Safety Profile of AMG 531 in Healthy Volunteers and in Thrombocytopenic Patients with Immune Thromobocytopenic Purpura.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1240-1240 ◽  
Author(s):  
Daniel E. Stepan ◽  
Elaine Sergis-Deavenport ◽  
Reggie Kelly ◽  
Jenny Christal ◽  
Chien-Feng Chen ◽  
...  

Abstract AMG 531 is a novel platelet-stimulating peptibody that targets the thrombopoietin (TPO) receptor, resulting in an increased production of platelets. AMG 531 lacks sequence homology to native TPO. To date, AMG 531 has been administered in two phase 1 studies in a total of 56 healthy volunteers and in three phase 2 studies, with the option to continue in an open-label extension study, in a total of 57 patients with immune thrombocytopenic purpura (ITP). In general, no deaths or detectable neutralizing antibodies have occurred across the AMG 531 development program. In the two phase 1 studies, AMG 531 increased platelet counts in a dose-response manner when administered as a single subcutaneous (SC) or intravenous (IV) dose of ≥ 1.0 μg/kg. The dynamics of the platelet response were as expected based on experience with other Mpl ligands. Single doses of AMG 531 ≤ 10.0 μg/kg were well tolerated. No deaths, serious or severe adverse events, or other events of clinical importance were reported at any dose administered (0.1, 0.3, 1.0, and 2.0 μg/kg SC; 0.3, 1.0, and 10.0 μg/kg IV). Commonly reported adverse events in healthy subjects receiving AMG 531 were mild to moderate headache (13% incidence in both studies), malaise/fatigue (4% and 6%), and various flu-like reactions. In the phase 2 studies in patients with ITP, AMG 531 was administered SC using weight-based dosing in all but one study. The maximum dose administered (in the extension study) was 23 μg/kg; most patients received doses of 3 to 9 μg/kg. AMG 531 has been administered as frequently as weekly for > 24 weeks. To date, three studies have been completed: two studies of two administrations on day 1 and 15, administered as unit (μg) doses in one study (N=16 and N=24, respectively), one placebo-controlled study of 6 weeks’ duration with weekly dosing (AMG 531 N=17; placebo N=4). One open-label extension (N=26) is ongoing. AMG 531 has been generally well tolerated in ITP patients. Most of the reported adverse events have been mild to moderate in severity; no dose-related trends have been observed. Across the four studies, the most commonly reported adverse events in patients receiving AMG 531 were headache (29%–54% incidence), contusion (15%–53%), and epistaxis (13%–41%). In the 6-week, weekly-dosing study, contusion and epistaxis were each reported for 50% and 41% of placebo-treated patients, respectively. Across the phase 2 studies, five patients treated with AMG 531 have experienced serious adverse events deemed as serious, unexpected, and reported as related to study drug. These include worsening of thrombocytopenia in three patients after completing treatment, headache and elevated LDH in one patient, and diffuse reticulin formation in the bone marrow reported as myelofibrosis in one patient. The reticulin formation is hypothesized to be due to an excessive accumulation of megakaryocytes in the bone marrow. AMG 531 was discontinued, and a follow-up bone marrow (after 3 months) showed improvement in reticulin. In summary, AMG 531 has been generally well tolerated and able to stimulate platelet production in a dose-response manner in healthy volunteers and ITP patients. Results suggest that both unit dosing and weight-based dosing provide a predictable platelet response. AMG 531 may represent a new treatment option for thrombocytopenic patients with ITP. Safety surveillance is ongoing to further establish the safety profile of AMG 531.

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 6597-6597 ◽  
Author(s):  
S. O’Brien ◽  
J. C. Byrd ◽  
T. J. Kipps ◽  
A. Forero-Torres ◽  
I. W. Flinn ◽  
...  

