2021 ASH: Efficacy of Oral Decitabine/Cedazuridine (ASTX727) in the CMML Subgroup from ASCERTAIN Phase 3 Study

View Poster:

Efficacy of Oral Decitabine/Cedazuridine (ASTX727) in the CMML Subgroup from the ASCERTAIN Phase 3 Study

Abstract:

Background/Introduction: Chronic Myelomonocytic Leukemia (CMML) is an uncommon MDS/MPN overlap syndrome that has historically been included under the umbrella of myelodysplastic syndromes (MDS) for clinical trial and treatment. As a result, DNA methyltransferase inhibitors (DNMTi) such as decitabine or azacitidine have been the established standard of care for the treatment of CMML. The oral bioavailability of these agents has been limited due to rapid degradation by cytidine deaminase (CDA) in the gut and liver so treatment has required intravenous infusion or subcutaneous injections daily for 5-7 days every month (m) adding significant burden to older cancer patients due to daily time commitment and travel to treatment centers. In the context of pandemic SARS-CoV-2, parenteral therapy also increases contact with medical settings with increased infection risk. Oral decitabine 35 mg/cedazuridine 100 mg (ASTX727) is an oral fixed dose combination of decitabine and the CDA inhibitor cedazuridine that produced equivalent exposure (99%; 90% CI 93% to 106%) to IV decitabine 20 mg/m in a randomized cross-over study (Garcia-Manero et al, ASH 2019), and Median overall survival (mOS) for the entire study population in the ASCERTAIN study was approximately 32 months (Savona, 2021). Here, we present outcome data for this study for the enrolled subpopulation of patients with CMML.

Methods: We used a randomized cross over design in which patients were randomized in the first 2 cycles 1:1 to either Sequence A: (decitabine 35 mg/ cedazuridine 100 mg in Cycle 1 followed by IV decitabine at 20 mg/m in Cycle 2), or Sequence B: (IV decitabine in Cycle 1 followed by oral decitabine/cedazuridine in Cycle 2). We conducted an intra-patient comparison of decitabine PK (primary PK endpoint: decitabine AUC equivalence over 5 days of dosing). Cycles were repeated every 28 days (unless delays were needed). All patients received oral decitabine/cedazuridine in Cycles 3 and above until disease progression or unacceptable toxicity. Patients were eligible per the FDA-approved label of IV decitabine (MDS patients by FAB classification including CMML, or MDS IPSS Intermediate-1, 2 or high-risk patients). Clinical endpoints were best response according to International Working Group (IWG) 2006 response criteria, transfusion independence for at least 8 or 16 consecutive weeks, overall survival, and safety. Adverse events (AEs) were graded by Common Terminology Criteria for Adverse Events (CTCAE) v 4.03.

Results: Of the 133 patients enrolled and treated in ASCERTAIN, 16 (12%) had a diagnosis of CMML with demographics and as follows:  median age 71.5 years, 69%Male/31%Female, median weight 87kg (range 65-124), 25%ECOG 0, 75% ECOG 1. Population disease characteristics were: 19% poor or intermediate risk cytogenetics, with median baseline hemoglobin 90 g/L, neutrophils 1.27 X 109/L, platelets 84 x 109/L, bone marrow blasts 5%, with 38% RBC transfusion dependent. Patients received a median of 7 cycles of therapy (range 3-24). Treatment-emergent adverse events of CTCAE Grade 3 or higher in > 10% of patients, independent of relationship to ASTX727, were cytopenias (neutropenia [69%], thrombocytopenia [63%], anemia [56%], leukopenia [19%]), febrile neutropenia (31%), fatigue (13%). Two patients (12.5%) had Complete responses (CR), 8 (50%) had marrow CR ([mCR], including 3 (19%) with hematologic improvement (HI); Overall response rate (ORR) [CR + PR+ mCR + HI] was 75%. Of six patients with baseline RBC transfusion dependence 3 (50%) became transfusion independent. Leukemia-free survival was 28.2 months and after a median follow up of more than 33 months, median overall survival had not been reached. Two patients (13%) went on to Hematopoietic Stem Cell Transplant (HCT).

Conclusions: In the overall study, oral decitabine/cedazuridine delivered equivalent PK exposure to 5 days of IV decitabine 20mg/m2 with a resultant clinical activity safety and efficacy profile in CMML patients consistent with the published literature (e.g. Zeidan, et al 2017) and the Phase 2 experience. The use of oral decitabine/cedazuridine is a reasonable approach in CMML patients.

References:
Garcia-Manero, et al ASH 2019
Savona, et al, Int. MDS Symposium, 2021
Zeidan, et al, Cancer 2017: 3754-3762.

