Precision BioSciences Presents Analysis of Azer-Cel, Allogeneic CAR T Product Attributes Related to In Vivo Pharmacokinetics, Pharmacodynamics, and Clinical Outcomes at 2022 American Society of Hematology Annual Meeting

On December 10, 2022 Precision BioSciences (Nasdaq: DTIL), a clinical stage gene editing company developing ARCUS-based ex vivo allogeneic CAR T and in vivo gene editing therapies, reported a novel, product-attributes analysis of its lead CD19 allogeneic CAR T candidate, Azercabtagene Zapreleucel (azer-cel; PBCAR0191) that shows a relationship between CAR T cell composition and effective cell dose with pharmacokinetics, pharmacodynamics, and clinical outcomes (Press release, Precision Biosciences, DEC 10, 2022, View Source [SID1234625028]). Data from this analysis, Effective Cell Dose and Functional Attributes of Azercabtagene Zapreleucel (azer-cel; PBCAR0191) Associated with Allogeneic CAR T-Cell Safety and Efficacy in Patients with Relapsed/Refractory B-Cell Lymphoma, were presented today during a poster session at the American Society of Hematology (ASH) (Free ASH 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!

"Autologous CAR T therapy remains one of the most promising approaches in the treatment of hematological malignancies. However, 30-60% of high-grade non-Hodgkin Lymphoma (NHL) patients relapse after treatment and, for up a proportion of patients, an effective autologous product cannot be manufacturedi," said Caron A. Jacobson, M.D., azer-cel clinical trial investigator and Medical Director for the Immune Effector Cell Therapy Program at Dana-Farber Cancer Institute. "Unlike autologous CAR T cell therapy, all allogeneic CAR T products are cryopreserved, which may alter the effective dose and composition of the post-thaw product. In this analysis, azer-cel cellular attributes were interrogated in the post-thaw product for possible relationship to in vivo pharmacokinetics, pharmacodynamics, and clinical outcomes for 44 subjects with NHL across multiple azer-cel dose levels and lymphodepletion regimens. The analysis found that CAR T expansion, a key determinant of durable response, strongly correlated with non-apoptotic CAR T cell dose."

Azer-cel is an investigational allogeneic anti-CD19 CAR T candidate currently in a Phase 1/2a clinical trial of adult subjects with relapsed or refractory NHL, who relapsed following treatment with an autologous CAR T.

"This is the first analysis of an allogeneic anti-CD19 CAR T product to examine the relationships between allogeneic CAR T cell composition, cell dose, and lymphodepletion with pharmacokinetics, pharmacodynamics, and clinical outcomes," said Alan List, M.D., Chief Medical Officer, Precision BioSciences. "These results indicate that the post-thaw CAR T product composition drives in vivo cell expansion potential and CAR T-related adverse events. We are continuing to use this information in real time, applying optimizations across our first- and second-generation allogeneic CAR T platforms with the goal of improving those attributes and characteristics that drive predictability, reliability, and performance of CAR T cell therapy."

The analysis also showed that CD4:CD8 ratio strongly correlated with in vivo CD4+ CAR T cell expansion. Similar to data reported in autologous CAR T studies, differentiated CD4+ CAR T cell dose correlated with Grade 3 or greater neurotoxicity. This was particularly observed in a subset of patients that were both CAR T relapsed and conditioned with an intensified lymphodepletion treatment regimen.

Oncternal Therapeutics Presents Updated Interim Data for Zilovertamab in Combination with Ibrutinib at ASH 2022

On December 10, 2022 Oncternal Therapeutics, Inc. (Nasdaq: ONCT), a clinical-stage biopharmaceutical company focused on the development of novel oncology therapies, reported updated interim clinical data from ongoing Phase 1/2 Study CIRM-0001 in an oral presentation at the American Society of Hematology (ASH) (Free ASH Whitepaper) 2022 Annual Meeting (Press release, Oncternal Therapeutics, DEC 10, 2022, View Source [SID1234625027]). In the study, zilovertamab, an investigational anti-ROR1 monoclonal antibody, is being evaluated in combination with ibrutinib in patients with mantle cell lymphoma (MCL), chronic lymphocytic leukemia (CLL) and in a recently opened cohort for patients with marginal zone lymphoma (MZL). The clinical trial is being conducted in collaboration with the University of California San Diego (UC San Diego) and has been partially funded by the California Institute for Regenerative Medicine (CIRM).

