New data at ASH underscore the potential for durable, clinically important responses with momelotinib for myelofibrosis patients

On December 11, 2022 GSK plc (LSE/NYSE: GSK) reported new 48-week data from the MOMENTUM phase III trial that showed a majority of patients treated with investigational momelotinib maintained their responses across key clinical measures including Total Symptom Score (TSS), Transfusion Independence (TI) rate, and Splenic Response Rate (SRR) in myelofibrosis patients previously treated with an approved Janus kinase (JAK) inhibitor (Press release, GlaxoSmithKline, DEC 11, 2022, View Source [SID1234625112]). Additionally, new analyses from MOMENTUM showed that TI response with momelotinib at week 24 was associated with overall survival. These data were presented at the 64th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting and Exposition (10-13 December) in New Orleans.

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!

Hesham Abdullah, Senior Vice President, Global Head of Oncology Development, GSK, said: "Myelofibrosis patients have significant medical needs including progressive anaemia resulting from the disease and often exacerbated by currently approved treatments. The data presented today reinforce the potential of momelotinib as a treatment option with a favourable impact on myelofibrosis symptoms and spleen volume, as well as on blood transfusions due to anaemia."

MOMENTUM is a global, randomised, double-blind phase III clinical trial of momelotinib (MMB) versus danazol (DAN) in patients with myelofibrosis who were symptomatic and anaemic and had been previously treated with an approved JAK inhibitor. The study was designed to evaluate the safety and efficacy of momelotinib for the treatment and reduction of the key manifestations of the disease: constitutional symptoms, blood transfusions (due to anaemia) and enlarged spleen. Patients were randomised at 2:1 to receive either momelotinib or danazol (n=130 and n=65, respectively). After 24 weeks of treatment, patients on danazol were allowed to crossover to receive momelotinib. Early crossover to momelotinib was available for confirmed splenic progression.

Primary analysis at week 24 met the primary endpoint of TSS reduction of ≥50% over the 28 days immediately before the end of week 24 compared to baseline TSS, using the Myelofibrosis Symptom Assessment Form. It also met key secondary endpoints including TI rate for ≥12 weeks immediately before the end of week 24 with haemoglobin levels ≥ 8 g/dL and SRR based on splenic volume reduction of ≥35% at week 24 from baseline.

48-week follow-up data from MOMENTUM presented at ASH (Free ASH Whitepaper) demonstrated the following (oral presentation #627):

TSS response at week 24 and maintained below baseline through week 48

TI response at week 24 and maintained response through week 48

SRR response at week 24 and maintained below baseline through week 48

MMB

31/32 (97%)*

36/40 (90%)

24/24 (100%)***

DAN with crossover to MMB at week 24

6/6 (100%)**

10/13 (77%)

2/2 (100%)

*12 of 61 (20%) non-responding patients who received momelotinib at week 24 had achieved a TSS response at week 48

**10 of 35 (29%) non-responding patients who received danazol and crossed over to momelotinib at week 24 achieved a new TSS response at week 48

***data available for 24 of 30 patients who received momelotinib and achieved splenic response at 24 weeks

In MOMENTUM, the most common Grade 3 or greater treatment emergent adverse events in the open-label period, similar to the double-blind period, were thrombocytopenia (9% for the continuous momelotinib treatment arm and 15% for the danazol to momelotinib treatment arm) and anaemia (9% for the continuous momelotinib treatment arm and 2% for the danazol to momelotinib treatment arm). Additionally, these efficacy and safety results in patients with thrombocytopenia were consistent with the overall population.

An additional analysis from the MOMENTUM clinical trial evaluated the impact of TI response on overall survival (abstract #3028). As previously presented, patients receiving treatment with momelotinib were more likely to achieve transfusion independence during the study period than patients treated with danazol (TI response at week 24 of 31% and 20% for the momelotinib and danazol arms, respectively; non-inferiority p=0.0064). Momelotinib patients were also less likely to require a transfusion during the study period (35% of momelotinib patients had zero units transfused compared to 17% of danazol patients; odds ratio=2.7; p=0.0107), and more likely to reduce transfusion burden. Data presented at ASH (Free ASH Whitepaper) with additional survival follow up suggests that TI response with momelotinib at week 24 is associated with overall survival (HR=0.27 for TI vs. non-TI; CI 95% 0.09, 0.80) compared to patients who were not TI.

