Syros Announces New Preclinical Data on SY-1365 Showing Potent Anti-Tumor Activity in Multiple Models of Heavily Pretreated Ovarian Cancer

On April 16, 2018 Syros Pharmaceuticals (NASDAQ: SYRS), a biopharmaceutical company pioneering the discovery and development of medicines to control the expression of genes, reported new preclinical data showing that SY-1365, its first-in-class selective cyclin-dependent kinase 7 (CDK7) inhibitor currently in a Phase 1 trial in patients with advanced solid tumors, demonstrated potent anti-tumor activity in multiple models of heavily pretreated ovarian cancer (Press release, Syros Pharmaceuticals, APR 16, 2018, View Source [SID1234525362]). These data were presented by Syros and its collaborators from the Dana-Farber Cancer Institute (DCFI) at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting in Chicago.

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!

"Despite recent advances in treating ovarian cancer, many patients either don’t respond or become resistant to treatment, and the outlook for these patients is poor," said Panagiotis A. Konstantinopoulos, M.D, Ph.D., Director of Translational Research and attending oncologist in the Gynecologic Oncology Program at DCFI and an Associate Professor of Medicine at Harvard Medical School. "SY-1365 stopped tumor growth in multiple preclinical models of ovarian cancer that had progressed following treatment with standard-of-care and targeted therapies. These data suggest SY-1365 has the potential to be an important new therapy for ovarian cancer that warrants further clinical investigation."

Researchers from Syros and DCFI evaluated the anti-tumor activity of SY-1365 in a broad panel of ovarian cancer cell lines, as well as in patient-derived xenograft (PDX) mouse models developed from patients treated with multiple prior therapies, including standard-of-care platinum-based therapies and a new class of targeted therapies known as PARP inhibitors. The data, which were presented in a poster discussion session and a poster session, show that SY-1365:
Induced cell death in numerous ovarian cancer cell lines.
Inhibited tumor growth in 10 of the 17 treatment-relapsed ovarian PDX models studied, including inducing complete regressions. Notably, these responses were observed irrespective of BRCA status or sensitivity to a PARP inhibitor.
Lowered expression of MCL1, a gene in the mitochondrial apoptosis pathway that is known to inhibit apoptosis, or programmed cell death.

The data also showed that sensitivity to SY-1365 was associated with low expression of BCLXL, a known apoptosis inhibitor, and RB1, a known tumor suppressor, pointing to potential biomarkers that may be predictive of response to SY-1365.

"We are very encouraged by these data," said David A. Roth, M.D., Chief Medical Officer of Syros. "CDK7 has emerged as a potentially important new drug target in cancer. By selectively inhibiting CDK7, SY-1365 has been shown to lower the expression of cancer driving genes and hit cancer cells at multiple points in the cell cycle, preferentially killing cancer cells and inhibiting tumor growth in preclinical models of a number of difficult-to-treat solid tumors and blood cancers. These newest data in ovarian cancer provide strong support for the planned expansion of our Phase 1 clinical trial into ovarian cancer and further highlight the promise of SY-1365 to make a profound difference for patients."

The ongoing Phase 1 trial of SY-1365 is a multi-center, open-label trial enrolling patients with advanced solid tumors. The primary objective of the trial is to assess the safety and tolerability of escalating doses of SY-1365, with the goal of establishing a maximum tolerated dose and a recommended Phase 2 dose and regimen. The dose-escalation phase is open and expected to enroll approximately 35 solid tumor patients for whom standard curative or palliative measures do not exist or are no longer effective. Following the dose-escalation phase, Syros plans to open expansion cohorts to further evaluate the safety and anti-tumor activity of SY-1365 as a single agent and in combination with standard-of-care therapies in multiple ovarian and breast cancer populations.
Syros expects to open the expansion phase of the trial in mid-2018 and to report data from the dose escalation portion of the trial in the fourth quarter of 2018.
About SY-1365

SY-1365 is a first-in-class selective cyclin-dependent kinase 7 (CDK7) inhibitor with potential across a range of difficult-to-treat solid tumors and blood cancers. In preclinical studies, SY-1365 has shown significant anti-proliferative and pro-apoptotic activity in difficult-to-treat cancers, including breast and ovarian cancers and acute leukemia. SY-1365 has also been shown to preferentially kill cancer cells over non-cancerous cells and lower the expression of cancer-driving genes, inducing significant anti-tumor activity, including complete tumor regressions, in preclinical models of these cancers. SY-1365 is currently in a Phase 1 clinical trial in patients with advanced solid tumors, with planned expansion cohorts to evaluate SY-1365 in multiple ovarian and breast cancer patient populations as a single agent and in combination with standard-of-care therapies. Additional details about the trial can be found using the identifier NCT03134638 at www.clinicaltrials.gov.

