TG Therapeutics Announces Data from a Phase 1 Study Evaluating TG-1701 as a Monotherapy and as a Triple Combination with Ublituximab and UKONIQ® at the 2021 European Hematology Association Virtual Congress

On June 11, 2021 TG Therapeutics, Inc. (NASDAQ: TGTX), reported data from TG-1701, the Company’s investigational once-daily, oral BTK inhibitor, as a monotherapy and as a triple therapy in combination with ublituximab, the Company’s novel glycoengineered anti-CD20 monoclonal antibody, and UKONIQ (umbralisib), the Company’s once-daily, inhibitor of PI3K-delta and CK1-epsilon in patients with front line or relapsed/refractory non-Hodgkin’s lymphoma (NHL) and chronic lymphocytic leukemia (CLL) (Press release, TG Therapeutics, JUN 11, 2021, View Source [SID1234583903]). Data from this trial were made available on demand this morning during the 3032 European Hematology Association (EHA) (Free EHA Whitepaper) Virtual Congress. The data presented today were previously presented at the American Society of Clinical Oncology (ASCO) (Free ASCO Whitepaper) annual meeting.

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 HIGHLIGHTS:

Poster Presentation Title: TG-1701, A Selective Bruton Tyrosine Kinase (BTK) Inhibitor, as Monotherapy and in Combination with Ublituximab and Umbralisib (U2) in Chronic Lymphocytic Leukemia (CLL) and Lymphoma

A total of 125 patients with R/R CLL or B-cell lymphoma have been treated with TG-1701, with patients receiving monotherapy in the dose-escalation cohort (n=25), 200 mg in a dose-expansion cohort (n=61), 300 mg in a CLL dose-expansion cohort (n=20), or TG-1701 in combination with U2 in the dose escalation cohort (n=19).
TG-1701 monotherapy was well tolerated and the maximum tolerated dose was not reached up to 400 mg QD.
Adverse Events (AEs) of special interest in patients treated with 200 mg and 300 mg QD of TG-1701 (n=81), included Grade 3 hypertension (4.9%), atrial fibrillation (1.2%), and no instances of major bleeding observed. Grade 3 AEs occurring in ≥10% of patients treated with U2+1701 included diarrhea (11%), neutropenia (11%), ALT increase (16%), and AST increase (16%), and Grade 4 AEs occurring in ≥10% of patients treated with U2+1701 included neutropenia (11%).
At a median follow up of 12.2 months in the 200 mg QD monotherapy expansion cohorts, overall response rates (ORR) were: 95% (19/20) in CLL, 65% (13/20) in mantle cell lymphoma (MCL), and 95% (19/20) in Waldenstrom macroglobulinemia (WM).
100% ORR observed at a median follow up of 8.6 months in the 300 mg CLL monotherapy cohort (n=19).
At a median follow up of 15.6 months, the 1701+U2 dose escalation (using doses of 100mg to 300 mg QD of TG-1701) resulted in 79% ORR, with 21% CR rate across patients with WM, CLL, marginal zone lymphoma (MZL), diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) (n=19).
Data presented at EHA (Free EHA Whitepaper) 2021 is available on the Publications page of the Company’s website at View Source

Sorrento Announces Clinical Research Agreement With Mayo Clinic and FDA Clearance for the First Phase 1b Pilot Study Using Sofusa Lymphatic Drug Delivery Technology to Deliver Ipilimumab in Patients With Melanoma

On June 11, 2021 Sorrento Therapeutics, Inc. (Nasdaq: SRNE, "Sorrento") reported a research collaboration agreement with Mayo Clinic to conduct human clinical proof of concept studies using the Sofusa Lymphatic Drug Delivery System (S-LDDS) technology across multiple products and indications (Press release, Sorrento Therapeutics, JUN 11, 2021, View Source [SID1234583902]). Sofusa is a drug delivery platform which delivers biologic therapies through the skin and directly into the lymphatic system with potential to improve the efficacy and safety of immuno-oncology therapies. Targeting delivery to the lymphatics should also enable reduced dosing as compared to traditional systemic infusions or subcutaneous injections.