2021 ASH: Oral decitabine/cedazuridine in Patients with Lower Risk Myelodysplastic Syndrome: a Longer-Term Follow-Up of from the ASCERTAIN Study

View Poster
Oral decitabine/cedazuridine in Patients with Lower Risk Myelodysplastic Syndrome: a Longer-Term Follow-Up of from the ASCERTAIN Study

Abstract:
Background/Introduction: Lower-risk (IPSS low risk and Int-1) myelodysplastic syndromes (MDS) are typically treated supportively to address cytopenias. DNA methyltransferase inhibitors (DNMTi) such as azacitidine and decitabine (DEC) are FDA-approved for higher risk MDS patients (pts), and while the DEC USPI includes IPSS Int-1 pts, it is not widely used in this population. Approved intravenous (IV) or subcutaneous (SC) regimens require 5-7 days of treatment every month burdening older cancer pts due to daily travel and treatment time and may increase potential risk from pandemic SARS-CoV-2 infection. Because DNMTis are rapidly degraded by cytidine deaminase (CDA) in the gut and liver, oral availability has only been recently possible. A randomized study with CC-486, an oral formulation of azacitidine, in the Int-1 population showed a median overall survival (mOS) of approximately 17 months for both placebo and treated patients (Garcia-Manero, 2021). Oral DEC 35 mg/cedazuridine 100 mg (ASTX727) or DEC-C, is an oral fixed dose combination (FDC) of DEC and the CDA inhibitor cedazuridine (CED) resulting in equivalent exposure (99%; 90% CI 93% to 106%) to standard IV DEC 20 mg/m2 for 5 days in an intra-patient randomized cross-over study (Garcia-Manero et al, ASH 2019). Here, we present data on patients with lower risk MDS from that study.
Methods: We used a randomized cross over design with pts randomized 1:1 in the first 2 cycles to either Sequence A: (DEC 35 mg/ CED 100 mg in Cycle 1 and IV DEC at 20 mg/m2 in Cycle 2), or Sequence B (IV DEC in Cycle 1 and oral DEC/CED in Cycle 2). Cycles were repeated every 28 days unless delays were needed, and all patients received oral DEC-C in Cycles 3+ until disease progression or unacceptable toxicity. We conducted an intra-patient comparison of DEC PK (DEC AUC equivalence over 5 days of dosing). Pts were eligible as per the FDA-approved label of IV DEC (MDS pts by FAB classification including CMML, or MDS IPSS Intermediate-1, 2 or high-risk pts). Clinical endpoints were best response as assessed by an independent expert panel according to IWG 2006 response criteria, transfusion independence (TI), overall survival (OS), and safety.
Results: Of the 133 pts treated in ASCERTAIN, 69 had a diagnosis of lower-risk MDS (93% Int-1, 7% LR). Median age was 70.0 years (range 45-87), 65% were male, median weight was 84 kg (range 50-127), median baseline hematologic parameters were: hemoglobin 89 g/L (range 69.8-146.5), WBCs 1.50 X 109/L (range 0.11-7.1), platelets (plt) 86 x 109/L (range 5-703), bone marrow blasts 4% (range 0-18), cytogenetics: 7 (10.1%) poor-risk, 21 (30.4%) intermediate risk, 37 (53.6%) better-risk, 4 (5.7%) missing or not evaluable. 27(39%) of the pts were RBC transfusion dependent (TD) and 6 (9%) plt TD. 17 (25%) had received prior MDS treatment, 3% prior DNMTi. Pts received a median of 9 cycles of therapy (range 1-28). Treatment-emergent adverse events of CTCAE Gr 3 or higher in >10% of pts, independent of relationship to ASTX727, included cytopenias (neutropenia [59%], thrombocytopenia [58%], anemia [48%], leukopenia [26%]), febrile neutropenia (32%), and pneumonia (19%). Sixteen pts (23%) achieved Complete Response (CR), 18 (26%) had marrow CR (mCR), including 9 (13%) with hematologic improvement (HI). Overall Response rate (ORR; CR + PR+ mCR + HI) was 57%. Of those RBC or plt TD at baseline, 13 (48%) became RBC TI and 4 (67%) became plt TI. With approximately 32 months of median follow up, neither median leukemia-free survival (mLFS) nor mOS had been reached (Figure 1). Twelve pts (17%) went on to allogeneic stem cell transplant.
Conclusions: Oral decitabine/cedazuridine given as a FDC in MDS pts produced equivalent PK exposure to 20 mg/m2 IV DEC; in lower risk MDS pts with treatment indicated, the agent was generally well-tolerated with prolonged treatment and could result in mLFS and mOS which exceeds 32 months. This FDC and other dosing regimens of oral DEC-C should be further studied in this patient population.
References:
Garcia-Manero, et al, ASH 2019
Savona, et al, Int. MDS Symp. 2021
Garcia-Manero, et al,. J. Clin. Onc. 2021 39:13, 1426-1436