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 presented in today’s oral session further strengthens our confidence in the significant clinical benefit the combination of zilovertamab and ibrutinib can provide to patients with MCL and CLL. The rapid onset of deep responses in patients with MCL supports our recently initiated randomized global registrational Phase 3 Study ZILO-301 of the combination of zilovertamab plus ibrutinib," said James Breitmeyer, M.D., Ph.D., Oncternal’s President and CEO. "The robust response rates and prolonged PFS seen for MCL and CLL patients expressing the p53 mutation/del(17p), who are underserved by current standard of care treatments, is especially exciting. We continue to believe that the inhibition of ROR1 with zilovertamab can play a key role in addressing important unmet medical needs in difficult-to-treat patients across various hematological malignancies."

The updated interim data presented during an oral presentation at the ASH (Free ASH Whitepaper) 2022 meeting include 33 patients with relapsed/refractory (R/R) MCL enrolled in the dose-finding and dose-expansion cohorts of Study CIRM-0001 (Part 1 + Part 2), of whom 28 were evaluable for efficacy as of the October 11, 2022 data cut-off date.

Patients had high-risk factors and were heavily pre-treated at study entry, with 52% having a high Ki-67 proliferative index (≥30%), 47% with p53 mutations or deletions in chromosome 17p (del(17p)), and 46% having an intermediate or high sMIPI prognostic score.
The objective response rate (ORR) was 89% (25 of 28 evaluable patients) and includes recently enrolled patients.
The complete response (CR) rate was 43% (12 of 28 evaluable patients) at 26 months and was already 18% (5 of 28 evaluable patients) at 3 months.
Historical data published for single agent ibrutinib for 370 patients with R/R MCL from three clinical trials showed an ORR of 66%, CR rate of 20% and median PFS of 12.8 months (Rule 2017, British Journal of Haematology).
The partial response (PR) rate was 46% (13 of 28 evaluable patients), and the stable disease (SD) rate was 4% (1 of 28 evaluable patients), for a total clinical benefit rate (CR, PR and SD) of 93%.
The median PFS has not been reached after a median follow-up of 19.5 months (95% CI: 19.4, 28.5), regardless of the number of prior systemic therapies.
Median PFS was also favorable in patients with high-risk features associated with disease difficult-to-treat with BTK inhibitors:
P53 mutation/del(17p): median PFS not reached (95% CI: 2.85, NE). Historical data for single agent ibrutinib in 20 patients with p53 mutation showed a median PFS of 4.0 months (Rule 2019, Haematologica).
Ki-67 ≥30%: median PFS 33.2 months (95% CI: 2.85, NE).
>1 prior systemic therapy: median PFS not reached (95% CI: 4.33, NE).
Thirty-four patients with CLL in the dose-finding and dose-confirming cohorts of this clinical trial (Part 1 & Part 2) as of the October 11, 2022 data cut-off date were all evaluable for efficacy. Patients had high-risk factors, and most were heavily pre-treated at study entry, with a median of two systemic prior therapies (range 1-9).

Landmark PFS was 100% at 42 months in patients with R/R CLL expressing p53 mutation/del(17p) treated with the combination of zilovertamab plus ibrutinib. The most recent data update from the ALPINE study in patients with R/R CLL expressing p53 mutation/del(17p) showed a landmark PFS of 77.6% at 24 months for zanubrutinib monotherapy and 55.7% at 24 months for ibrutinib monotherapy (Brown 2022, ASH (Free ASH Whitepaper)).
Landmark PFS was 95% at 24 months in all patients with R/R CLL treated with the combination of zilovertamab plus ibrutinib. The most recent data update from the ALPINE study in R/R CLL patients showed a landmark PFS at 24 months of 79.5% for zanubrutinib and 67.3% for ibrutinib monotherapy (Brown 2022, ASH (Free ASH Whitepaper)).
The median overall survival (OS) was not reached at 40 months for patients with CLL with p53 mutation/del(17p).
Thirty-one patients with CLL have also been enrolled in the randomized efficacy cohort of this clinical trial (Part 3), of which 23 were evaluable for efficacy. Data from this cohort are maturing. The median PFS had not been reached for either group as of the October 11, 2022 cut-off date after a median follow up of 29 months.