A New Drug Application and Marketing Authorisation Application for momelotinib is currently under review with the US Food and Drug Administration (FDA) and European Medicines Agency (EMA), respectively. Momelotinib is not currently approved in any market.

About momelotinib
Momelotinib is an investigational oral treatment for myelofibrosis. Momelotinib not only inhibits the Janus kinase (JAK) 1 signalling pathway and JAK2 signalling pathways, but also activin A receptor type I (ACVR1).[i],[ii],[iii],[iv] Inhibition of JAK1 and JAK2 may improve constitutional symptoms and splenomegaly.[i],[ii],[iv] Additionally, direct inhibition of ACVR1 leads to a decrease in circulating hepcidin, which is often elevated in myelofibrosis and contributes to anaemia.[i],[ii],[iii],[iv]

About myelofibrosis
Myelofibrosis is a rare and potentially fatal cancer characterized by the build-up of excessive scar tissue in the bone marrow, which interferes with the production of healthy blood cells and can lead to: severe low blood counts, including anaemia and thrombocytopenia; constitutional symptoms such as weakness and fatigue; and splenomegaly or an enlarged spleen.[v],[vi],[vii] Myelofibrosis affects about 1 in 500,000 people worldwide.[viii] At diagnosis, approximately 40% of patients are anaemic and nearly all myelofibrosis patients will eventually develop anaemia.[ix],[x],[xi] Patients will often require blood transfusions, and more than 30% will discontinue treatment due to anaemia.[xii] Anaemia and transfusion dependence are strongly correlated with poor prognosis, reduced quality of life and shortened survival

Poseida Therapeutics Presents Preclinical Data from P-FVIII-101 Gene Therapy for Hemophilia A at the 64th ASH Annual Meeting & Exposition

On December 11, 2022 Poseida Therapeutics, Inc. (Nasdaq: PSTX), a clinical-stage cell and gene therapy company advancing a new class of treatments for patients with cancer and rare diseases, reported that the Company will present preclinical data from its P-FVIII-101 gene therapy program, partnered with Takeda, at the 2022 American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting being held in New Orleans and virtually December 10–13, 2022 (Press release, Poseida Therapeutics, DEC 11, 2022, View Source [SID1234625071]). The data establish preclinical proof of principle for the treatment of Hemophilia A using P-FVIII-101, a non-viral liver-directed gene therapy utilizing Poseida’s Super piggyBac delivery system, which could potentially lead to a functional cure.

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!

Poseida Therapeutics (PRNewsfoto/Poseida Therapeutics, Inc.)

"We are very excited by these new P-FVIII-101 data, which demonstrate normalization of FVIII levels in an animal model of Hemophilia A," said Brent Warner, President, Gene Therapy at Poseida Therapeutics. "Most importantly, we have demonstrated the use of a fully non-viral gene therapy to address the underlying cause of Hemophilia A, providing key preclinical proof of principle for our program. We look forward to our continued work on this program together with our partner, Takeda."

Details of the oral presentation are as follows:

Title: Sustained Factor VIII Activity Following Single Dose of Non-Viral Integrating Gene Therapy
Presenter: Brian Truong, Ph.D.
Presentation Date and Time: Today, December 11, 2022 at 10:15 AM CT
Session Name: 321. Coagulation and Fibrinolysis: Basic and Translational
Publication Number: 400
Location: Ernest N. Morial Convention Center, 293-294

P-FVIII-101 utilizes the Company’s non-viral, nanoparticle-based delivery system together with SPB, which enables increased transgene cargo capacity, stable integration into the genome, potential for re-dosing, and potentially simpler manufacturing processes. The data to be presented show that P-FVIII-101 achieved and sustained normalized (>50%) hFVIII activity following a single dose and delivered therapeutic FVIII activity in mice following single and repeat doses, indicating the potential for dose titration. Durable responses were observed following a single dose reported over the study period of seven months. The data support that with SPB the therapeutic transgene expression cassette can be stably integrated into the genome of liver cells and provide consistent and durable therapeutic activity.