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 first study resulting in this agreement is MC20711, a Phase 1b study of the administration of Ipilimumab Intra-Lymphatically using the Sofusa DoseConnect in Patients with Metastatic Melanoma. This is an investigator-initiated study developed by Svetomir Markovic, MD, PhD, and his team. "Checkpoint therapies have shown good results in some patients; however, response rates are still low, and this may be due to poor exposure to drug targets which reside in lymph nodes. By delivering checkpoint therapies directly to the lymphatics, we expect to see an increase in clinical response and potentially a decrease in systemic side effects" – Mike Royal, MD, CMO for Sorrento Therapeutics.

This agreement builds upon the previously announced exclusive licensing agreement where Sorrento licensed Mayo Clinic’s proprietary Antibody-Drug-Nanoparticle albumin-bound Immune Complex platform technology. "Both of these agreements are part of our strategic focus to combine potent antibodies from our G-MAB library with next-generation therapeutic platform technologies to deliver unparalleled safety and efficacy" – Henry Ji, President and CEO, Sorrento Therapeutics.

Mayo Clinic, Dr. Svetomir Markovic and Dr. James Jakub have financial interests in the technology referenced in this release. Mayo Clinic will use any revenue it receives to support its not-for-profit mission in patient care, education, and research.

Momelotinib Oral Presentation at European Hematology Association Demonstrates Association Between Transfusion Independence and Improved Overall Survival

On June 11, 2021 Sierra Oncology, Inc. (SRRA), a late-stage biopharmaceutical company on a quest to deliver targeted therapies that treat rare forms of cancer, reported data presented at the European Hematology Association (EHA) (Free EHA Whitepaper) Annual Meeting highlighting transfusion independence with momelotinib is associated with improved overall survival, including in patients with anemia at baseline (Press release, Sierra Oncology, JUN 11, 2021, View Source [SID1234583900]). In addition, transfusion independence is seen irrespective of baseline degree of anemia, platelet count or transfusion status.

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!

"Momelotinib has previously been shown to provide an anemia benefit for myelofibrosis patients, resulting in higher rates of transfusion independence. The data presented at EHA (Free EHA Whitepaper) are particularly exciting because they suggest myelofibrosis patients randomized to momelotinib who achieve or maintain transfusion independence at Week 24 achieve a clinically relevant improvement in overall survival, the gold standard measurement of oncology treatment.," said Barbara Klencke, MD, Chief Medical Officer at Sierra Oncology. "The correlation between Week 24 transfusion independence response and overall survival observed with momelotinib is unique among JAK inhibitors. This suggests that the likelihood of achieving or maintaining transfusion independence may become an important consideration for physicians when recommending myelofibrosis treatment options. With this in mind, we wanted to understand which patients were most likely to maintain or achieve transfusion independence by Week 24. In other data presented at EHA (Free EHA Whitepaper), we report observing higher rates of transfusion independence with momelotinib relative to ruxolitinib, regardless of a patient’s baseline degree of anemia, platelet count or transfusion status."

Transfusion Independence is Associated with Improved Overall Survival in Myelofibrosis Patients Receiving Momelotinib

Efficacy data examining the association between transfusion independence and overall survival for momelotinib patients from SIMPLIFY-1 (JAKi-naïve) and SIMPLIFY-2 (JAKi-exposed) were presented in an oral presentation by Ruben Mesa, MD, Director of the Mays Cancer Center, UT Health San Antonio, MD Anderson Cancer Center. Previously published data demonstrated higher transfusion independence response rates in the momelotinib arms of SIMPLIFY-1 (67% vs. 49%) and SIMPLIFY-2 (43% vs. 21%) compared with ruxolitinib or best available therapy, respectively. Additionally, robust overall survival was observed with extended momelotinib treatment in both JAKi-naïve and JAKi-exposed patients (median not yet reached in SIMPLIFY-1 and 34.3 months in SIMPLIFY-2) as reported at ASH (Free ASH Whitepaper) 2020. These new analyses being presented at EHA (Free EHA Whitepaper) 2021 suggest JAKi-naïve patients receiving momelotinib achieve or maintain transfusion independence (TI) at Week 24 have favorable overall survival compared to non-responders, with a similar trend observed in SIMPLIFY-2.