The combination of zilovertamab plus ibrutinib has been well tolerated as of the October 11, 2022 cut-off date, with treatment emergent adverse events consistent with or slightly improved compared to those reported for ibrutinib alone. There have been no dose-limiting toxicities and no serious adverse events attributed to zilovertamab alone. Atrial fibrillation was observed in only 9.4% of the patients and febrile neutropenia in 1.2% of patients.

About the CIRM-0001 Study
The CIRM-0001 Study is a Phase 1/2 trial evaluating zilovertamab in combination with ibrutinib in separate groups of patients with CLL, MCL or MZL. Enrollment of the dose-finding cohorts in CLL and MCL, dose-expansion cohort in CLL and MCL and randomized Phase 2 cohort in CLL has been completed. An additional dose-expansion cohort of up to 10 patients with MZL has started enrolling patients. Additional information about the CIRM-0001 clinical trial and other clinical trials of zilovertamab may be accessed at ClinicalTrials.gov.

About Zilovertamab
Zilovertamab is an investigational, humanized, potentially first-in-class monoclonal antibody targeting Receptor tyrosine kinase-like Orphan Receptor 1 (ROR1). A Phase 3 registrational trial evaluating zilovertamab plus ibrutinib versus ibrutinib plus placebo in relapsed or refractory Mantle Cell Lymphoma (MCL) has been initiated (NCT05431179). Zilovertamab is also being studied in a Phase 1/2 clinical trial in combination with ibrutinib for the treatment of patients with MCL, chronic lymphocytic leukemia (CLL) or marginal zone lymphoma (MZL), in a collaboration with the University of California San Diego (UC San Diego) School of Medicine and the California Institute for Regenerative Medicine (CIRM). In addition, Oncternal is supporting two investigator-sponsored studies being conducted at the UC San Diego School of Medicine, a Phase 1b clinical trial for patients with metastatic castration-resistant prostate cancer (mCRPC), and a Phase 2 clinical trial of zilovertamab in combination with venetoclax, a Bcl-2 inhibitor, for patients with relapsed/refractory CLL. Both are open for enrollment.

ROR1 is a potentially attractive target for cancer therapy because it is an onco-embryonic antigen, not usually expressed on adult cells, but its expression confers a survival and fitness advantage when reactivated and expressed by tumor cells. Researchers at the UC San Diego School of Medicine discovered that targeting a critical epitope on ROR1 was key to specifically inhibiting ROR1-expressing tumors. This led to the development of zilovertamab which binds this critical epitope of ROR1, highly expressed on many different cancers but not on normal tissues. Preclinical data showed that when zilovertamab bound to ROR1, it blocked Wnt5a signaling, inhibited tumor cell proliferation, migration and survival, and induced differentiation of the tumor cells. The FDA has granted Orphan Drug Designations to zilovertamab for the treatment of patients with MCL and CLL/small lymphocytic lymphoma. Zilovertamab is in clinical development and has not been approved by the FDA for any indication.

December 10, 2022: MaaT Pharma Presents Compelling Consolidated MaaT013 Clinical Data at the 64th ASH Annual Meeting

On December 10, 2022 MaaT Pharma (EURONEXT: MAAT – the "Company"), a French clinical-stage biotech and a pioneer in the development of Microbiome Ecosystem TherapiesTM (MET) dedicated to improving survival outcomes for patients with cancer, reported data from the continuing compassionate use program (EAP) in France for MaaT013 at the Annual Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting in New Orleans, U.S. To see the abstract, click here (Press release, MaaT Pharma, DEC 10, 2022, View Source [SID1234625025]).

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 EAP consolidated results, presented in an oral format on December 10 at 5:15 pm CET/ 10.15am CST by Prof. Mohamad Mohty, Head of the Clinical Hematology and Cellular Department at the Saint-Antoine Hospital and Sorbonne University, include data from 81 patients treated with MaaT013, the company’s high-richness, high-diversity lead Microbiome Ecosystem TherapyTM (MET) for hospital use in an acute setting. Patients developing refractory acute Graft-versus-Host Disease with gastrointestinal involvement (GI-aGvHD) following hematopoietic cell transplantation demonstrated an overall response rate (GI-ORR) of 56% at day 28 following MaaT013 treatment. In patients responding to MaaT013 therapy, the overall survival (OS) rate at 12 months was 59% (compared to 14% for non-responders) indicating a significant clinical benefit with MaaT013.