"Although gene therapy has the potential to deliver functional cures for Hemophilia A, current approaches face challenges – both with durability and the ability to re-dose – and are not appropriate for use in juvenile patients," said Denise Sabatino, Ph.D., Research Associate Professor of Pediatrics at the Perelman School of Medicine at the University of Pennsylvania and Children’s Hospital of Philadelphia (CHOP) and an author on the oral presentation. "The data being presented today show that P-FVIII-101 has the potential to correct a deficiency in FVIII to near normal levels in juvenile mice, providing a path forward for a more tolerable, durable treatment for Hemophilia A in pediatric patients. Current treatment options are not curative and require lifelong treatment, and P-FVIII-101 may have the potential to significantly improve outcomes for people with Hemophilia A."

In October 2021, Poseida announced that it had entered into a research collaboration and exclusive license agreement with Takeda to utilize the Company’s proprietary genetic engineering platform technologies for the research and development of gene therapies, including P-FVIII-101. The companies plan to continue preclinical studies to advance the program toward an Investigational New Drug (IND) application.

About P-FVIII-101
P-FVIII-101 is a liver-directed gene therapy partnered with Takeda combining Poseida’s Super piggyBac platform and nanoparticle delivery technologies for the in vivo treatment of Hemophilia A. Hemophilia A is a bleeding disorder caused by a deficiency in Factor VIII production with a high unmet need. P-FVIII-101 utilizes the piggyBac gene integration system delivered via lipid nanoparticle, which has demonstrated stable and sustained Factor VIII expression in animal models.

HARPOON THERAPEUTICS PRESENTS UPDATED INTERIM RESULTS AT ASH 2022 FOR NOVEL T CELL ENGAGER HPN217 IN RELAPSED/REFRACTORY MULTIPLE MYELOMA

On December 11, 2022 Harpoon Therapeutics, Inc. (Nasdaq: HARP), a clinical-stage immuno-oncology company developing novel T cell engagers, reported updated interim data from its Phase 1 clinical trial evaluating single-agent HPN217 in relapsed/refractory multiple myeloma (RRMM) in a poster presentation at the 64th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting and Exposition being held in person and virtually in New Orleans (Press release, Harpoon Therapeutics, DEC 11, 2022, View Source [SID1234625070]). HPN217 targets B-cell maturation antigen (BCMA) and is based on Harpoon’s proprietary Tri-specific T cell Activating Construct (TriTAC) platform designed to recruit a patient’s own immune cells to kill tumor 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!

The interim results, as of the data cut-off date of October 17, 2022, showed that HPN217 demonstrated continued evidence of clinical activity and a tolerable safety profile in heavily pre-treated patients with RRMM (62 patients treated across fixed dose and step dose regimens). HPN217 was active across a wide dose range (2.15 to 24 mg), with 77% (10/13) ORR observed across the highest step doses (12 and 24 mg). A majority of responders had decreases in the serum BCMA biomarker (sBCMA, a marker correlated with disease prognosis) by week two of treatment. Additionally, 86% (18/21) of responders remain on study treatment with sustained response, with many responders on treatment for over a year. Three patients in the study were evaluated for minimal residual disease (MRD), and all three were MRD negative (<10-5). sBCMA remained undetectable at 9 months in many responders who achieved very good partial response (VGPR) or better.

Low-grade CRS occurred in 29% of patients across the highest step dose regimens (12% Grade 1 and 18% Grade 2) and was seen primarily in the earliest doses. No Grade 3 or higher CRS or any immune effector cell associated neurotoxicity syndrome (ICANS) events have been observed.

"The encouraging initial clinical activity with deepening and durable responses observed in patients who have received multiple prior lines of therapy, combined with a generally well-tolerated safety profile, suggest the investigational T cell engager HPN217 may offer meaningful clinical benefits for patients with relapsed/refractory multiple myeloma," said Al-Ola A. Abdallah, M.D., of University of Kansas Medical Center, a Principal Investigator in this study. "I look forward to continuing to study this promising drug candidate in these patients with advanced disease for whom there remains a significant unmet need for new treatment options."