Key Data Presented

In SIMPLIFY-1 (JAKi-naïve patients)
Patients randomized to momelotinib who were TI responders show a three-year overall survival of 80% compared to 50% for non-responders (HR=0.30; p<0.0001)
Patients with anemia at baseline (RBC transfusion <12 weeks before baseline, or baseline Hgb <10 g/dL) show a similar survival advantage for momelotinib patients who achieve a TI response at Week 24 compared to TI non-responders (HR=0.32; p=0.0006)
In SIMPLIFY-2 (JAKi-exposed patients)
Patients randomized to momelotinib who were TI responders at Week 24 show a trend toward better OS compared to TI non-responders (HR=0.52; p=0.0652)
Improved Transfusion Independence Rates for Momelotinib vs. Ruxolitinib in Anemic JAKi Naïve Myelofibrosis Patients Independent of Baseline Platelet or Transfusion Status

Progressive anemia is a common occurrence in myelofibrosis, with nearly all patients requiring transfusions as their disease advances. As noted in the EHA (Free EHA Whitepaper) oral presentation, JAKi-naïve patients receiving momelotinib who maintain or achieve transfusion independence at Week 24 have favorable overall survival compared to non-responders, with a similar trend observed in SIMPLIFY-2.

Data presented in a poster presentation by Jean-Jacques Kiladjian, MD, PhD, Professor of Clinical Pharmacology, Paris Diderot University and Consultant Hematologist and Head of the Clinical Investigation Center, Saint Louis Hospital, show that the prognostically important Week 24 TI rates in JAKi-naïve myelofibrosis patients were consistently higher in anemic patients receiving momelotinib compared to ruxolitinib in all patient subsets with baseline hemoglobin <14 g/dL and all subsets defined by baseline platelet count and baseline transfusion status. Combined with data from the oral presentation, these data suggest the goal of achieving TI should become an important driver of treatment decisions in myelofibrosis.

ONK Therapeutics’ CSO Prof. M O’Dwyer and his Academic Group Present New Data Showing the Benefit of Knocking Out the Inhibitory Receptor CD96 on Human NK Cells in the Context of Multiple Myeloma at EHA 2021

On June 11, 2021 ONK Therapeutics Ltd, an innovative natural killer (NK) cell therapy company, reported that new data presented by John Daly from the academic lab of CSO, Prof. Michael O’Dwyer at the Annual European Hematology Association (EHA) (Free EHA Whitepaper) 2021 Virtual Congress illustrating the merit of knocking out the checkpoint inhibitory receptor for CD96 on NK cells in the context of MM (Press release, ONK Therapeutics, JUN 11, 2021, View Source [SID1234583898]).

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 data is based on research studies carried out at the National University of Ireland, Galway (NUI Galway), College of Medicine, Nursing and Health Sciences, by Prof. O’Dwyer’s academic research group.

Contradictory data had previously shown that CD96 could be both an activating and an inhibitory receptor on human NK cells, depending on the tumor type being examined. The data presented during the poster presentation demonstrate conclusively that in the case of MM cells expressing CD155, CD96 is a human NK cell inhibitory receptor, regulating both cytotoxicity and cytokine release. Experiments using CRISPR/Cas9-mediated KO of CD96 in primary NK cells resulted in enhanced cytotoxicity and cytokine release compared to controls. Studies carried out in parallel also demonstrated a similar beneficial effect of CRISPR/Cas9-mediated KO of TIGIT in primary NK cells.

Furthermore, NK cells obtained from MM patient bone marrow had particularly high levels of CD96 expression, suggesting NK cells in the bone marrow of MM patients are more likely to be susceptible to CD155 mediated immune-evasion.

These new insights support knocking out inhibitory checkpoint receptors, including CD96 KO and TIGIT KO as a promising approach to improving the functionality of off-the-shelf engineered NK cell therapies for the treatment of cancer.

