrated circuit. This SLIC probiotic strain is naturally capable of finding and growing within tumors in the body, but the genomic integration of this circuit ensures greater stability of the system and higher levels of therapeutic release.

"We have demonstrated that the engineered bacteria remain functional and localized within the tumor as the bacteria grow in mice for at least two weeks after treatment, preventing the microbes from affecting healthy tissue," says Gurbatri. Testing in mouse models further demonstrated that unlike previous iterations of the circuit, SLIC was able to clear tumors after a single dose, adding to its translational potential. Because the circuit is integrated into the genome, the stability of the platform greatly increases, thus negating the need for multiple injections of bacteria.

The research team used this probiotic delivery system to release nanobodies blocking PD-L1 and CTLA-4 within tumors in mouse models of lymphoma and colorectal cancer. It is already known that tumors express these checkpoints to stop the immune system, specifically T cells, from functioning properly. The goal of blocking PD-L1 and CTLA-4 is to remove the "brakes" and enable T cells to attack the cancer. A direct comparison to clinically relevant antibodies against the same target showed that their probiotic therapy was more effective, leading to complete tumor regression and prevention of metastatic formation in early and late-stage mouse models of lymphoma.

Leveraging the versatility of this system, the researchers sought to treat more difficult cancers, like colorectal, that have been less responsive to traditional immunotherapies. In this additional model, they paired the immune checkpoint nanobodies with a cytokine to further stimulate the immune system. A single dose of this probiotic cocktail resulted in tumor regression with no observed side-effects.

Says Gurbatri, "We showed that a triple combination of immunotherapies could effectively reduce tumor growth in a cancer that is generally less responsive to immunotherapy. We’ve demonstrated that one dose of our probiotic therapy results in continuous localized drug release and clearance of the bacteria population once tumors have cleared. These elements could be particularly beneficial in a clinical setting, where patients want and need minimally invasive and self-sustained therapies."

The biomedical engineers worked closely with colleagues, including Assistant Professor Nicholas Arpaia, in the microbiology and immunology departments at Columbia University Irving Medical Center. The team is currently performing further safety and toxicology studies of their engineered probiotic in genetically modified mouse models of cancer. They are also collaborating with physicians on the translational aspects of their work and have also founded a company, GenCirq Inc., to translate their promising technology to patients.

About the Study

The study is titled "Engineered probiotics for local tumor delivery of checkpoint blockade nanobodies."

Authors are: Candice R. Gurbatri 1, Ioana Lia 1, Rosa Vincent 1, Courtney Coker 1, Samuel Castro 1, Piper M. Treuting 2, Taylor E. Hinchliffe 1, Nicholas Arpaia 3, 4, Tal Danino 1, 4, 5.

1 Department of Biomedical Engineering, Columbia Engineering

2 Department of Comparative Medicine, University of Washington, Seattle

3 Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University

4 Herbert Irving Comprehensive Cancer Center, Columbia University

5 Data Science Institute, Columbia University

The study was supported in part by the NIH Pathway to Independence Award (R00CA197649- 02 to T.Danino.), DoD Idea Development Award (LC160314 to T.Danino.), DoD Era of Hope Scholar Award (BC160541 to T.Danino.), Breast Cancer Research Foundation AACR (Free AACR Whitepaper) Career Development Award for Translational Breast Cancer Research (PC 516160 to T.Danino.), National Institute of General Medical Sciences of the National Institutes of Health (R01GM069811to T.Danino.), Bonnie J. Addario Lung Cancer Foundation Young Innovators Team Award (YITA, to T.Danino. and N.Arpaia.) and the National Science Foundation Graduate Research Fellowship (1644869 to C.Gurbatri.).

C.Gurbatri., N.Arpaia., and T.Danino. have filed a patent application ("Programmable bacteria for the treatment of cancer") with the U.S. Patent and Trademark Office (U.S. patent application no. PCT/US19/42795) related to this work. T.Danino. and N.Arpaia. have a financial interest in GenCirq Inc.

OSE Immunotherapeutics Announces Artificial Intelligence Antibody Drug Development Collaboration with MAbSilico

On February 12, 2020 OSE Immunotherapeutics reported to incorporate innovative problem-solving solutions like AI for the development of new monoclonal antibodies (Press release, OSE Immunotherapeutics, FEB 12, 2020, View Source [SID1234554357]). MAbSilico solutions have already been tested and validated by OSE Immunotherapeutics and will be used for six antibody programs, including novel bispecific antibodies. AI and numerical simulation can guide therapeutic antibody discovery, help reduce the risk of failure and accelerate the pre-clinical development process of these drug candidates before clinical tests.

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!

Nicolas Poirier, Chief Scientific Officer of OSE Immunotherapeutics, stated: "We are delighted with this collaboration, we constantly strive to introduce innovative technologies to develop first-in-class products in immuno-oncology and autoimmune diseases. Due to the devastating nature of these diseases, our development strategies need to be accelerated and artificial intelligence solutions for drug discovery offered by MAbSilico can be a great asset to achieve this goal."

While all MAbSilico’s commercialized solutions are included in this three-year agreement, OSE Immunotherapeutics also gains early access to MAbSilico’s SaaS (Software as a Service) and technologies in development including those for the conception of therapeutic antibodies optimized for bioproduction. OSE provides internal data to MAbSilico in order to feed their algorithms in development and deliver new technology faster.

Puard, Chief Executive Officer of MAbSilico, stated: "It is a pleasure to start this collaboration with one of the top French Biotech. This partnership demonstrates the need to fasten new AI-based solutions for antibody drug discovery. We believe that with the trust and collaboration of OSE, we will accelerate the release of our software and new technologies."