"These data provide further validation of our proprietary TriTAC T cell engager platform, demonstrating robust clinical activity for HPN217 at higher doses, while maintaining tolerability in this heavily refractory patient population," said Luke Walker, M.D., Chief Medical Officer of Harpoon Therapeutics. "These data support our continued clinical development efforts, and we look forward to continuing dose optimization with ongoing patient enrollment in the Phase 1 trial expected to reach completion in the first half of 2023."

For more details about the ASH (Free ASH Whitepaper) Annual Meeting, please visit: View Source

The poster (publication #3240) will be available on Harpoon’s website following today’s presentation.

Conference Call and Webcast Details

Harpoon’s management will host a live call/webcast on Monday, December 12, 2022, at 4:30 ET/3:30 CT/1:30 PT, to review the interim results of its Phase 1 HPN217 clinical program and provide an update on other pipeline programs. The live call may be accessed by dialing 1-877-407-9039 for domestic callers and 1-201-689-8470 for international callers with conference ID code number 13734677. A live webcast of the call will be available from the Events and Presentations section of Harpoon’s website here and will be archived there shortly after the live event.

About HPN217

HPN217 targets B-cell maturation antigen (BCMA) and is based on Harpoon’s proprietary Tri-specific T cell Activating Construct (TriTAC) platform designed to recruit a patient’s own immune cells to kill tumor cells.

In November 2019, Harpoon Therapeutics and AbbVie announced a licensing agreement and option to advance HPN217 and expand an existing discovery collaboration. Under the terms of the agreement, AbbVie may exercise its option to license HPN217 after completion of the Phase 1 clinical trial.

In March 2022, the FDA granted Fast Track designation to HPN217, underscoring its potential to address a serious unmet medical need for patients with relapsed, refractory multiple myeloma.

About the HPN217 Clinical Trial

HPN217 is being evaluated in an ongoing Phase 1, multicenter, open-label dose escalation study designed to evaluate safety, tolerability, pharmacokinetics (PK) and clinical activity in patients with relapsed/refractory multiple myeloma who have had at least three prior systemic treatments, including a proteasome inhibitor, an immunomodulatory drug and an anti-CD38 antibody, including patients with prior exposure to BCMA therapy. Primary objectives are characterization of safety, tolerability, PK and determination of the recommended Phase 2 dose.

As of the cutoff date on October 17, 2022, maximum tolerated dose has not yet been reached in the step-dose regimen. Assessment of the Q2 cohort dosing schedule is ongoing.

For additional information about the trial, please visit www.clinicaltrials.gov using the identifier NCT04184050.

Aptose Announces Updated Clinical Responses, Breadth of Activity, and Safety Across Four Dose Levels of Tuspetinib in Difficult-to-Treat Acute Myeloid Leukemia Populations

On December 11, 2022 Aptose Biosciences Inc. ("Aptose") (NASDAQ: APTO, TSX: APS) reported a clinical update of its lead oral myeloid kinome inhibitor, tuspetinib (formerly HM43239), as responses continue to emerge from a Phase 1/2 trial, and from its oral, dual lymphoid and myeloid kinase inhibitor, luxeptinib (formerly CG-806) in an ongoing Phase 1a/b trial (Press release, Aptose Biosciences, DEC 11, 2022, View Source [SID1234625069]).

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!

Tuspetinib, a once daily oral agent designed to target FLT3, SYK, and JAK kinases but avoid targets that drive toxicities, safely delivered complete remissions (CR/CRh/CRi/CRp) as a monotherapy across four dose levels (40mg, 80mg, 120mg, and 160mg) in acute myeloid leukemia (AML) patients that previously had been failed by chemotherapy, Bcl-2 inhibitors, hypomethylating agents, competitor FLT3 inhibitors, and hematopoietic stem cell transplants. Data are being presented at the 2022 American Society of Hematology (ASH) (Free ASH Whitepaper) annual meeting by lead investigator Naval G. Daver, M.D., Associate Professor in the Department of Leukemia at MD Anderson Cancer Center, showing tuspetinib delivers single agent responses in very ill and heavily pretreated relapsed or refractory AML patients of mutationally-defined populations, including those with AML harboring wild-type FLT3, ITD or TKD mutated FLT3, or mutated forms of NPM1, MLL,TP53, NRAS, KRAS, DNMT3A, RUNX1, various slicing factors, and other genes. "The welcome blend of safety and breadth of activity seen with tuspetinib in AML patients makes this an ideal candidate for combination therapy," remarked Dr. Daver.