Harpoon Therapeutics Launches Immuno-Oncology Pipeline With Compelling Data From Lead Programs at the 2018 American Association for Cancer Research (AACR) Annual Meeting

On April 16, 2018 Harpoon Therapeutics, a biotechnology company pioneering a new class of T cell engaging therapeutics based on its proprietary TriTAC platform, reported that preclinical data supporting the ongoing development of the platform and its two lead molecules, HPN424 and HPN536 (Press release, Harpoon Therapeutics, APR 16, 2018, View Source [SID1234525414]). These programs are the first of four programs using TriTAC technology, which has been designed for superior tumor penetration and efficacy in combating solid tumors. The company, which announced a series B financing and a partnership with AbbVie in 2017, anticipates filing investigational new drug (IND) applications and entering the clinic with these two compounds in the next 12 months. Data were presented at the 2018 American Association of Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting in Chicago, held April 14-18, 2018.

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!

"Harpoon created TriTAC as a best-in-class T cell engager platform optimized for the treatment of solid tumors," said Jerry McMahon, PhD, President and CEO, Harpoon Therapeutics. "The platform allows us to bring the success of T cell engagers targeting hematologic malignancies, like Blincyto, to solid tumor treatment. At Harpoon, we have optimized serum half-life, maximized tumor penetration, and engineered superior stability and manufacturability. These features have been the foundation for our discovery and development of a robust pipeline of drug candidates targeting PSMA, mesothelin, BCMA and DLL3."

Harpoon presented preclinical data which highlighted the novel aspects of its proprietary TriTAC platform to potentially overcome the limitations of existing bispecific antibody-based and CAR-T therapies. The TriTAC platform uses a single flexible polypeptide comprised of three binding domains, and is designed to be the smallest, half-life extended T cell engaging format without the potential liabilities associated with conventional bispecific antibodies.

"We are excited to share these preclinical data for HPN424 and HPN536, which are designed to elicit targeted tumor cell destruction for both metastatic castration-resistant prostate cancer (mCRPC) and mesothelin-expressing tumors, respectively, such as lung, ovarian and pancreatic cancers," said Holger Wesche, PhD, Senior Vice President, Research. "We believe our new data underscore the significant impact our approach could make in advancing the field of immuno-oncology, and look forward to evaluating our compounds in clinical testing."

HPN424 Data Showed Potent T Cell Killing of Prostate Cancer Cells and Serum Half-Life Extension

HPN424 is a 50-kD single polypeptide containing three binding domains — for human PSMA, human serum albumin (HSA) and human CD3. In preclinical studies, HPN424 demonstrated single-digit picomolar potency for PSMA-dependent T cell killing in a panel of human prostate cancer cell lines, which translated to in vivo efficacy with efficacious doses in the low µg/kg range. HPN424 was well tolerated in a multi-dose safety study in non-human primates and showed a serum half-life of 80 hours supporting once-weekly administration for a Phase 1 dose-escalation study in mCRPC patients planned to begin this year.

HPN536 Data Showed Potent Destruction of Mesothelin-Positive Cancer Cells and Evidence of Mechanism in a Primate Study

HPN536 is a 50-kD single polypeptide containing three binding domains — for human mesothelin (MSLN), HSA and human CD3. In preclinical studies, HPN536 demonstrated single-digit picomolar potency for MSLN-dependent T cell killing in a panel of human cancer cell lines derived from mesothelioma, pancreatic, non-small cell lung and ovarian tumors, and in vivo efficacy with efficacious doses in the low µg/kg range. An exploratory safety study in non-human primates showed that HPN536 was well tolerated and supported weekly dosing in humans. The compound was also shown to elicit T cell activation resulting in mesothelial hypertrophy and lymphocyte infiltration, which strongly supports tissue penetration, the mechanism of action of the TriTAC platform, and the planned Phase 1 study.