6597 Background: Lumiliximab, an anti-CD23 monoclonal antibody, had pharmacologic activity and an outstanding safety profile in a recently completed phase 1, single-agent study. Based on evidence of clinical activity, favorable safety profile, and preclinical data suggesting synergy with both fludarabine and rituximab, we initiated a combination study of lumiliximab with FCR in previously treated patients. Methods: Patients ≥18 years of age with relapsed CD23+ B-cell CLL were eligible for this open-label, dose-escalation, phase 1/2 study. Sample size was planned for ≤37 patients. Patients received either 375 mg/m2 or 500 mg/m2 of lumiliximab in combination with each 28-day cycle of FCR for 6 cycles. Primary objectives were to determine the safety profile, recommended phase 2 dose, and clinical activity of lumiliximab with FCR. Results: Accrual began in June 2004; 30 of the 31 patients were enrolled by December 2005; data are available for 28 patients. No dose-limiting toxicity was noted in the phase 1 component (375 mg/m2 dose, n=3, and 500 mg/m2 dose, n=6) and 500 mg/m2 was chosen for the phase 2 dose. All enrolled patients had progressive, symptomatic CLL as defined by NCI criteria, median 2 prior treatments (range, 1 to 9), median age 58, 64% males, 96% WHO performance status of ≤1. Seventeen patients experienced CTC Grade 3 or 4 adverse events (hematologic toxicity typically associated with FCR). Sixteen patients completed ≥3 cycles of treatment and were evaluated for response using NCI-WG criteria: 7 (44%) patients with confirmed complete responses (CRs); 1 with unconfirmed CR (pending marrow confirmation), 3 with partial responses (PRs), 4 with PRs awaiting confirmation, and 1 with disease progression. Twelve patients are not yet evaluable for response. Conclusions: Lumiliximab in combination with FCR is well tolerated and may enhance the activity of FCR for treatment of patients with progressive CLL after prior therapy. [Table: see text]


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. TPS7568-TPS7568
Author(s):  
Stephen Opat ◽  
Robert Marcus ◽  
Craig Anthony Portell ◽  
William Reed ◽  
Melannie Co ◽  
...  

TPS7568 Background: Bruton tyrosine kinase (BTK) plays a critical role in B-cell receptor signaling, mediating B-cell proliferation, migration, adhesion and survival. BTK inhibition has emerged as a strategy for targeting B-cell malignancies, including MZL. In preclinical studies, zanubrutinib was shown to be a potent, irreversible, highly specific BTK inhibitor with excellent oral bio-availability and favorable pharmacokinetic/pharmacodynamic properties. Clinical data to date have shown that complete and sustained 24-hour BTK occupancy is associated with durable responses and suggested that zanubrutinib is generally well tolerated with low rates of serious adverse events. Preliminary results from the MZL cohort enrolled in the open-label, multicenter, phase 1 study demonstrated responses in 7 of 9 patients for an overall response rate (ORR) of 78%. Cumulative safety data also showed that zanubrutinib monotherapy was associated with infrequent incidence of atrial fibrillation and major hemorrhage and infrequent drug discontinuation due to treatment-related adverse events. This study is designed to evaluate the safety and efficacy of zanubrutinib in patients with R/R MZL. Methods: This ongoing global phase 2, single-arm, open-label study is examining zanubrutinib monotherapy in patients with R/R MZL who have received one or more prior lines of systemic therapy. Patients are treated with oral zanubrutinib at 160 mg twice-daily until progressive disease, unacceptable toxicity, or withdrawal of consent. Eligible patients must have histologically confirmed MZL, have received prior anti-CD20 antibody therapy, and have measurable disease. Disease response is assessed per the 2014 Lugano Classification for non-Hodgkin lymphoma. The primary endpoint is ORR determined by independent review committee (IRC). Key secondary endpoints include ORR by investigator assessment, time to and duration of response, time to treatment discontinuation, progression-free survival (all determined by IRC and investigator assessments), and overall survival and safety. Recruitment is ongoing.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2676-2676 ◽  
Author(s):  
P.W. Wijermans ◽  
Michael Lübbert ◽  
Maria R. Baer ◽  
James L. Slack

CMML is a preleukemia that results in cytopenias, dysplastic morphology of bone marrow and peripheral blood cells, produces large numbers of ineffective monocytes, and is notoriously hard to treat. The recent revision of the WHO classification excludes CMML from the myelodysplastic syndromes (MDS), and reclassifies it as a myelodysplastic/myeloproliferative disease. Decitabine (Dacogen™) is a cytosine analog that reverses aberrant DNA hypermethylation, leading to re-expression of silenced tumor suppressor genes. The reclassification of CMML has led to a review of CMML patients treated with decitabine. Overall response rates (ORR) (complete response [CR] + partial response [PR]) from one pivotal phase 3 trial (D-0007) and two phase 2 trials (PCH 95-11, PCH 97-19) in the subset of patients with CMML receiving decitabine were reviewed. The two phase 2 trials were open-label and single-arm, with a recommended minimum of 4 treatment cycles and a maximum of 8 cycles. The phase 3 trial used a 1:1 randomized comparison of decitabine plus supportive care (SC) vs SC alone with a maximum of 10 cycles of therapy. The phase 3 trial study design dictated that patients be removed from therapy following 8 cycles of decitabine if CR was not achieved, and following 6 cycles in the absence of PR. Patients who maintained a CR for 2 cycles were removed from therapy. For consistency across trials, all decitabine-treated patients were evaluated using the phase 2 response criteria (CR was defined by normocellular bone marrow with <5% blasts, and normal Hgb, WBC, and platelet counts, and PR required 50% decrease in blast count, increases in Hgb by >1.5 mmol/L, WBC count by >1000, and platelet count by >50,000). A total of 28 patients diagnosed with CMML are included in this review. Similar demographics and disease characteristics were observed in all 3 studies, with an average age of 70.2 years and 71% of patients male. A baseline WBC of >20,000 was observed in 8/28 (29%) patients and baseline bone marrow blasts >5% were observed in 11/28 (39%) patients. All clinical responses were centrally reviewed. The ORR was 25% (14% CR + 11% PR). Hematologic improvement was observed in 11% of patients and stable disease in 39% of patients. The decitabine adverse event profile seen in CMML patients was similar to observations in other hematologic patient populations, with myelosuppression and related infectious complications. These data demonstrate encouraging activity for decitabine in CMML, and suggest that studies in other myeloproliferative diseases may be warranted.


Neurosurgery ◽  
2019 ◽  
Vol 66 (Supplement_1) ◽  
Author(s):  
Michael L Levy ◽  
John Crawford ◽  
Nabil Dib ◽  
Lev Verkh ◽  
Nikolai Tankovich ◽  
...  

Abstract INTRODUCTION Stroke is a leading cause of disability, affecting patients for years. We examined safety and preliminary efficacy for a cellular therapy targeting patients with chronic disabling stroke, a population for whom limited treatment options currently exist. METHODS Entry criteria included ischemic stroke > 6 mo prior, substantial disability (subject confined to wheelchair, had home-nursing care, or needed assistance with activities of daily living), and NIH Stroke Scale (NIHSS) score = 6 to 20. Enrollees received a single intravenous dose of allogeneic mesenchymal bone marrow cells. Phase 1 used a dose escalation design (3 tiers, n = 5 each). Phase 2 (n = 21) was an expanded safety cohort. The primary endpoint was safety over 1-yr. Secondary endpoints examined behavior, with a pre-specified focus at 6-mo. RESULTS In phase 1, each dose (0.5, 1.0, and 1.5 million cells/kg body weight) was found safe, so phase 2 subjects received 1.5 million cells/kg. At baseline, subjects (n = 36) averaged 4.2 ± 4.6 yr poststroke, age 61.1 ± 10.8 yr, NIHSS score 8.7 ± 3.0, and Barthel Index 65 ± 29. Two were lost to follow-up, one was withdrawn, and two died (unrelated to study treatment). There were 15 serious adverse events, none possibly or probably related to study treatment. Two adverse events were possibly related to study treatment, a UTI and IV site irritation. Treatment was safe based on serial exams, EKGs, laboratory tests, and pan-computed tomography scans. Change from baseline to 6-mo post-transfusion was significant for all 4 secondary endpoints: Barthel Index (6.8 ± 11.4 points, P = .002), NIHSS (−1.25 ± 1.7 points, P < .001), MiniMental Status Exam (1.8 ± 2.8 points, P < .001), and Geriatric Depression Scale (−1.6 ± 3.8 points, P = .015). At baseline 11.4% (4/35 subjects) had Barthel Index = 95 to 100 (favorable outcome); at 6-mo, 27.3% (9/33); by 12-mo, 35.5% (11/31). CONCLUSION Transfusion of mesenchymal bone marrow cells in patients with disabling chronic stroke was safe and significantly improved several behavioral measures. The initial results were potentially equivalent to or exceeded those of prior studies utilizing stereotactic implantation. These data support proceeding to a randomized, placebo-controlled study.


2018 ◽  
Vol 36 (6_suppl) ◽  
pp. 411-411 ◽  
Author(s):  
Yohann Loriot ◽  
Andrea Necchi ◽  
Se Hoon Park ◽  
Jesús García-Donas ◽  
Robert A Huddart ◽  
...  

411 Background: Although immune checkpoint inhibitors (ICI) have improved outcomes in some pts with platinum-resistant mUC, many pts (eg, pts with TCGA luminal 1 tumors, many of whom are FGFRa) may not benefit. ERDA, a pan-FGFR (1-4) inhibitor, demonstrated promising phase 1 activity: 11 partial responses among 24 FGFRa mUC pts. We report efficacy and safety of ERDA in the ongoing global open-label phase 2 study BLC2001 (NCT02365597). Methods: Pts had measurable mUC with specific FGFR2/ FGFR3 mutations or translocations per central lab Janssen assay, ECOG 0-2, and were chemorefractory (progressed during/following ≥ 1 line of prior systemic chemo or ≤ 12 mos of [neo]adjuvant chemo). Cisplatin-ineligible, chemo-naïve pts, and prior ICI treatment were allowed. Pts were randomized 1:1 to 28-d cycles of oral 6 mg/d continuous dosing (6 C) or 10 mg/d intermittent 7 d on/7 d off dosing (10 I) ERDA; the dose was further uptitrated if no significant treatment-related adverse events (TRAEs) were observed. The primary end point was ORR. Results: 78 pts received 6 C and 33 pts received 10 I (10 I cohort stopped early) ERDA. 31 pts in 6 C arm were further uptitrated. Across arms, 50% had ≥ 2 prior lines of therapy; 93% were chemorefractory. Confirmed ORRs (RECIST 1.1) were 35% and 24%, and disease control rates (CR+PR+SD) were 74% and 73% in the 6 C and 10 I arms, respectively. Adverse events (AEs) were manageable, and there were no treatment-related deaths (Table). Treatment is ongoing in 10 pts. Conclusions: ERDA (6 C or 10 I) has promising efficacy and tolerability in pts with FGFRa mUC. Based on these results and ERDA pharmacometric modeling, dosing was optimized at 8 mg/d (continuous), and this cohort is ongoing. Phase 3 study is planned. Clinical trial information: NCT02365597. [Table: see text]


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 736-736 ◽  
Author(s):  
Anne-Marie Duliege ◽  
Donald M. Arnold ◽  
Ralph Boccia ◽  
Michael Boxer ◽  
Nichola Cooper ◽  
...  

Abstract Introduction. The phase 3 randomized, placebo-controlled trials of the SYK inhibitor fostamatinib in adult patients with ITP were followed by an open-label extension (OLE) study to examine durability of response to fostamatinib and to allow patients in the placebo group in the phase 3 trials to crossover to fostamatinib. This abstract provides an additional year of data from the OLE study in patients with up to 3 years of fostamatinib treatment. Methods. Patients were adults with persistent or chronic ITP who had failed ≥1 prior therapy and had ≥3 platelet counts below 30,000/μL at screening. Patients initiated fostamatinib treatment at 100mg BID PO and increased to 150 mg BID based on tolerability and if platelets were <50,000/μL after 1 month. A stable platelet response was defined as a platelet count ≥50,000/μL that was maintained during the second 12-week period of fostamatinib treatment. The pre-specified analysis in the OLE study was the achievement and maintenance of a stable platelet response for 12 months. A post-hoc analysis was conducted on overall response, defined as achieving ≥1 platelet count of ≥50,000/µL within 12 weeks of beginning active treatment. All analyses excluded counts within 4 weeks of a rescue therapy. The data cutoff is 8 March 2018. Results. In the OLE study, 123 patients received fostamatinib, including 44 who had received placebo in the phase 3 studies. At baseline, the median age was 52 years (range 20-88), and 7% had persistent ITP (<12 months duration). The median duration of disease was 8.4 years, and the median platelet count was 16,000/μL at the start of treatment. Prior to the phase 3 studies, patients had tried a median of 3 (range 1-13) unique ITP treatments including splenectomy (35%; median 13 years earlier), corticosteroids (95%), immunoglobulins (53%), TPO-RA (47%), immunosuppressants (43%), and rituximab (32%). The median duration of fostamatinib exposure was 8.9 months (range 1.5-41.3). The median treatment compliance was 98%. At the time of analysis, 42 of 123 subjects (34%) continued fostamatinib treatment. Main reasons for discontinuation included lack of a platelet response after Week 12 (36%), study-specific adverse event (AE) (7%), and other AEs (7%). Of 27 patients with a stable response, 21 (78%) have maintained the response at Month 12 of fostamatinib treatment, and 15 (56%) at Month 24. At Month 12, median platelet count for the 49 subjects with data at that time point was 72,000/µL (range: 9000-333,000 µL). For the 32 subjects with data at Month 24, median platelet count was 80,500/µL (range: 7000-315,000/µL). See figure. An overall platelet response was achieved by 57/123 (46%) patients. AEs were reported by 95 (77%) patients and were mild/moderate in 92 (75%). See table. The most common AEs were diarrhea and hypertension, which were manageable with targeted treatment, fostamatinib dose modifications, or treatment withdrawal (5 patients withdrew due to diarrhea and none due to hypertension). Serious adverse events (SAE) were reported in 28 patients (23%), were considered unrelated to fostamatinib in 23 patients (19%) and included bleeding-related SAEs in 11 subjects (9%), thrombocytopenia in 6 subjects (5%), epistaxis in 3 (2%), sepsis in 2 (2%) and transaminases increased in 2 (2%). Adverse events were consistent with those reported during the placebo-controlled trials. Conclusion. In this open-label extension study of the two phase 3, placebo-controlled trials, 56% of subjects with a stable response maintained the response for ≥24 months. No new safety signals have been detected during long-term treatment of ITP with fostamatinib. Figure. Figure. Disclosures Duliege: Rigel: Employment, Equity Ownership. Arnold:UCB: Consultancy; Amgen: Consultancy, Research Funding; UCB: Consultancy; Amgen: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Bristol Myers Squibb: Research Funding; Novartis: Consultancy, Research Funding; Bristol Myers Squibb: Research Funding. Boccia:Amgen: Research Funding, Speakers Bureau; Pfizer: Consultancy; Abbvie: Speakers Bureau; AstraZeneca: Research Funding, Speakers Bureau; Celgene: Speakers Bureau; Sandoz: Consultancy; Genentech: Research Funding, Speakers Bureau; BMS: Research Funding. Boxer:Rigel: Speakers Bureau; Incyte: Speakers Bureau; AbbVie: Speakers Bureau. Cooper:Amgen, Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Hill:Novartis: Honoraria; Novartis: Honoraria. Zayed:Rigel: Employment, Equity Ownership. Tong:Rigel: Employment, Equity Ownership. Bussel:Uptodate: Honoraria; Protalex: Consultancy; Novartis: Consultancy, Research Funding; Prophylix: Consultancy, Research Funding; Momenta: Consultancy; Amgen Inc.: Consultancy, Research Funding; Rigel: Consultancy, Research Funding.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 3059-3059 ◽  
Author(s):  
Karen Kelly ◽  
Jeffrey R. Infante ◽  
Matthew H. Taylor ◽  
Manish R. Patel ◽  
Michael S. Gordon ◽  
...  

3059 Background: Avelumab is a fully human IgG1 anti–PD-L1 antibody with clinical activity in several tumor types. Pooled safety data from a large phase 1 trial in various tumors and a phase 2 trial in Merkel cell carcinoma (NCT01772004, NCT02155647) were analyzed to further characterize the safety profile of avelumab. Methods: Patients (pts) received avelumab 10 mg/kg 1-hour IV Q2W until progression, unacceptable toxicity, or withdrawal. Treatment-related adverse events (TRAEs) were graded by NCI CTCAE. In post hoc analyses, immune-related adverse events (irAEs) were identified via an expanded AE list and medical review, and infusion-related reaction (IRR) events were identified based on prespecified MedDRA terms, occurring within 1 day or related symptoms that resolved within 2 days of infusion. Results: In 1,738 pts analyzed (phase 1, n = 1,650; phase 2, n = 88) who received ≥1 dose of avelumab for a median of 12 weeks (range 2-138), the most common any grade TRAEs were fatigue (n = 307, 18%), IRR (n = 295, 17%), and nausea (n = 150, 9%). 177 pts (10%) had a grade ≥3 TRAE; most common were fatigue and elevated lipase (17 [1%] each). TRAEs led to discontinuation in 107 pts (6%). Four pts (0.2%) died due to a TRAE. Any grade irAEs occurred in 247 pts (14%), which were grade ≥3 in 39 pts (2%) and considered serious in 43 pts (2%). The most common any grade irAEs were thyroid disorder (n = 98, 6%) and rash (n = 90, 5%). Other irAEs (eg, colitis, hepatitis, pneumonitis, adrenal insufficiency, myositis) each occurred in < 2%. irAEs led to discontinuation in 34 pts (2%). IRR or related symptoms (eg, chills, pyrexia, hypersensitivity) occurred in 439 pts (25%), which were grade 3 in 9 pts (0.5%) and grade 4 in 3 pts (0.2%). An IRR occurred at first infusion in 79% and within first 4 doses in 99%; 63/439 pts (14%) had IRR recurrence in later cycles. IRR led to dose interruption in 152 (9%), infusion rate reduction in 124 (7%), and discontinuation in 35 pts (2%). Conclusions: This large pooled analysis confirms that avelumab has an acceptable safety profile. A minority of pts experienced a grade ≥3 TRAE or irAE and discontinuation due to TRAEs was uncommon. IRRs mostly occurred at first infusion and the rate of recurrence was low. Clinical trial information: NCT01772004, NCT02155647.


2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
Jonathan Barratt ◽  
Angelique Mittan ◽  
Suzanne Roy ◽  
Cailin Sibley ◽  
Colleen Stromatt ◽  
...  

Abstract Background and Aims IgA nephropathy (IgAN), the leading cause of primary glomerulonephritis, is an autoimmune disease with no approved treatments.1 Progression to end-stage-renal disease occurs in up to 45% of IgAN patients, requiring dialysis or kidney transplant to manage.2-4 A critical step in IgAN pathogenesis is the production of galactose-deficient IgA1 (Gd-IgA1) leading to the generation of anti-Gd-IgA autoantibodies and the formation of immune complexes that result in kidney inflammation and damage.5 A Proliferation-Inducing Ligand (APRIL), a soluble factor that regulates B cell differentiation, proliferation and survival of plasma cells, and IgA class-switching is elevated in patients with IgAN6, 7. IgAN patients with high plasma APRIL levels are reported as having higher levels of Gd-IgA1 and proteinuria and lower estimated glomerular filtration rates compared to those with lower plasma APRIL levels.7 BION-1301 is a novel humanized blocking antibody targeting APRIL. The primary objective of Study ADU-CL-19 is to assess the short-term safety and tolerability of BION-1301 in Healthy Volunteers (HV) and IgAN patients and to secondarily assess the short-term pharmacokinetics (PK), pharmacodynamics (PD), immunogenicity, and preliminary clinical activity of BION-1301. The primary objective of Study ADU-CL-24 is to characterize the long-term safety of BION-1301 in IgAN patients who completed treatment in ADU-CL-19 while secondarily assessing the long-term PK, PD, immunogenicity, and preliminary clinical activity of BION-1301. Method The Phase 1 study (ADU-CL-19; NCT03945318) comprises 3 parts. Parts 1 and 2 are double-blind, randomized, placebo-controlled single and multiple ascending dose designs in HV; both parts have been completed. Part 3 is an ongoing multicenter (US and UK), multiple-dose, two cohort design in approximately 20 patients with IgAN (10/cohort). Key eligibility criteria for Part 3 includes: (1) urine protein ≥0.5 g/24h or baseline UPCR ≥0.5 g/g, (2) stable/optimized dose of ACE-I/ARB or intolerant to ACE-I/ARB, and (3) biopsy-verified diagnosis of IgAN within the past 10 years. In Part 3, patients in Cohort 1 are receiving BION-1301 at 450mg every 2 weeks for 3 months. The dose and schedule for Cohort 2 will be determined by the Safety Review Team (SRT) based on data from the first 5 patients. After 3 months of treatment, patients continue safety follow-up for approximately 6 months unless deciding to enroll in the open-label extension (OLE) study, withdraw from the study, or are lost to follow up. Study ADU-CL-24 (NCT04684745) is a Phase 2 Open-Label Extension (OLE) of Study ADU-CL-19. Eligibility for the OLE study is restricted to those patients who completed at least 75% of their intended doses as well as the End of Treatment (EOT) visit in Study ADU-CL-19. Patients who enroll directly from the ADU-CL-19 EOT visit are not required to attend an additional screening visit. This is encouraged to maintain un-interrupted dosing. Once enrolled in the OLE, patients receive BION-1301 at the same dose and regimen as assigned in the parent study for up to 2 years. The dose, route, and regimen of BION-1301 may change after review of emergent safety, PK, PD and efficacy data by the SRT. Results Parts 1 and 2 of the Phase 1 study in HV are complete and the data were presented at ASN in 20208 Part 3 of the Phase 1 and the Phase 2 OLE study are ongoing, and interim results from the first dose cohort in IgAN patients are being presented in a separate poster at this meeting. Conclusion The design of the Phase 1 and Phase 2 OLE studies described in HVs and IgAN patients have enabled the generation of short- and long-term safety, PK, PD, immunogenicity and preliminary efficacy data for BION-1301. The Phase 1 short-term data will guide the design of future later-stage trials of BION-1301, while the long-term data from OLE will enable a greater understanding of the potential long-term risk/benefit profile of BION-1301 in IgAN patients.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2822-2822 ◽  
Author(s):  
Hagop M. Kantarjian ◽  
Elias J. Jabbour ◽  
Guillermo Garcia-Manero ◽  
Tapan M. Kadia ◽  
Courtney D. DiNardo ◽  
...  

Abstract Background: DFP-10917 is a nucleoside analog similar to cytarabine with a unique mechanism of action when administered at a low dose. Upon prolonged administration, DFP-10917 is converted to its nucleotide form and incorporated into tumor DNA, causing DNA strand breaks. Resulting G2/M phase-arrest by cell-checkpoint regulators ultimately leads to apoptosis of tumor cells. Methods: In the Phase 1, DFP-10917 was administered by 7-day continuous infusion (CI) followed by 21 days rest (Phase 1 Stage 1) or 14-day CI followed by 14 days rest (Phase 1 Stage 2) in patients (pts) with relapsed or refractory acute leukemia, to determine the maximum tolerated dose (MTD), recommended phase 2 dose (RP2D) and dose-limiting toxicities (DLT). Phase 2 was an open label, single arm, two-stage study of DFP-10917 administration at the RP2D using a 14-day CI in pts with relapsed or refractory acute myeloid leukemia (AML), or newly diagnosed AML pts ≥60 years not fit for intensive chemotherapy. A Simon 2-stage optimal design was used whereby if ≥1/10 pts responded (CR, CRi, CRp and PR) then an additional 19 pts were to be enrolled. Overall, if ≥4/29 pts respond, DFP-10917 warrants further investigation. A total of 39 AML pts were enrolled in Phase 1 with mean age 65 years (yrs), range 26-85 yrs. In Phase 1 Stage 1 (7-day CI, n=27) 26 pts received DFP-10917 at 8 escalating doses ranging from 4 to 35 mg/m2/day. One pt had CRi after the first treatment cycle of DFP-10917 at 6 mg/m2/day x 7 days. At 35 mg/m2, 1 pt experienced a cycle 1 DLT of grade 3 diarrhea. The starting dose for Phase 1 Stage 2 was calculated as 2/3 the cumulative 7-day dose at the MTD of 35 mg/m2/day divided by 14-day resulting in a dose of 10mg/m2/day×14 days. In Phase 1 Stage 2, (14-day CI, n=12), DFP-10917 at 10 mg/m2/day×14 days resulted in DLTs of prolonged hypo-cellularity, and the MTD/RP2D was defined as 6mg/m2/day×14days. Two pts had CR-one pt received 5 cycles of treatment and the second had continuous CR for over 22 cycles of DFP-10917 treatment. A total of 30 pts, all refractory or relapsed AML, were enrolled in Phase 2, mean age 70 yrs, range 45-88 yrs. Phase 2 pts were treated with DFP-10917 at 6 mg/m2/day x 14 days CI. An overall response rate (ORR) of 48% was observed (6 CRs including 2 transitioned to stem cell transplantation (SCT), 7 CRi including 3 transitioned to SCT, and 1 CRp). Among the 14 responding pts, 3 were refractory, 4 were salvage-1 and 7 were salvage-2 or greater. The median Overall Survival was 6.9 months and median Duration of Response was 3.5 months. The main drug-related AEs >grade 3 were: neutropenia (50%), thrombocytopenia (43%) or anemia (37%). Mild to moderate severity drug-related gastrointestinal AEs were frequently observed (43%) as well as fatigue (13%). No Phase 2 pts discontinued DFP-10917 treatment due to unacceptable drug-related toxicity. Conclusions: The RP2D of DFP-10917 was established at 6 mg/m2/day for 14-day CI with 14-day rest. In Phase 2, DFP-10917 demonstrated important anti-leukemia activity. The safety results of DFP-10917 continuous infusion indicated a tolerable safety profile in pts with relapsed or refractory AML. Overall, DFP-10917 shows promise as salvage treatment in pts with refractory or relapsed AML. Clinical trial information: NCT01702155 Disclosures Kantarjian: ARIAD: Research Funding; Bristol-Myers Squibb: Research Funding; Amgen: Research Funding; Pfizer Inc: Research Funding; Delta-Fly Pharma: Research Funding; Novartis: Research Funding. DiNardo:Novartis: Research Funding; Abbvie: Research Funding; Agios: Research Funding; Celgene: Research Funding; Daiichi Sankyo: Research Funding. Jain:Infinity: Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Seattle Genetics: Research Funding; Incyte: Research Funding; ADC Therapeutics: Consultancy, Honoraria, Research Funding; Celgene: Research Funding; Novartis: Consultancy, