Positive Primary Analysis from the Phase 2 OPTIC Study of Ponatinib (Iclusig®) in Chronic Phase-Chronic Myeloid Leukemia (CP-CML) to be Presented at the 2021 ASCO Annual Meeting

On May 20, 2021 Incyte (Nasdaq:INCY) reported that data from the primary analysis of the Phase 2 OPTIC (Optimizing Ponatinib Treatment In CML) trial will be presented during an oral session (Abstract #7000) at the 2021 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting, held virtually June 4-8, 2021 (Press release, Incyte, MAY 20, 2021, View Source [SID1234580364]). The OPTIC trial – an ongoing, randomized, open-label study prospectively evaluating response-based dosing regimens of ponatinib (Iclusig) over a range of three starting doses (45mg, 30mg, 15mg) followed by dose reduction to 15mg with the aim of optimizing efficacy and safety in patients with chronic-phase chronic myeloid leukemia (CP-CML) who are resistant to prior tyrosine kinase inhibitor (TKI) therapy – met its primary endpoint.

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 trial, sponsored by Takeda and co-funded by Incyte, evaluated treatment in patients with resistant disease, with and without mutations. The results show that the optimal benefit-risk profile for ponatinib in patients with CP-CML was achieved with a starting dose of 45mg/day, and upon response (achieving ≤1% BCR-ABL1IS), dose reduction to 15mg/day. The results also suggest a clinically-manageable safety profile for ponatinib. Data from the OPTIC interim analysis (cutoff date of July 2019), which evaluated 216 patients with a median follow-up of 21 months, were previously reported. The primary analysis (cutoff date of May 2020) evaluated 283 patients with a median follow-up of 32 months. All patients in the OPTIC trial were evaluable for the primary endpoint at the time of this analysis.

"The OPTIC primary analysis reinforces the positive response that is achievable with ponatinib in appropriate patients with CP-CML, and the ability that response-based regimens have to maximize efficacy, while maintaining a manageable safety profile," said Luca Marini, M.D., Regional Vice President, Head of European Medical Affairs, Incyte. "The OPTIC trial provides additional confirmation on the optimization of ponatinib dosing and reinforces its role as a meaningful treatment option for patients."

Key findings from the OPTIC primary analysis include:

The maximum rates of ≤1% BCR-ABL1IS at 12 months, the primary endpoint, were achieved in the 45mg/day starting dose cohort (44.1%), and 73.3% of patients in this cohort maintained response with the dose reduction to 15mg/day. The 30mg/day and 15mg/day starting dose cohorts also demonstrated benefit (29.0% and 23.1% ≤1% BCR-ABL1IS at 12 months, respectively), especially in patients with less-resistant disease and with a T315I mutation.
Positive survival outcomes were estimated in all three arms, with an 89.3% 36-month overall survival (OS) probability anticipated for the 45mg starting dose cohort and 73.3% progression-free survival (PFS) anticipated for the same cohort.
This indicates that the dose-reduction strategy did not impact OS regardless of prior second-generation TKI resistance or the presence of BCR-ABL1 mutations.
Rates of arterial occlusive events (AOEs) observed at the time of primary analysis (6% overall and 9.6% in the 45mg starting dose cohort) suggest a clinically manageable safety profile.
Safety data include:
Among all study participants (N=283), the most common treatment-emergent adverse events (TEAEs) Grade 3 or greater were thrombocytopenia (27%), neutropenia (17%) and anemia (7%).
Reported AOEs were 10%, 5% and 3% for the 45mg, 30mg, 15mg/day starting dose cohorts, respectively. Grade 3 or greater AOEs were 5%, 5% and 3% for the 45mg, 30mg and 15mg/day starting dose cohorts, respectively.
Reported serious AOEs were 4%, 4% and 3% for the 45mg, 30mg, 15mg/day starting dose cohorts, respectively. There were four deaths related to AEs (two sudden deaths and two pneumonia).
"As a physician, I am pleased by the results of the OPTIC trial evaluating patients with resistant CP-CML, who are in need of additional options to improve outcomes," said Dr. Gianantonio Rosti, M.D., Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" (IRST S.r.l.) Istituto di Ricovero e Cura a Carattere Scientifico. "It is encouraging to see the positive benefit-risk profile that can be achieved with ponatinib through a response-based dosing regimen, therefore providing efficacy while also managing the risk for arterial occlusive events."

Incyte has an exclusive license from Takeda Pharmaceuticals International AG to commercialize ponatinib in the European Union and 29 other countries, including Switzerland, UK, Norway, Turkey, Israel and Russia. Iclusig is marketed in the U.S. by Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited.

About the OPTIC Trial

OPTIC (Optimizing Ponatinib Treatment In CML) is a randomized, dose-ranging Phase 2 trial designed to evaluate three starting doses (15mg, 30mg, 45mg) of ponatinib in patients with resistant chronic-phase chronic myeloid leukemia (CP-CML) or who had documented history of presence of T315I mutation after receiving any number of prior tyrosine kinase inhibitors (TKIs). Dose reduction at response occurred per study protocol. The trial is expected to inform the optimal use of ponatinib in these patients. The primary endpoint of the trial is achieving ≤1% BCR-ABL1 at 12 months. A total of 283 patients were enrolled at clinical sites around the world. Dose reduction at response occurred per study protocol.

For more information about the OPTIC study, please visit View Source

About CML

CML – a rare malignancy – is one of four main types of leukemia; it is a result of a genetic mutation that takes place in early, immature versions of myeloid cells, which form red blood cells, platelets and most types of white blood cells. Subsequently, an abnormal gene called BCR-ABL1 forms, turning the damaged cell into a CML cell. CML typically progresses slowly, but it can change into a fast-growing acute leukemia that is hard to treat.

About Ponatinib (Iclusig) Tablets

Ponatinib (Iclusig) targets not only native BCR-ABL but also its isoforms that carry mutations that confer resistance to treatment, including the T315I mutation, which has been associated with resistance to other approved TKIs.

In the EU, Iclusig is approved for the treatment of adult patients with chronic phase, accelerated phase or blast phase chronic myeloid leukemia (CML) who are resistant to dasatinib or nilotinib; who are intolerant to dasatinib or nilotinib and for whom subsequent treatment with imatinib is not clinically appropriate; or who have the T315I mutation, or the treatment of adult patients with Philadelphia-chromosome positive acute lymphoblastic leukemia (Ph+ ALL) who are resistant to dasatinib; who are intolerant to dasatinib and for whom subsequent treatment with imatinib is not clinically appropriate; or who have the T315I mutation.

Click here to view the Iclusig EU Summary of Medicinal Product Characteristics.

Incyte has an exclusive license from Takeda Pharmaceuticals International AG to commercialize ponatinib in the European Union and 29 other countries, including Switzerland, UK, Norway, Turkey, Israel and Russia. Iclusig is marketed in the U.S. by Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited

IconOVir Bio Announces Preclinical Proof-of-Concept Data for Proprietary Mutations Designed to Improve Selectivity of Novel Oncolytic Viruses, Including Lead Product Candidate ICVB-1042

On May 20, 2021 IconOVir Bio, Inc. (IconOVir), a preclinical-stage biotechnology company pioneering the next generation of oncolytic virus (OV) therapy to improve the treatment of patients with cancer, reported new preclinical data demonstrating proof-of-concept for a proprietary combination of two selectivity mutations, made to Adenovirus (Ad) proteins E1A and E4-ORF6/7, both of which are incorporated into its rationally designed OV product candidates (Press release, IconOVir Bio, MAY 20, 2021, View Source [SID1234580363]). The data, which were generated by IconOVir’s partners at the Salk Institute for Biological Studies, were shared in an abstract (e14543) titled "Highly selective and potent p16-CDK-RB-E2F targeted oncolytic virus therapies," which was included in the 2021 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting Proceedings, an online supplement to the Journal of Clinical Oncology.

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!

"IconOVir was founded on the belief that OVs can be rationally engineered to deliver deeper, more durable benefit to patients," said Mark McCamish, M.D., Ph.D., President and Chief Executive Officer of IconOVir. "Our proprietary platform enables rapid, iterative and combinatorial engineering, screening and optimization of next-generation OVs, enabling us to impart desirable attributes to novel viruses while still maintaining the qualities that make Ad an attractive OV backbone. In the case of ICVB-1042, we incorporate a series of mutations to optimize potency, selectivity and tumor tropism, and potentially enable both intratumoral and intravenous administration. The abstract shared at ASCO (Free ASCO Whitepaper) describes the first pair of mutations that we introduced into ICVB-1042, which improve tumor selectivity without impacting replicative efficiency or sacrificing lytic potency. We look forward to advancing ICVB-1042 into clinical studies in 2022, as we pursue our mission of curing cancer and restoring life to every patient, everywhere."

In normal cells, the p16/RB tumor suppressor pathway regulates E2F transcription factors, which activate critical genes for DNA replication. However, in nearly all solid tumors, one or more mutations in the p16-CDK-RB-E2Fpathway genes aberrantly activate E2F and are critical in driving uncontrolled tumor proliferation. While difficult to target with traditional therapeutic modalities, these tumor suppressor loss of function mutations represent ideal targets for driving durable anti-tumor responses.

Ad has evolved viral proteins, such as E1A, that bind to RB to activate E2F transcription of viral and cellular genes required for viral replication. On this basis, multiple first-generation Ad OV candidates have been engineered to contain RB binding domain mutations in the E1A gene in an effort to prevent E2F activation and replication in healthy cells. However, IconOVir’s partners at the Salk Institute have shown that another viral protein, E4-ORF6/7, activates E2F independently of E1A and drives virus replication in normal cells, even when the E1A gene has been mutated.

In the abstract published in the ASCO (Free ASCO Whitepaper) Annual Meetings Proceedings, the Salk Institute team describes data showing that combining E1A and E4-ORF6/7 mutations in an Ad OV yields an improved selectivity/efficacy profile over first-generation OVs engineered with E1A-RB mutations alone by conferring E2F-dependent tumor-selective replication without negatively impacting lytic potency in cancer cells. Furthermore, this high degree of tumor-selectivity is maintained when combined with additional mutations to enhance the lytic potency, tropism and systemic bioavailability of Ad OVs, as in IconOVir’s lead candidate, ICVB-1042.

"While OV-based therapies have delivered meaningful benefit to select patient populations, first-generation therapies are limited in their lytic potency, tumor selectivity, and systemic bioavailability, which has prevented the full realization of the modality’s potential," said Clodagh O’Shea, Ph.D., Wicklow Chair and Professor of Molecular and Cell Biology at the Salk Institute and Scientific Founder and Chair of the Scientific Advisory Board of IconOVir. "The data announced today demonstrate proof-of-concept for our belief that rationally designed OVs can overcome these historical challenges, by introducing mutations that enable viruses to specifically replicate in tumor cells, while leaving healthy cells unharmed. I look forward to collaborating with the IconOVir team as they advance ICVB-1042, the first rationally engineered OV to incorporate the dual E1A and E4-ORF6/7 mutation, into clinical development next year."

IconOVir’s lead product candidate, ICVB-1042, is derived from Ad, a common cold virus. In preclinical studies, ICVB-1042 has been shown to infect and kill a broad range of tumor cells, including head and neck, bladder, lung and breast, suggesting that it could have potential utility in a wide range of solid tumor indications. IconOVir is advancing ICVB-1042 through preclinical development, with plans to file an Investigational New Drug application with the U.S. Food and Drug Administration in the first half of 2022.

Heat Biologics Announces Promising Survival Data of HS-110 in Two Treatment Settings of Lung Cancer; Selected for Presentation at 2021 American Society of Clinical Oncology Annual Meeting

On May 20, 2021 Heat Biologics, Inc. (Nasdaq: HTBX), a clinical-stage biopharmaceutical company focused on developing first-in-class therapies to modulate the immune system, reported that the latest data from HS-110, the Company’s lead product, has been accepted for poster presentation at the 2021 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting, to be held virtually from June 4-8, 2021 (Press release, Heat Biologics, MAY 20, 2021, View Source [SID1234580362]). The ASCO (Free ASCO Whitepaper) Annual Meeting is the largest international conference to showcase the latest advancements in oncology.

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!

Abstract Title: Interim results of viagenpumatucel-L (HS-110) plus nivolumab in previously treated patients (pts) with advanced non-small cell lung cancer (NSCLC) in two treatment settings

Session Title: Poster Session: Lung Cancer—Non-Small Cell Metastatic

Abstract Number: 9100

Lead Author and Presenter: Roger B. Cohen, MD, Perelman School of Medicine at the University of Pennsylvania

According to ASCO (Free ASCO Whitepaper), more than 5,400 abstracts were reviewed for the 2021 ASCO (Free ASCO Whitepaper) Annual Meeting. Additional information about the conference may be accessed at View Source

About HS-110
HS-110 is a first-in-class, off-the-shelf, allogeneic cell therapy designed to utilize gp96 for immune activation against multiple tumor testis antigens. Phase 2 trial of HS-110 in combination with Bristol-Myers Squibb’s (BMS) OPDIVO (nivolumab) has completed enrollment in patients with incurable or metastatic NSCLC. OPDIVO is a programmed death-1 immune checkpoint inhibitor (PD-1 inhibitor). HS-110 has broad potential for providing multiple treatment options to NSCLC patients in combination with a PD-1 inhibitor. Positive interim survival data has been demonstrated in two distinct treatment settings in previously treated NSCLC patients who have not been treated with PD-1 inhibitor or programmed death-ligand 1 inhibitor (PD-L1 inhibitor) as well as patients who have progressed during or after previous treatment with a PD-1 or PD-L1 [PD-(L)1] inhibitor. Combination of HS-110 and PD-(L)1 therapies may confer additional survival benefit.

H3 Biomedicine Announces Presentation of Four Abstracts at the 2021 American Society of Clinical Oncology (ASCO) Annual Meeting

On May 20, 2021 H3 Biomedicine Inc. (H3), a U.S.-based precision medicine research & development subsidiary of Eisai Co., Ltd., reported the presentation of four posters providing updated investigational data on its H3B-6545 clinical program for breast cancer and its H3B-6527 clinical program for hepatocellular carcinoma at the 2021 American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) Annual Meeting, being held virtually June 4–8, 2021 (Press release, H3 Biomedicine, MAY 20, 2021, View Source [SID1234580361]).

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 continue to be encouraged by the insights gained through our evaluation of genomically and clinically defined patient populations in these two oncology programs in areas of tremendous unmet medical need," said Antonio Gualberto, MD, PhD, Chief Medical Officer of H3. "We look forward to further highlighting our innovative, data-driven approach to precision oncology research and to showcasing the potential clinical benefit of our product candidates for patients with cancer at ASCO (Free ASCO Whitepaper) 2021."

The investigational clinical data abstracts published online today for the H3B-6545 and H3B-6527 studies reflect data as of March 31, 2021 and Jan 04, 2021, respectively. Updated results from both studies will be presented at ASCO (Free ASCO Whitepaper).