As of October 6, 2022, 60 heavily pretreated relapsed/refractory AML patients were enrolled at multiple centers and treated at doses escalating from 20 mg to 200 mg, with further dose exploration at the 40 mg, 80 mg, 120 mg and 160 mg dose levels. Prior to Aptose licensing tuspetinib, Hanmi Pharmaceutical Company demonstrated complete remissions at the 80 mg dose level. As of January 1, 2022, Aptose assumed control of clinical trial activities and has demonstrated additional complete remissions at the 120 mg, 160 mg, and now the 40 mg dose levels. Many responders were bridged successfully to hematopoietic stem cell transplant (HSCT), while others not eligible for HSCT remained on tuspetinib with a durable response and no drug related myelosuppression even after months of continuous dosing.

The noteworthy safety and potency profile position tuspetinib, in both FLT3 mutated (FLT3+) and wildtype (FLT3-WT) AML patients, potentially to become the kinase inhibitor of choice to combine with venetoclax and hypomethylating agents to deliver high response rates without exacerbated myelosuppression or life-threatening toxicities and potentially to become the preferred agent for maintenance therapy to prevent relapse after HSCT or drug-induced complete remissions. Such roles can define the ultimate therapeutic success for patients and commercial success for tuspetinib.

"While the superior target and safety profile, and proven breadth of activity of tuspetinib compared to competitive compounds in development advocate for tuspetinib to participate in broader and more sizable commercial markets," said William G. Rice, Ph.D., Chairman, President, and Chief Executive Officer, "responses generated by tuspetinib in mutationally-defined populations of high unmet need may also provide accelerated approval opportunities."

Highlights of Updated Tuspetinib Data

In addition to 5 CRc and 1 PR reported at ASH (Free ASH Whitepaper) 2021, 4 new CRc and 3 new PR have been generated thus far during 2022.
New responses during 2022 were achieved with 160 mg,120 mg, 80 mg, and 40 mg
Among efficacy evaluable patients treated with 80 mg, 120 mg, or 160mg, the following response rates were achieved:
FLT3+ 8 of 21 (38.1%)
FLT3-WT 4 of 21 (19%), plus additional CRi since data cutoff date
FLT3+ with prior FLT3i 3 of 11 (27.3%)
FLT3+/NPM1+ 4 of 6 (66.7%)
FLT3+/NPM1+/DNMT3A+ 3 of 4 (75%)
N/K-RAS+ 3 of 8 (37.5%)
Significant bone marrow leukemic blast reductions were observed broadly in FLT3+ and FLT3 wildtype patients across multiple dose levels, comparable to reported gilteritinib data, but in more heavily pre-treated relapsed and refractory AML patients (waterfall chart available on Aptose website).
Vignettes of patient experiences highlight the potency and breadth of tuspetinib to deliver complete remissions among several mutationally-defined populations with a diversity of adverse mutations.
Tuspetinib continued to show a favorable safety profile with only mild AEs and no DLTs up to 160 mg per day, and no drug discontinuations from drug related toxicity.
No drug related SAE, drug related deaths, differentiation syndrome
No drug related AE of QT prolongation
No DLT through 160 mg level – one DLT of muscle weakness at 200 mg (not rhabdomyolysis)
No observed muscle destruction – no AE of elevated creatinine phosphokinase (CPK)
Avoids many of the typical toxicities observed with other tyrosine kinase inhibitors
Aptose has identified a safe therapeutic range with a broad therapeutic window, spanning the dose levels of 40, 80, 120 and 160 milligrams.
For the APTIVATE expansion trial that has initiated patient enrollment, Aptose has selected 120 mg as the initiating single agent expansion dose and 80 mg as the initiating dose selected for combination with venetoclax. The trial is designed to confirm activity through patient enrichment of specific mutationally defined AML populations, including FLT3-mutant patients who have been failed by a prior FLT3 inhibitor, as supported by fast-track designation and a significant response rate to date.
More detail on the data is available on the presentations page of the Aptose website here.

Aptose also provided an update of the luxeptinib clinical program:

Luxeptinib Key Highlights

Luxeptinib is an oral, first-in-class FLT3 and BTK kinase inhibitor in Phase 1 a/b clinical studies for the treatment of myeloid hematologic malignancies. This small molecule demonstrates potent inhibition of wild type and all mutant forms of FLT3 (including internal tandem duplication, or ITD, and mutations of the receptor tyrosine kinase domain and gatekeeper region) and cures animals of AML in the absence of toxicity in murine leukemia models.

Just recently, a CR was achieved with a diffuse large B-cell lymphoma (DLBCL) patient at the end of Cycle 22 with 900mg BID of the original G1 formulation. Previously, an MRD-negative CR was reported with a R/R AML patient receiving 450mg BID of the original G1 formulation. Collectively, these findings demonstrate luxeptinib is active against AML as well as lymphoid malignancies.
The original G1 formulation of luxeptinib was hampered by poor absorption. The new "G3" formulation was designed and developed for more rapid absorption (early Tmax), more efficient absorption (use lower doses), longer retention (longer t1/2), and greater accumulation (higher steady state levels).
The new G3 formulation this year was tested as a single dose in 20 patients from a Phase 1 clinical program of luxeptinib. Modeling of the pharmacokinetic (PK) properties of G3 predicts steady-state plasma exposure from continuous dosing with 50 mg of G3 (every 12 hours, Q12h) should be comparable to that of 900 mg of the original G1 formulation Q12h, representing up to an 18-fold improvement in bioavailability with G3.
Aptose recently announced dosing of the first patient with continuous dosing of the G3 formulation (50 mg BID) in an ongoing Phase 1a/b clinical trial in patients with relapsed or refractory AML. A second patient now has initiated continuous dosing with the G3 formulation.
The G3 formulation may result in greater exposures of luxeptinib and additional responses in these difficult-to-treat patient populations.
Aptose expects that 9-15 patients will determine if G3 is safe and achieves desired exposures to deliver clinical responses.
As announced prior, Aptose will be holding a conference call and data review today:

Aptose Corporate Update Details

Date & Time: Sunday, Dec 11, 2022, 10:00 AM EST

Participant Webcast Link: Link

Participant Dial-in:

Toll Free Investors Dial: 1-877-407-9039

Toll/International Investors Dial: 1-201-689-8470

Conference ID: 13734698

The slides will be available on Aptose’s website here and the webcast of the presentation will be archived shortly after the conclusion of the event at the link above.

Interim Data From Phase III Study Presented at ASH 2022 Show Hemlibra (emicizumab-kxwh) Achieved Meaningful Bleed Control in Infants From Birth

On December 11, 2022 Genentech, a member of the Roche Group (SIX: RO, ROG; OTCQX: RHHBY), reported interim results from the Phase III HAVEN 7 study (Press release, Genentech, DEC 11, 2022, View Source [SID1234625056]). The study shows Hemlibra (emicizumab-kxwh) achieved meaningful bleed control with a favorable safety profile in infants (up to 12 months) with severe hemophilia A without factor VIII inhibitors: 77.8% of participants did not have any bleeds that required treatment and 42.6% did not have any treated or untreated bleeds at all. These results help support the use of Hemlibra in this population, in which it is already approved in many countries around the world. The new data were presented at the 64th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting and Exposition taking place in New Orleans from December 10-13, 2022.

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!

"Hemophilia can substantially reduce quality of life for those affected, starting at infancy, which is especially distressing for parents and caregivers. We continue to explore Hemlibra’s potential benefits to a broad range of people with hemophilia A."