ABSTRACT INFORMATION

Abstract #1773
Title: "HPN424, a half-life extended, PSMA/CD3-specific TriTAC for the treatment of metastatic prostate cancer"
Date and Time: April 16, 2018, 8:00 AM – 12:00 PM CT
Session: Therapeutic Antibodies, Including Engineered Antibodies 1

Abstract #1781
Title: "HPN536, a T cell-engaging, Mesothelin/CD3-specific TriTAC for the treatment of solid tumors"
Date and Time: April 16, 2018, 8:00 AM – 12:00 PM CT
Session: Therapeutic Antibodies, Including Engineered Antibodies 1

Abstract #3814
Title: "TriTACs are novel T cell-engaging therapeutic proteins optimized for the treatment of solid tumors and for long serum half-life"
Date and Time: April 17, 2018, 8:00 AM – 12:00 PM CT
Session: Therapeutic Antibodies, Including Engineered Antibodies 3

Bexion Pharmaceuticals, Inc. Presentations Accepted at 2018 ASCO Annual Meeting

On April 16, 2018 Bexion Pharmaceuticals, Inc., a clinical-stage biopharmaceutical company focused on rare brain tumors, reported that the four abstracts submitted for presentation at the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting held June 1-5, 2018 in Chicago, Illinois have all been accepted (Press release, Bexion, APR 16, 2018, View Source [SID1234525565]).

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 accepted presentations is entitled "First-in-human, First-in-class Phase 1a Study of BXQ-350 for Solid Tumors and Gliomas".

The ASCO (Free ASCO Whitepaper) Annual Meeting brings together more than 32,000 oncology professionals from around the world to discuss state-of-the-art treatment modalities, new therapies, and ongoing controversies in the field.

About BXQ-350

BXQ-350 is a unique formulation of a synthetically produced, human lysosomal protein, Saposin C (sphingolipid activator protein, or SapC), and the phospholipid dioleoylphosphatidylserine (DOPS).

Aptose Presents New Preclinical Data on CG’806 Pan-FLT3/ Pan-BTK Inhibitor at 2018 AACR Annual Meeting

On April 16, 2018 Aptose Biosciences Inc. (NASDAQ:APTO) (TSX:APS) reported the presentation of preclinical data demonstrating the robust cell killing ability of CG’806, a pan-FLT3/pan-BTK inhibitor, in multiple types of AML and B-cell malignancies (Press release, Aptose Biosciences, APR 16, 2018, View Source;p=RssLanding&cat=news&id=2342648 [SID1234525329]). Data further demonstrated that CG’806 targets multiple pathways and overcomes drug resistance seen with other inhibitors. The data were presented in a poster on Sunday, April 15, 2018 at the 2018 American Association for Cancer Research (AACR) (Free AACR Whitepaper) Conference being held April 14-18, in Chicago, IL.

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 poster, entitled CG’806, a first-in-class pan-FLT3/pan-BTK inhibitor, targets multiple pathways to kill diverse subtypes of acute myeloid leukemia and B-cell malignancy in vitro, explores the potency and molecular mechanisms of the pan-FLT3/pan-BTK inhibitor CG’806 in hematologic malignancies relative to other FLT3 or BTK inhibitors commercialized or in development. The Aptose research team led by Dr. Hannah Zhang, Senior Director of Research, demonstrated that in FLT3-ITD AML cells, CG’806 induced apoptosis through inhibition of FLT3 signaling, and CG806 was approximately 10-fold more potent than quizartinib. Although FLT3-ITD is found in approximately 30% of AML patient, most AML patients express wild type (WT) FLT3. CG’806 was superior to quizartinib, gilteritinib and crenolanib FLT3 inhibitors in FLT3-WT AML cell lines. In B cell malignancies, BTK signaling plays a pivotal pathogenic role. CG’806 decreased BTK phosphorylation in all malignant B cell lines tested and inhibited cell proliferation and colony formation 50-6,000 times more potently than ibrutinib, an effect explained by the ability of CG’806 to target multiple rescue pathways rather than merely the exclusive inhibition of BTK signaling.

CG’806 demonstrated the ability to target all wild type (WT) and mutant forms of FLT3 and BTK and to inhibit multiple signaling pathways, producing killing of diverse subtypes of hematologic malignancies driven by different genomic aberrations.

"This study directly compares CG’806 to other FLT3 or BTK inhibitors in development and confirms the potent and extended activity we have seen with the molecule," said William G. Rice, Ph.D., Chairman and Chief Executive Officer of Aptose. "As a pan-FLT3/pan-BTK multi-kinase inhibitor that can eliminate tumors in the absence of toxicity in animal models, CG’806 has demonstrated the ability to kill a broad range of AML and B-cell malignancies through inhibition of multiple oncogenic pathways. We are eager to pursue its clinical development."
Separately, Aptose and Oregon Health & Science University (OHSU) Knight Cancer Center researchers also announced new data on CG’806 presented at AACR (Free AACR Whitepaper) (see press release here). Both poster presentations will be published in the AACR (Free AACR Whitepaper) Conference Proceedings. The posters can also be accessed here or at the Publications & Presentations section of the Aptose website, www.aptose.com.