H3’s ASCO (Free ASCO Whitepaper) abstract titles are as follows:

H3B-6545
Abstract Number: 1018
Title: Phase I/II Study of H3B-6545, a Novel Selective Estrogen Receptor Covalent Antagonist (SERCA), in Estrogen Receptor Positive (ER+), Human Epidermal Growth Factor Receptor 2 Negative (HER2-) Advanced Breast Cancer
Poster Discussion Session: Breast Cancer – Metastatic
Presenter: Erika P. Hamilton, Sarah Cannon Research Institute, Tennessee Oncology

Abstract Number: e13025
Title: Phase 1b Study of H3B-6545 in Combination with Palbociclib in Women with Metastatic Estrogen Receptor–positive (ER+), Human Epidermal Growth Factor Receptor 2 (HER2)-negative Breast Cancer
Poster Session: Breast Cancer – Metastatic (Online Publication Only)
Presenter: Stephen R.D. Johnston

Abstract Number: e13022
Title: Relative Bioavailability of H3B-6545 Tablets vs Capsules and Drug-Drug Interaction between H3B-6545 and Pantoprazole
Poster Session: Breast Cancer – Metastatic (Online Publication Only)
Presenter: Alan Xiao

H3B-6527
Abstract Number: 4090
Title: Phase 1 Study of H3B-6527 in Hepatocellular Carcinoma (HCC) or Intrahepatic Cholangiocarcinoma (ICC)
Poster Session: Gastrointestinal Cancer – Gastroesophageal, Pancreatic and Hepatobiliary
Presenter: Teresa Macarulla

ABOUT H3B-6545
Estrogen receptor alpha (ERα) plays an important oncogenic role in the genesis and progression of luminal breast cancers and historically has been a target of endocrine therapies. However, recently, hotspot mutations in ERα have been detected in nearly 30% of endocrine-therapy resistant metastases. Functional studies have shown that these ERα mutations can confer ligand-independent activation of the ERα pathway and can promote partial resistance to existing classes of ER-directed therapies. H3B-6545, a first-in-class small molecule selective estrogen receptor covalent antagonist (SERCA) demonstrates activity in tumor models that harbor wild-type or mutant ERα. H3B-6545 activity against ERα mutants resistant to standard therapy provides an opportunity to target a currently unmet medical need both as a single agent and in combination with other breast cancer therapies.

ABOUT H3B-6527
Receptor tyrosine kinases (RTKs) can be dysregulated in cancer cells and can frequently promote abnormally rapid tumor growth and development. Hepatocellular carcinoma (HCC) can be driven in this way by hyperactivation of the fibroblast growth factor 19 (FGF19)/fibroblast growth factor receptor 4 (FGFR4) pathway. H3B-6527 is a selective, orally bioavailable, and covalent inhibitor of FGFR4 that has demonstrated tumor regression in several preclinical models of HCC. H3B-6527 is being tested specifically in patients with FGFR4-dysregulated advanced HCC.

The abstracts discuss investigational uses of agents in development and are not intended to convey conclusions about efficacy or safety. There is no guarantee that such investigational agents will successfully complete clinical development or gain health authority approval.

GSK announces sale of stake in Innoviva Inc

On May 20, 2021 GlaxoSmithKline reported that it has agreed with Innoviva, Inc ("Innoviva") – a royalty management company – to sell all of its approximately 32 million shares of common stock of Innoviva back to Innoviva at a price of $12.25 per share, raising gross proceeds of approximately $392 million (Press release, GlaxoSmithKline, MAY 20, 2021, View Source [SID1234580360]). Following settlement of the transaction, GSK will no longer hold any Innoviva stock.

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 terms of GSK’s long-standing and successful respiratory collaboration with Innoviva remain unchanged. Under this agreement GSK pays royalties to Innoviva on TRELEGY ELLIPTA, RELVAR/BREO ELLIPTA and ANORO ELLIPTA. In 2020, TRELEGY ELLIPTA sales were £819m (+59% CER (constant exchange rates)); RELVAR/BREO ELLIPTA sales were £1.1bn (+17% CER) and ANORO ELLIPTA sales were £547m (+8% CER).

This equity disposal releases capital to enable GSK to make further investment behind the Group’s strategic priorities.

Innoviva’s most recent closing share price was $12.29. The five-day volume weighted average price was $12.67.