Tweet this
The burden of severe hemophilia A in infants and on their parents and caregivers is significant. The World Federation of Hemophilia treatment guidelines consider the standard of care in hemophilia to be regular prophylaxis initiated at a young age, as studies have shown that early prophylaxis improves long-term outcomes, while reducing the risk of intracranial hemorrhage. However, for many infants with hemophilia A, prophylaxis is not started until after the first year of life because of the high treatment burden. Hemlibra provides a flexible treatment option that can be administered subcutaneously from birth at different dosing frequencies.

"These initial results support the benefit of starting Hemlibra from birth given that early preventative treatment is essential in infants," said Levi Garraway, M.D., Ph.D., chief medical officer and head of Global Product Development. "Hemophilia can substantially reduce quality of life for those affected, starting at infancy, which is especially distressing for parents and caregivers. We continue to explore Hemlibra’s potential benefits to a broad range of people with hemophilia A."

HAVEN 7 is a Phase III, multi-center, open-label study evaluating the efficacy, safety, pharmacokinetics and pharmacodynamics of Hemlibra in infants with severe hemophilia A without factor VIII inhibitors. The results of this interim analysis, which included data from 54 participants, showed that 77.8% of participants (n=42) did not have any bleeds which required treatment, while 42.6% (n=23) did not have any treated or untreated bleeds at all. There were no treated spontaneous bleeds in any participants, and all treated bleeds were traumatic. A total of 77 bleeds occurred in 31 participants (57.4%); 88.3% were traumatic. Mean model-based annualized bleeding rate (ABR) at the time of interim analysis was 0.4 (95% CI: 0.23–0.65) for treated bleeds.

Hemlibra’s safety profile was consistent with previous studies, with no new safety signals observed. Nine people (16.7%) reported a Hemlibra-related adverse event (AE), all of which were local injection site reactions. Eight participants (14.8%) reported 12 serious AEs, unrelated to Hemlibra. There were no deaths, thromboembolic events or cases of thrombotic microangiopathy, reinforcing Hemlibra’s favorable safety profile. No intracranial hemorrhages occurred.

Primary analysis will be conducted at 52 weeks. The study also has an additional seven-year follow-up period to collect long-term data such as safety and joint health outcomes, further building upon our understanding of the benefit of Hemlibra in this population.

EUHASS Database and ATHN 7 Study

Genentech also presented data from the European Haemophilia Safety Surveillance (EUHASS) database and the prospective observational ATHN 7 study at ASH (Free ASH Whitepaper) 2022. Data from EUHASS, which collects real-world safety data on treatments for inherited disorders, showed the safety profile of Hemlibra in people with hemophilia A was favorable and consistent with clinical trial data. Data from ATHN 7, exploring the efficacy of Hemlibra in women with hemophilia A, showed two of the three female participants had no bleeds; the third had one treated bleed associated with a dental procedure and one untreated bleed associated with menses. Ongoing evaluation is vital to further understand the safety and efficacy profile of Hemlibra in this rare and under-represented population.

Hemlibra is approved as a treatment for people with hemophilia A with factor VIII inhibitors in more than 110 countries worldwide, and for people without factor VIII inhibitors in more than 100 countries worldwide. It has been studied in one of the largest clinical trial programs in people with hemophilia A with and without factor VIII inhibitors, including eight Phase III studies.

About Hemlibra

Hemlibra is a bispecific factor IXa- and factor X-directed antibody. It is designed to bring together factor IXa and factor X, proteins required to activate the natural coagulation cascade and restore the blood clotting process for hemophilia A patients. Hemlibra is a prophylactic (preventative) treatment that can be administered by an injection of a ready-to-use solution under the skin (subcutaneously) once weekly, every two weeks or every four weeks. Hemlibra was created by Chugai Pharmaceutical Co., Ltd. and is being co-developed globally by Chugai, Roche and Genentech.

Hemlibra U.S. Indication

Hemlibra is a prescription medicine used for routine prophylaxis to prevent or reduce the frequency of bleeding episodes in adults and children, ages newborn and older, with hemophilia A with or without factor VIII inhibitors.

Important Safety Information

What is the most important information to know about Hemlibra?

Hemlibra increases the potential for blood to clot. People who use activated prothrombin complex concentrate (aPCC; Feiba) to treat breakthrough bleeds while taking Hemlibra may be at risk of serious side effects related to blood clots.