In aGvHD patients refractory to 1st-line (steroids) and 2nd-line (ruxolitinib) treatments (n=31) and receiving MaaT013 as a 3rd-line therapy, 65% demonstrated a GI-ORR at day 28. The 12-month OS rate in the MaaT013-responding group was 74%. This patient population resembles the ongoing Phase 3 ARES clinical trial (NCT04769895) being conducted in Europe.

"The clinical benefits we continue to observe with MaaT013 are promising and reinforce the potential for our MET to improve survival outcomes for aGvHD patients when early treatment lines are unsuccessful" said Hervé Affagard, CEO and co-founder of MaaT Pharma. "The year 2022 will be remembered as a turning point for the microbiome therapeutics industry as it continues to mature. We expect to see an acceleration in the field following the first FDA approval for a microbiome-based product in preventing C. difficile infection as well as promising clinical results in various infectious disease and oncology indications."

Prof. Mohamad Mohty added, "Patients with severe acute GvHD that have undergone several lines of treatments have a high mortality risk with no currently proven salvage treatment options. The results observed with MaaT013 are very encouraging especially in the 3rd-line setting where we see that the survival outcome is significantly improved. Since the patient population being treated with MaaT013 in the company’s Phase III ARES pivotal study in Europe is similar to those treated in the EAP, we are hopeful that the clinical trial results will corroborate these positive results and will provide an important benefit to all patients who are in need."

Key clinical findings with MaaT013 in compassionate use in France (Early Access Program or ‘EAP’)

In the EAP, 81 patients with steroid-dependent or steroid-resistant, Grade II-IV, gastrointestinal aGvHD were treated with MaaT013 from July 2018 to May 2022

45 out of 81 (56%) showed objective GI response at day 28 of which 30 patients (37%) had a complete response, 11 patients (14%) had a very good partial response, and 4 patients (5%) showed a partial response.
Overall survival (OS) in MaaT013-responding patients at the 12-month follow-up was 59%, compared to 14% in non-responders (OS in all included patients was 39% at 12 months).
Considering GvHD response in all organs (GI, skin, liver), 38 out of 78 patients (49%) showed an objective response rate (ORR) at day 28, of which 24 patients (31%) had a complete response, 11 patients (14%) had a very good partial response, and 3 patients (4%) showed a partial response.
At the time of treatment, all patients had either Grade II (11%), Grade III (51%) or Grade IV (38%) aGvHD (MAGIC Classification).
Patients received MaaT013 after 1 to 6 (median: 2; 66/81 received ruxolitinib) lines of treatment
68 out of 81 patients (84%) were steroid resistant of which 33 out of 68 (49%) showed an objective GI response at day 28; among these patients, 21 patients (31%) had a complete response, 9 patients (13%) had a very good partial response, and 3 patients (4%) showed a partial response.
66 out of 81 patients (81%) were refractory to ruxolitinib (any treatment line), of which, 37 out of 66 (56%) showed an objective GI response at day 28; of these 25 patients (38%) had a complete response, 9 patients (14%) had a very good partial response, and 3 patients (5%) showed a partial response
31 out of 66 patients were ruxolitinib-refractory in 2nd-line and MaaT013 was administered as a 3rd-line treatment; 20 out of 31 patients (65%) showed an objective GI response at day 28; among these patients, 19 patients (61%) had a complete response, and 1 patient (3%) had a very good partial response.
13 out of 81 patients (16%) were steroid-dependent of which 12 out of 13 patients (92%) showed an objective GI response at day 28; among these patients, 9 patients (69%) had a complete response, 2 patients (15%) had a very good partial response, and 1 patient (8%) showed a partial response.

Evaluation of MaaT013 in the Phase III pivotal clinical trial ARES

MaaT Pharma announced the first patient enrolled in the Phase III, open label, single arm, ARES pivotal trial for MaaT013 in March 2022, with a safety and data review by an independent data safety and monitoring board (DSMB), after enrollment of half of the patients in the study, expected in the first half of 2023.

As of today, MaaT013 has been safely administered to more than 160 patients in Europe in clinical trials and in the Expanded Access Program in France. Indeed, additionally to clinical trials, MaaT Pharma has been pursuing, since 2019, the compassionate use program in France, approved by the ANSM, to faster access to MaaT013 for patients with unmet medical need, mainly for indications in acute Graft-versus-Host disease. This program also allows the Company to strengthen its supply chain and production capacities to safely provide MaaT013, on a regular basis to 24 transplantation centers in France.

About MaaT013

MaaT013 is a standardized, high-richness, high-diversity Microbiome Ecosystem TherapyTM containing ButycoreTM (group of bacterial genera known to produce immuno-regulatory metabolites). It aims to restore the symbiotic relationship between the patient’s functional gut microbiome and their immune system to correct the responsiveness and tolerance of immune functions and reduce steroid-resistant, gastrointestinal-predominant aGvHD. MaaT013 has been granted Orphan Drug Designation by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). MaaT013 is an off-the-shelf, healthy-multi-donors-derived product intended for acute, hospital use.

LAVA-051, a novel bispecific gamma-delta T-cell engager (Gammabody™), in relapsed/refractory MM and CLL: pharmacodynamic and early clinical data

On December 10, 2022 LAVA Therapeutics N.V. (Nasdaq: LVTX), a clinical-stage immuno-oncology company focused on developing its proprietary Gammabody platform of bispecific gamma delta T cell engagers to transform the treatment of cancer, reported a poster presentation highlighting updated data, including safety, pharmacodynamics (PD) and pharmacokinetics (PK) from the ongoing Phase 1/2a clinical trial of LAVA-051 in patients with relapsing/refractory (R/R) chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) at the 64th American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting and Exposition, taking place in New Orleans, Louisiana and virtually December 10–13, 2022 (Press release, Lava Therapeutics, DEC 10, 2022, View Source [SID1234625024]). The presentation includes initial data from patients receiving LAVA-051 subcutaneously, along with updates on the intravenous dosing-cohorts.

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!

"To date, the Phase 1 data, as presented, showed that dose escalation of LAVA-051 up to 200 µg could be achieved in patients with R/R MM and CLL without evidence of dose-limiting toxicity and cytokine release syndrome (CRS). Often, such toxicities are a significant safety challenge for T-cell engager therapies," said Arnon Kater, M.D., Ph.D., chairman of the Dutch/Belgium HOVON CLL working group and professor of translational hematology at the Amsterdam University Medical Center, and LAVA-051 clinical trial investigator. "I am pleased that this first clinical study with a gamma delta T-cell engager has progressed into more relevant dose levels." In the phase 1/2a study of LAVA-051 in patients with relapsed/ refractory (R/R) CLL, MM and AML (NCT04887259), the primary objectives are to investigate safety and tolerability of LAVA-051 and determine the recommended Phase 2 dose (RP2D) of LAVA-051. The secondary objectives include evaluation of PK, PD, immunogenicity, and preliminary anti-tumor activity.

In addition to the favorable safety profile demonstrated as of the data cutoff (November 11, 2022), LAVA-051 showed predictable and linear pharmacokinetics and on-mechanism pharmacodynamic parameters consistent with Vγ9Vδ2-T cell engagement, including increasing occupancy of patient Vγ9Vδ2-T cells with LAVA-051 and consistent increases in the expression of T-cell activation markers. Moreover, potential signs of clinical activity of LAVA-051 were seen.

"The LAVA Therapeutics team is committed to transforming treatment for people living with cancer," said Stephen Hurly, president and chief executive officer of LAVA Therapeutics. "We are pleased with the encouraging findings so far from this clinical trial and dose escalation is continuing in the US and EU. I am excited about the potential of LAVA-051 as a novel therapy that may overcome the challenges associated with current T cell-engager approaches."

Details of the poster presentation session are as follows:

Abstract #: 2014
Abstract Title: LAVA-051, a Novel Bispecific Gamma-Delta T-Cell Engager (Gammabody), in Relapsed/Refractory MM and CLL: Pharmacodynamic and Early Clinical Data
Session Name: Cellular Immunotherapies: Early Phase and Investigational Therapies: Poster I
Session #: 704
Session Date: Saturday, December 10, 2022
Session Time: 5:30 p.m.–7:30 p.m. CT
Presenter: Arnon P. Kater, M.D., Ph.D., chairman of the Dutch/Belgium HOVON CLL working group and professor of translational hematology at the Amsterdam University Medical Center

A PDF copy of the presentation is available here.

About LAVA-051
LAVA-051 is a humanized Gammabody designed to activate both Vγ9Vδ2 (Vgamma9 Vdelta2) T cells and type 1 NKT cells to kill CD1d-expressing tumor cells. LAVA-051 consists of two single domain antibodies linked via a short five amino acid glycine-serine linker. One domain antibody recognizes the Vδ2 chain of the Vγ9Vδ2 T cell receptor, and the other domain antibody is specific for CD1d, a glycoprotein involved in the presentation of (glyco)lipid antigens to distinct T cell populations including type 1 NKT cells, that can be expressed on a wide range of hematologic malignancies, including chronic lymphocytic leukemia, multiple myeloma, and acute myeloid leukemia.

Kura Oncology Presents Updated Clinical Data from KOMET-001 Trial of Menin Inhibitor Ziftomenib at American Society of Hematology Annual Meeting

On December 10, 2022 Kura Oncology, Inc. (Nasdaq: KURA), a clinical-stage biopharmaceutical company committed to realizing the promise of precision medicines for the treatment of cancer, reported updated clinical data from KOMET-001, a Phase 1/2 trial of the Company’s potent and selective menin inhibitor, ziftomenib, including an encouraging safety and tolerability profile and clinical activity in patients with relapsed/refractory acute myeloid leukemia (AML) (Press release, Kura Oncology, DEC 10, 2022, View Source [SID1234625023]).

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!

These data are being featured during an oral session today at the American Society of Hematology (ASH) (Free ASH Whitepaper) Annual Meeting. A copy of the presentation is available on Kura’s website at www.kuraoncology.com/pipeline/#publications.

"NPM1-mutant and KMT2A-rearranged AML represent diseases of significant unmet need for which no approved targeted therapies exist," said Harry Erba, M.D., Ph.D., Director of the Leukemia Program at the Duke Cancer Institute. "Notably, NPM1-mutant disease accounts for approximately 30% of new AML cases annually. Although typically associated with a more favorable prognosis, the risk of relapse remains high after initial chemotherapy for NPM1-mutant AML, especially when other poor risk mutations such as FLT3 are present as well. Relapsed/refractory NPM1 mutated AML is associated with a poor prognosis. These data reported today demonstrate encouraging activity and manageable toxicity of ziftomenib in heavily pretreated AML patients with NPM1 mutations."

In the Phase 1a dose-escalation trial, ziftomenib demonstrated a wide therapeutic window and encouraging monotherapy activity in an all-comer population of 30 patients with relapsed/refractory AML, including a complete remission (CR) with no evidence of minimal residual disease (MRD) in an NPM1-mutant patient with DNMT3A and KMT2D co-mutations. The patient entered the trial having progressed through seven prior lines of therapy and remains on ziftomenib after two years.

In order to inform an optimal Phase 2 dose and in consultation with the U.S. Food and Drug Administration (FDA) and its Project Optimus initiative, Kura conducted a Phase 1b trial with two randomized expansion cohorts, each comprised of NPM1-mutant and KMT2A-rearranged AML patients. A total of 53 patients were ultimately treated in the Phase 1b trial: 17 at 200 mg and 36 at 600 mg. These patients were heavily pretreated and received a median of three prior lines of therapy (range 1-11), with the majority of patients having received prior venetoclax and a quarter having progressed after at least one prior stem cell transplant. As of the data cutoff on October 24, 2022, 10 of the patients treated at 600 mg remained on ziftomenib and 17 were still in follow-up. Median duration of response has not been reached.

Ziftomenib demonstrated optimal clinical benefit at 600 mg with a 30% CR rate (6/20) in patients with NPM1-mutant AML, compared to 17% (1/6) at 200 mg. Notably, four of the six NPM1-mutant patients who achieved a CR at 600 mg had IDH and/or FLT3 co-mutations. Overall, four of the seven patients with IDH co-mutations achieved a CR on ziftomenib. Of the five patients assessed for MRD at 600 mg, three were MRD negative.

Although meaningful clinical benefit was observed in patients with KMT2A rearrangements, symptoms of differentiation syndrome prevented most patients from receiving sufficient therapy to attain response criteria for CR or CR with partial hematologic recovery (CRh), and only one patient achieved a CR/CRh.

Continuous daily dosing of ziftomenib has been well tolerated. Reported adverse events most often were consistent with features of underlying disease. No evidence of drug-induced QTc prolongation was observed. Six patients discontinued due to adverse events; however, none of these were considered treatment related. The most common treatment-emergent adverse event observed was differentiation syndrome (DS), a known adverse event related to AML treatments that promote differentiation of AML cells. Of the 20 NPM1-mutant patients treated at 600 mg, four (20%) experienced DS; three of these events were less than Grade 3, and only one of these events (5%)was Grade 3. For KMT2A-rearranged patients, rates of DS were similar across doses, and approximately 38% of patients experienced DS; 25-30% of treated KMT2A-rearranged patients experienced Grade 3 or greater events.