ASH 2021: A Phase 1 Study Evaluating ASTX727 (decitabine and cedazuridine) and Venetoclax in Combination Therapy in Newly Diagnosed AML Patients Unfit for Intensive Induction Chemotherapy

View Poster:

A Phase 1 Study Evaluating ASTX727 (decitabine and cedazuridine) and Venetoclax in Combination Therapy in Newly Diagnosed AML Patients Unfit for Intensive Induction Chemotherapy

Abstract:

Introduction: The combination of a DNA methyltransferase inhibitor (DNMTi; parenteral azacitidine or decitabine) with the BCL2 inhibitor venetoclax is a newly established standard-of-care regimen for patients with newly diagnosed acute myeloid leukemia (AML) ineligible to receive intensive induction chemotherapy (DiNardo et al, 2020). Replacing the parenteral DNMTi with an oral DNMTi with equivalent exposure may provide the benefit of reducing patient and caregiver burden of chronic parenteral therapy, and may help responding patients stay on treatment longer. ASTX727 (a fixed-dose combination of decitabine 35 mg and cedazuridine 100 mg) is an oral DNMTi that provides equivalent exposure to its parenteral DNMTi at standard dosing (Savona et al, 2020) and is under evaluation in combination with venetoclax as an all-oral regimen.

Methods: This is an ongoing Phase 1 study being conducted at 7 US medical centers (ClinicalTrials.gov NCT04657081). Newly diagnosed AML patients 75 years or older, or with comorbidities that preclude use of intensive induction chemotherapy are eligible. The primary objective is to evaluate the effect of ASTX727 on the PK of venetoclax. Key secondary objectives are to evaluate the effect of venetoclax on the PK of ASTX727, and to determine the safety and efficacy profile for the combination. For Cycle 2 and beyond, ASTX727 is administered orally daily on days 1-5 and venetoclax 400 mg is administered orally daily on days 1-28 of 28-day cycles. For Cycle 1, ASTX727 is given in the same dose schedule while venetoclax is given as a ramp-up on days 1 and 2 according to the venetoclax US prescribing information (USPI); therefore, the PK studies are conducted during Cycle 2. Delay of subsequent cycles and venetoclax dose modifications for hematologic toxicities and anti-fungal concomitant medications follow the venetoclax USPI. Response assessments are evaluated using the 2017 ELN criteria (Döhner et al, 2017) and the CRh criterion (complete response [CR] with partial hematologic recovery) defined as those patients achieving marrow CR criteria but not peripheral blood count criteria and demonstrating an absolute neutrophil count >500/μL and platelet >50,000/μL (Kantarjian et al, 2017).

Results: At the data cut-off date of August 1, 2021, 15 AML patients have enrolled and received study treatment. Median age is 78 years (range 66 – 84) and 9 (60%) are males. Of the 12 patients with data, 2 (17%), 6 (50%), and 4 (33%) patients are in the favorable, intermediate, and adverse risk ELN categories, respectively. Of the 15 dosed patients, 6 (40%) are diagnosed with AML with myelodysplasia-related changes. The median duration of exposure is 2 cycles (range 1-5) and 1.7 months (range 0.8-5.6). The most common adverse events (AEs) of Grade 2 or higher occurring in ≥10% of patients include neutropenia (5, 33%), febrile neutropenia (2, 13%), anemia (2, 13%), thrombocytopenia (2, 13%), vomiting (2, 13%), pneumonia (2, 13%), peripheral edema (2, 13%), hypertension (2, 13%) and vascular access complication (2, 13%). There are 17 serious AEs experienced in 7 patients; a grade 3 pneumonia and a grade 3 dysphagia are the only serious AEs assessed as related to ASTX727 and/or venetoclax and both events occurred in the same patient. Both AEs were part of the patient’s medical history. Two deaths have occurred to date: one patient due to rapidly progressive disease during Cycle 2 and one patient who achieved a best response of CRh transitioned to hospice due to progressive multiple myeloma. Of 9 patients with response assessments and evaluable data, 3 (33%) achieved CR and 4 (44%) achieved CRh as the best response for a composite CR+CRh rate of 78%. Preliminary PK data available from 9 patients show venetoclax exposures are not affected by coadministration of ASTX727and are similar to historical data. Exposures of decitabine and cedazuridine are consistent with the range seen in previous studies. Updated PK, safety, and efficacy data will be provided in December 2021.

Conclusions: A preliminary analysis of ASTX727 and venetoclax combination therapy in patients with newly diagnosed AML unfit for intensive induction chemotherapy demonstrate expected PK data, and a similar safety and efficacy profile to the approved combination therapy of a DNMTi and venetoclax. These preliminary data suggest that an all-oral regimen of a DNMTi in combination with venetoclax is feasible and should be investigated further.

2021 Virtual 16th MDS Conference: Prolonged survival observed in 133 MDS patients treated with oral decitabine/cedazuridine

View Poster
Prolonged survival observed in 133 MDS patients treated with oral decitabine/cedazuridine

Abstract: 

Background and aims

DNMTi are active in MDS treatment, however chronic parenteral therapy constitutes a burden for these patients, often elderly with co-morbid conditions. Oral decitabine(35 mg)/cedazuridine(100mg) given Days 1-5 every 28 days produces equivalent pharmacokinetic exposure (AUV) to 20 mg/m2 IV decitabine dosing (Garcia-Manero, ASH 2019).

Methods

This randomized, cross-over study enrolled MDS/CMML subjects appropriate to receive IV decitabine per the US label. Subjects either received IV decitabine or oral decitabine/cedazuridine, followed by the converse in C2, allowing intrapatient PK comparison. All subjects received oral decitabine/cedazuridine for subsequent cycles providing longer term safety and efficacy data.

Results

133 patients (IPSS HR: 16%, Int-1: 48%, Int-2: 20%, LR:4%, CMML:12%) were enrolled (US and Canada). The median age was 71y; 65% Male; 41% RBC and 9% platelet transfusion dependent, respectively. Subjects received a median of 9 cycles of treatment and 26% proceeded to HCT, typically after 4-6 cycles. The most common adverse events of thrombocytopenia, neutropenia, and anemia were consistent with expected AEs with parenteral DNMTi. Complete Response(CR) was achieved in 22%(95% CI 15.1,29.8), and overall response (CR + Partial Response + marrow CR + Hematologic Improvement) of 62% (95% CI 52.8, 69.9) was similar to seen with parenteral DNMTi.  K-M estimated mOS was 31.7 months.

Conclusion

Oral decitabine/cedazuridine is the only DNMTi demonstrating equivalent pharmacokinetic exposure to its IV form, and led to expected equivalent responses, with mOS of 31.7mo in this study. Additional studies using oral decitabine/cedazuridine in combination with new oral agents for hematological disease are warranted.

2021 Blood Virtual Congress: Clinical Efficacy and Safety of Oral Decitabine/Cedazuridine in 133 Patients with Myelodysplastic Syndromes (MDS) and Chronic Myelomonocytic Leukemia (CMML)

View Poster:
Clinical Efficacy and Safety of Oral Decitabine/Cedazuridine in 133 Patients with Myelodysplastic Syndromes (MDS) and Chronic Myelomonocytic Leukemia (CMML)

Abstract:

Aim: Confirm fixed dose combination (FDC) of oral decitabine/cedazuridine produces similar clinical activity vs. IV decitabine. Background: An oral FDC of 35 mg decitabine and 100 mg of CDA inhibitor cedazuridine has shown 99% (90% CI 93% to 106%) equivalent exposure to 20 mg/m2 IV decitabine in a randomized cross-over study(1). If oral decitabine/cedazuridine treatment produces similar clinical results its use may decrease the burden associated with chronic parenteral hypomethylating agent (HMA) therapy in MDS and CMML.

Methods: Randomized cross over design: 133 subjects treated in US or Canada.
Primary PK endpoint: decitabine AUC equivalence over 5 days of dosing. Efficacy endpoints: best response per IWG 2006, transfusion independence, OS, and safety. AEs were graded by CTCAE v 4.03.

Results:  Patient Characteristics: median age 71.0 years; 65% male; 88%MDS/12%CMML; 43% either RBC or platelet baseline transfusion-dependent; 25% poor-risk cytogenetics, and 42% baseline BM blasts >5%. Best Response: CR in 29/133 patients (22%), mCR with HI:17% (without HI 16%), and HI: 7.5%, for an overall objective response (CR+mCR+HI) of 62%; 26% proceeded to transplant. With median follow up of 24.7 months, median OS had not been reached. Treatment-Emergent AEs (Grade ≥3 regardless of causality): thrombocytopenia (61%), neutropenia (58%), anemia (51%), febrile neutropenia (32%), leukopenia (25%), and pneumonia (18%), of patients treated with oral decitabine/cedazuridine (excluding IV decitabine cycle).

Conclusion: Efficacy and safety from oral decitabine 35 mg/ cedazuridine 100 mg daily for 5 days every 28 days are consistent with historical clinical data from standard IV decitabine 20 mg/m2 daily for 5 days. Oral decitabine/cedazuridine is the only oral HMA with systemic exposure equivalent to its injectable drug. Further investigation of oral decitabine/cedazuridine in all-oral combination studies is warranted and underway.

References:  Garcia-Manero, et al, Blood 2019; 134 (Supplement_1): 846. doi: https://doi.org/10.1182/blood-2019-122980