These serious side effects include:

Thrombotic microangiopathy (TMA), a condition involving blood clots and injury to small blood vessels that may cause harm to one’s kidneys, brain, and other organs
Blood clots (thrombotic events), which may form in blood vessels in the arm, leg, lung, or head
Patients should talk to their doctor about the signs and symptoms of these serious side effects, which can include

Confusion
Stomach, chest, or back pain
Weakness
Nausea or vomiting
Swelling, pain, or redness
Feeling sick or faint
Decreased urination
Swelling of arms and legs
Yellowing of skin and eyes
Eye pain, swelling, or trouble seeing
Fast heart rate
Numbness in your face
Headache
Shortness of breath
Coughing up blood
If patients experience any of these symptoms during or after treatment with Hemlibra, they should get medical help right away.

Patients should carefully follow their healthcare provider’s instructions regarding when to use an on demand bypassing agent or factor VIII, and the dose and schedule to use for breakthrough bleed treatment. If aPCC (Feiba) is needed, patients should talk to their healthcare provider in case they feel they need more than 100 U/kg of aPCC (Feiba) total.

Patients’ bodies may make antibodies against Hemlibra, which may stop Hemlibra from working properly. Patients should contact their healthcare provider immediately if they notice that Hemlibra has stopped working for them (e.g., increase in bleeds).

The most common side effects of Hemlibra include: injection site reactions (redness, tenderness, warmth, or itching at the site of injection), headache, and joint pain. These are not all of the possible side effects of Hemlibra. Patients can speak with their healthcare provider for more information.

What else should patients know about Hemlibra?

Patients should see the detailed "Instructions for Use" that comes with Hemlibra for information on how to prepare and inject a dose of Hemlibra, and how to properly throw away (dispose of) used needles and syringes.

Patients should stop taking their prophylactic bypassing therapy the day before they start Hemlibra
Patients may continue taking their prophylactic factor VIII for the first week of Hemlibra
Hemlibra may interfere with laboratory tests that measure how well blood is clotting and create an inaccurate result. Patients should speak with their healthcare provider about how this may affect their care.

Medicines are sometimes prescribed for purposes other than those listed in a Medication Guide. Patients should only use Hemlibra for the condition it was prescribed. Patients should not give Hemlibra to other people, even if they have the same symptoms that they have. It may harm them.

Patients should tell their healthcare provider about all the medicines they take, including prescription medicines, over-the-counter medicines, vitamins, or herbal supplements. Patients should keep a list of them to show their healthcare provider and pharmacist.

Before using Hemlibra, patients should tell their healthcare provider about all of their medical conditions, including if they are pregnant, plan to become pregnant, are breastfeeding, or plan to breastfeed.

Since Hemlibra was tested in males, there is no information on whether Hemlibra may impact an unborn baby or breast milk. Females who are able to become pregnant should use birth control during treatment.

Side effects may be reported to the FDA at (800) FDA-1088 or www.fda.gov/medwatch. Side effects may also be reported to Genentech at (888) 835-2555.

Please see Important Safety Information, including Serious Side Effects, as well as the Hemlibra full Prescribing Information and Medication Guide.

About hemophilia A

Hemophilia A is an inherited, serious disorder in which a person’s blood does not clot properly, leading to uncontrolled and often spontaneous bleeding. Hemophilia affects around 20,000 people in the United States, with hemophilia A being the most common form and approximately 50-60% of people living with a severe form of the disorder.

People with hemophilia A either lack or do not have enough of a clotting protein called factor VIII. In a healthy person, when a bleed occurs, factor VIII brings together the clotting factors IXa and X, which is a critical step in the formation of a blood clot to help stop bleeding. Depending on the severity of their disorder, people with hemophilia A can bleed frequently, especially into their joints or muscles. These bleeds can present a significant health concern as they often cause pain and can lead to chronic swelling, deformity, reduced mobility and long-term joint damage.

A serious complication of treatment is the development of inhibitors to factor VIII replacement therapies. Inhibitors are antibodies developed by the body’s immune system that bind to and block the efficacy of replacement factor VIII, making it difficult, if not impossible, to obtain a level of factor VIII sufficient to control bleeding.