About CG’806
CG‘806 is an oral, first-in-class pan-FLT3/pan-BTK multi-kinase inhibitor. This small molecule demonstrates potent inhibition of wild type and mutant forms of FLT3 (including internal tandem duplication, or ITD, and mutations of the receptor tyrosine kinase domain and gatekeeper region), eliminates acute myeloid leukemia (AML) tumors in the absence of toxicity in murine xenograft models, and represents a potential best-in-class therapeutic for patients with AML. Likewise, CG’806 demonstrates potent, non-covalent inhibition of the wild type and Cys481Ser mutant forms of the BTK enzyme, as well as other oncogenic kinase pathways operative in B cell malignancies, suggesting CG’806 may be developed for various B cell malignancy patients (including CLL, MCL, DLBCL and others) that are resistant/refractory/intolerant to covalent BTK inhibitors. CG’806 is currently in preclinical development in partnership with CrystalGenomics.

Idera Pharmaceuticals Enters into a Clinical Development Support Agreement with Pillar Partners Foundation to Expand the Clinical Research on IMO-2125 beyond PD-1 Refractory Melanoma

On April 16, 2018 Idera Pharmaceuticals, Inc. ("Idera") (NASDAQ:IDRA), a clinical-stage biopharmaceutical company focused on the discovery, development and commercialization of novel oligonucleotide therapeutics for oncology and rare diseases, reported it has entered into a clinical development support agreement with Pillar Partners Foundation ("Pillar Partners") (Press release, Idera Pharmaceuticals, APR 16, 2018, View Source [SID1234525345]). Under the terms of the agreement Pillar Partners will provide direct funding to support three investigator initiated clinical trials to further strategically expand the clinical research of IMO-2125, Idera’s toll-like receptor ("TLR") 9 agonist into broader melanoma populations and other solid tumors. For these trials, Idera will provide IMO-2125. Idera is currently enrolling a Phase 3 ("ILLUMINATE-301") trial of intratumoral administration of IMO-2125 in combination with ipilimumab in patients with anti-PD-1 refractory metastatic melanoma.

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 three trials within the terms of this agreement are:
A Phase 1/2 open label study of intratumoral IMO-2125 in combination with intratumoral ipilimumab and IV nivolumab in a protocol open to multiple tumor types including non-small cell lung cancer ("NSCLC"), melanoma, squamous cell carcinoma of the head and neck and urothelial carcinoma. The principal investigator initiating this trial is Aurélien Marabelle, MD, PhD, Clinical Director of the Cancer Immunotherapy Program at Institut Gustave Roussy, Villejuif, France.

A Phase 2 study of intratumoral IMO-2125 in combination with IV pembrolizumab in patients with NSCLC. The principal investigator initiating this trial is Arafat Tfayli, MD, FRCP, Professor of Clinical Medicine, Director of Research, NK Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon.

A Phase 2 placebo controlled study of intradermal administration of IMO-2125 in patients with T3/T4 primary melanoma scheduled to undergo a combined re-excision and sentinel node biopsy procedure. The principal investigators initiating this trial are Bas Koster, MD, Fons van den Eertwegh MD, PhD, and Tanja de Gruijl, PhD, who is Professor of Translational Tumor Immunology and Co-Director of the Cancer Immunology Program at the VU University Medical Center, Cancer Center Amsterdam, The Netherlands.
"We are eager to expand our knowledge and understanding of the various cancer types and combinations in which IMO-2125 can play a significant role in improving outcomes beyond our current registrational focus with our ILLUMINATE 301 program," stated Joanna Horobin, M.B., Ch. B., Idera’s Chief Medical Officer. "We look forward to working with these investigators to provide the support they need to initiate these trials before the end of the year," said Shah Rahimian, MD, Idera’s Oncology Medical Lead. "
"We have long believed and understood that the mechanism for IMO-2125 has broad potential and plays a central role in IO combinations beyond PD-1 refractory melanoma and through this financial grant, we are able to help light the spark to further expand our ability to test this hypothesis in multiple tumor types with expert clinical investigators," stated Youssef El Zein, Managing Partner, Pillar Invest Corporation.