E-poster presentation title: Knockout of CD96 or TIGIT using CRISPR/Cas9 enhances NK induced
cytotoxicity and cytokine production in the presence of CD155 expressing myeloma cells.

Author(s)/Presenters: John Daly, Mark Gurney, Michael O’Dwyer.

Session title: Myeloma and other monoclonal gammopathies – Biology & Translational Research.

Abstract number: EP941

Date and Time: Available on the virtual platform as an e-poster Friday, June 11 at 9:00 CEST.

Download a copy HERE.

Chris Nowers, ONK Therapeutics’ CEO said, "The studies carried out by Prof. O’Dwyer’s academic research group are expanding our deep understanding of NK cell biology and are helping to confirm certain gene constructs and edits that will enhance NK cell cytotoxicity, cytokine production and persistence in the tumor microenvironment. Gene edited NK cells lacking CD96 and/or TIGIT could therefore be beneficial for treating CD155 expressing malignancies, such as Multiple Myeloma."

ONK Therapeutics was formed based on technology and intellectual property developed at NUI Galway by Prof. O’Dwyer. The Company is developing off-the-shelf, optimized NK cell therapies for cancer that utilize dual-targeting of the death receptor pathway in addition to incorporating a CAR, along with further strategies that utilize novel gene editing approaches to enhance persistence, metabolic profile, and cytotoxic potential. ONK has exclusive licenses to a broad intellectual property (IP) against a wide range of NK cell checkpoints, including CD96 and TIGIT.

Keros Therapeutics Presents Preclinical Data from ALK2 and KER-050 Hematology Programs at the European Hematology Association EHA2021 Virtual Congress

On June 11, 2021 Keros Therapeutics, Inc. ("Keros") (Nasdaq: KROS), a clinical-stage biopharmaceutical company focused on the discovery, development and commercialization of novel treatments for patients suffering from hematological and musculoskeletal disorders with high unmet medical need, reported data from the ALK2 and KER-050 hematology programs at the European Hematology Association (EHA) (Free EHA Whitepaper) EHA (Free EHA Whitepaper)2021 Virtual Congress held from June 9 through June 17, 2021 (Press release, Keros Therapeutics, JUN 11, 2021, View Source [SID1234583897]).

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"We are pleased to be able to present multiple posters at this year’s EHA (Free EHA Whitepaper)2021 Virtual Congress. Our preclinical studies continue to elucidate the relationship between ALK2 inhibition, hepcidin and serum iron. These data support that ALK2 inhibition may be a potential treatment option for indications associated with high hepcidin, including anemia of inflammation ("AI") and iron refractory iron-deficiency anemia ("IRIDA"). In addition, the data suggests that ALK2 inhibition has the potential to mobilize and reduce tissue iron in diseases of iron overload," said Jasbir S. Seehra, Ph.D., Chief Executive Officer of Keros. "We also presented new preclinical data for KER-050 that demonstrated its rapid and durable effects on erythropoiesis and increases in circulating erythropoietin, which we believe provides a strong rationale for investigating KER-050 as a treatment for ineffective hematopoiesis in MDS and myelofibrosis."

Details of the presentations are as follows:

Inhibition of ALK2 Through Administration of a Small Molecule Inhibitor Decreased Hepcidin, Increased Serum Iron and Ameliorated Anemia in an Induced Model of Anemia of Inflammation

ALK2 is a potential therapeutic target in anemia resulting from chronic inflammation – Abstract Number: EP839
To induce a model of chronic kidney disease ("CKD"), mice were dosed daily for six weeks with 50 mg/kg of adenine, leading to iron deficiency anemia (37.6% decrease in serum iron; p<0.0001), associated with increased circulating interleukin-6 ("IL6") (2.42-fold; p<0.05) and hepcidin (3.49-fold; p<0.01). After completing the six weeks of adenine-administration, we administered 5 mg/kg of either a selective small molecule ALK2 kinase inhibitor ("KTI-2338"), or vehicle daily for 10 days in the CKD mice, and observed a reversal of the CKD-related changes in the KTI-2338-treated mice. We observed an increase in serum iron (108.2%; p<0.0001), decrease in hepcidin (85.4%; p<0.0001) and improvements in red blood cell ("RBC") count (7.2%; ns), hemoglobin ("HGB") (10.9%; p<0.0001) and hematocrit (10.3%; p<0.05) compared to vehicle. Reticulocyte hemoglobin ("RET-HGB") content, a measure of the incorporation of iron into hemoglobin, increased 9.9% (p<0.01) with KTI-2338 compared to vehicle-treated CKD mice, normalizing to baseline levels.