Kura believes the higher incidence of DS observed in the KMT2A-rearranged patients is due to their much higher incidence of disease in extramedullary (outside of the bone marrow) sites, induced to differentiate by the high tissue penetrance demonstrated by ziftomenib preclinically. By combining ziftomenib with appropriate standards of care, the Company believes it can reduce this extramedullary disease burden and consequent DS symptoms, keep patients on ziftomenib therapy longer and drive superior treatment outcomes in patients with KMT2A-rearranged AML.

"We are excited by these data and the potential for ziftomenib to improve the lives of patients with acute leukemias," said Stephen Dale, M.D., Chief Medical Officer of Kura Oncology. "In addition to encouraging activity as a monotherapy in patients with NPM1 mutations, we believe ziftomenib is supportive of future combination strategies, with no predicted adverse drug-drug interactions and oral daily dosing that should enable convenient administration with standards of care. We believe that rational combination approaches will also help to mitigate DS in the KMT2A-rearranged population, maximizing patients’ time on therapy and ultimately leading to improved outcomes for patients in dire need of new therapeutic options."

Regulatory Update

Kura also announced that 600 mg has been determined as the recommended Phase 2 dose for ziftomenib in NPM1-mutant AML following a positive Type C meeting with the FDA. Agreement was also reached on key elements of a registration-enabling study design, and the Company is now preparing to initiate the Phase 2 registration-directed trial. Kura expects to dose the first patient in the first quarter of 2023, followed by a series of combination studies in frontline and relapsed/refractory AML that will prioritize development with venetoclax and FLT3 in combination.

"We believe our growing body of data support ziftomenib’s position as a potential best-in-class menin inhibitor," said Troy Wilson, Ph.D., J.D., President and Chief Executive Officer of Kura Oncology. "Through our team’s relentless hard work and dedication, we believe we have optimized the benefit-risk profile and paths forward in both the NPM1-mutant and KMT2A-rearranged subsets. The large number of patients treated in our Phase 1 experience provides a robust data set to support our Phase 2 registration-directed trial as well as combination studies. We have already begun the work needed to initiate both the first potentially registration-enabling study for ziftomenib as well as multiple studies in combination with standards of care and in earlier lines of therapy to realize the full potential of ziftomenib in the treatment of acute leukemias."

Investor Event

Kura’s management will host an investor event at 11:15 a.m. CT / 12:15 p.m. ET today, December 10, 2022, following the oral presentation of updated data from the KOMET-001 clinical trial at the ASH (Free ASH Whitepaper) Annual Meeting in New Orleans. The event will feature members of the Kura management team along with two investigators from the KOMET-001 clinical trial. A live webcast of the event will be available in the Investors section of Kura’s website at www.kuraoncology.com, with an archived replay available shortly after the conclusion of the event.

About Acute Myeloid Leukemia

AML is the most common acute leukemia in adults and begins when the bone marrow makes abnormal myeloblasts (white blood cells), red blood cells or platelets. Despite the many available treatments for AML, prognosis for patients remains poor. Approximately 50% of patients with AML who achieve a CR after induction therapy relapse within one to three years. NPM1-mutations are among the most common genetic alterations, representing approximately 30% of AML. While patients with NPM1-mutant AML have high response rates to frontline therapy, relapse rates are high and survival outcomes are poor. Median overall survival is only six months following relapse for NPM1-mutant patients. KMT2A-rearrangements are less frequent, representing approximately 5-10% of AML, however these patients have a poor prognosis with high rates of resistance and relapse following standard of care therapies. Currently, there are no approved therapies indicated for NPM1-mutant or KMT2A-rearranged leukemias.

About Ziftomenib

Ziftomenib (KO-539) is a novel, once-daily, oral investigational drug candidate targeting the menin-KMT2A/MLL protein-protein interaction for treatment of genetically defined AML patients with high unmet need. In preclinical models, ziftomenib inhibits the KMT2A/MLL protein complex and exhibits downstream effects on HOXA9/MEIS1 expression and potent anti-leukemic activity in genetically defined preclinical models of AML. Ziftomenib has received Orphan Drug Designation from the U.S. Food and Drug Administration for the treatment of AML. Additional information about clinical trials for ziftomenib can be found at kuraoncology.com/clinical-trials-komet.