These data demonstrate that inhibition of ALK2 improved hematological markers of anemia, serum hepcidin and serum iron levels in a mouse model of AI resulting from chronic kidney disease characterized by elevated IL6, suggesting that targeting ALK2 inhibition could potentially treat anemia arising from high hepcidin.

Administration of an ALK2 Inhibitor in a Mouse Model of Iron Overload Resulted in Significant Reductions in Liver Non-Heme Iron

ALK2 inhibition, a novel therapeutic approach to iron overload – Abstract Number: EP842
Administration of KTI-2338 reduced circulating hepcidin levels and increased serum iron in wild-type mice after three days of daily dosing. Specifically, we observed a reduction in hepcidin as soon as four hours post the third administration of KTI-2338, which was sustained through 12 hours post-administration, as well as an increase in serum iron eight hours post-administration, which peaked at 16 hours post-administration.

Mice were dosed daily with 100 mg/kg of iron dextran to induce iron overload and subsequently dosed with either KTI-2338 (5 mg/kg) or vehicle. Mice dosed with iron dextran became iron overloaded and exhibited a substantial 47-fold increase in hepatic iron compared to mice that received vehicle (p<0.0001). Our initial results indicated that, after 63 hours of dosing with KTI-2338, there were significant reductions in liver non-heme iron content (62%). Subsequent analysis using a more selective iron assay confirmed that treatment with KTI-2338 reduced non-heme iron (13.4%) compared to vehicle treated mice (p<0.006).

These data suggest that, in conditions with iron overload, ALK2 inhibition may potentially be used to remove excess iron from the liver, potentially improving the effectiveness of chelation therapy and excretion.

Treatment with ALK2 Antibodies to Neutralize the ALK2 Receptor Reduced Serum Hepcidin and Increased Circulating Iron in Non-Human Primates, a Preclinical Model Highly Representative of Human Biology

Administration of ALK2 neutralizing antibodies to cynomolgus monkeys led to a sustained decrease in hepcidin, increase in circulating iron and increase in erythrocyte hemoglobin – Abstract Number: EP840
Keros has developed two fully human antibodies, KTI-016 and KTI-018, that are designed to specifically bind to and neutralize the ALK2 receptor. To determine the pharmacokinetic and pharmacodynamic properties of these antibodies, female cynomolgus monkeys received a single subcutaneous dose (3 mg/kg) of either antibody. Serum drug concentrations and indices of iron and hematology were assessed intermittently over an 8-week period. KTI-016 and KTI-018 were both rapidly absorbed, reached Cmax within 48 hours and had half-lives of 49.1 hours and 33.9 hours, respectively.

Six hours after administration, KTI-016 and KTI-018 reduced serum hepcidin by 50.3% and 55.6%, respectively. The reduction in hepcidin peaked starting at 48 hours post-administration, at 77.8% in the KTI-016-treated group and 77.2% in the KTI-018-treated group. These decreases remained through day 10 before returning to baseline by day 14. A corresponding increase in circulating iron occurred starting at 24 hours following administration, peaking at 63.9% (p<0.01) and 72.4% (p<0.001) in the KTI-016- and KTI-018-treated groups, respectively. This response was also sustained through day 10, returning to baseline by day 14.

KTI-016 and KTI-018 were also observed to have comparable effects on increasing RET-HGB, red blood cell hemoglobin ("RBC-HGB") and mean corpuscular hemoglobin concentration ("MCHC") (data from the two antibodies were combined). RET-HGB increased by 4.9% (p<0.001) at 3 days post-dose and remained elevated for at least 10 days. Increases in RBC-HGB content were observed initially at 35 days post-dose, with a 4.2% increase (p<0.0001) at day 56. We also observed increases in MCHC starting at 42 days post-dose, with a 3.7% increase (p<0.001) at day 56.

We believe iron mobilized by treatment with KTI-016 and KTI-018 was incorporated into hemoglobin, as evidenced by the observed increases in RET-HGB, RBC-HGB and MCHC. Accordingly, these data demonstrate that these antibodies may be a potential treatment for anemias arising from elevated hepcidin, such as IRIDA and AI.

Preclinical Study of KER-050 Demonstrated Effects on Multiple Stages of Erythroblast Maturation and Increased Circulating Erythropoietin

KER-050, an inhibitor of TGF-β superfamily signaling, observed to have a rapid, dynamic, and durable effect on erythropoiesis – Abstract Number: EP758
Mice treated with a single dose of a research form of KER-050 ("RKER-050") showed rapid and sustained increases in RBCs and HGB, observed from 12 hours (RBCs +8%; p<0.001 and HGB +9%; p<0.005) through day 51 (RBCs + 8% p<0.001), compared to vehicle-treated mice.

Treatment with RKER-050 also resulted in a dynamic mobilization of erythroblasts from the bone marrow into circulation. At 12 hours post-administration, we observed a 20% reduction in bone marrow ("BM") proerythroblasts that corresponded to a 37% increase in erythroblasts at 24 hours post-administration, which we believe indicates that early progenitors were differentiating into erythroblasts. We also observed an increase in mature erythroblasts, which suggests the differentiation of late-stage progenitors. This maturation of erythroblasts corresponded to an increase in circulating reticulocytes ("RETs") at 12 hours post-administration (+18%; p<0.05), as well as to an increase in RBCs observed at 48 hours post-dose (+8% p<0.0001).

The initial increase in RETs observed at 12 hours post-administration was reduced (-15%; p<0.05) at 24 hours post-administration, which we believe indicates that RETs matured to RBCs. At 48 hours post-administration, RET numbers returned to vehicle-comparable levels, which we believe indicates that the RET pool was replenished. This data potentially demonstrates that there was a continued progression of erythroblasts through maturation. This increased differentiation and maturation of erythroblast contributed to a sustained upregulation of erythropoiesis at day 14 post-dose.

In the RKER-050-treated mice, erythropoietin upregulation was not observed until day 4. However, erythropoietin was significantly increased from day 7 (+68%, p<0.0001) through day 37 (4-fold increase, p<0.05) which potentially indicates that erythropoietin has a contributing role in sustaining the RKER-050-mediated increase in RBCs and HGB, compared to vehicle, through at least day 51.

We believe the observed rapid and durable effect on erythropoiesis via multiple stages of erythroblast maturation (both early- and late-stage) as well as the observed increase in circulating erythropoietin provides a strong rationale for investigating KER-050 as a potential treatment for ineffective erythropoiesis in MDS and myelofibrosis.

About KER-050

Keros’ lead protein therapeutic product candidate, KER-050, is an engineered ligand trap comprised of a modified ligand-binding domain of the Transforming Growth Factor-Beta receptor known as activin receptor type IIA that is fused to the portion of the human antibody known as the Fc domain. KER-050 is being developed for the treatment of low blood cell counts, or cytopenias, including anemia and thrombocytopenia, in patients with myelodysplastic syndromes, or MDS, and in patients with myelofibrosis. In October 2020, Keros announced the dosing of the first two participants in its Phase 2 clinical trial evaluating KER-050 for the treatment of anemia and thrombocytopenia in very low-, low-, or intermediate-risk MDS. Keros expects to report initial data from Part 1 of this trial by the end of June 2021. Additionally, Keros plans to commence an open-label Phase 2 clinical trial evaluating KER-050 for the treatment of patients with myelofibrosis-associated cytopenias in the third quarter of 2021 and expects to report initial data from this trial in 2022.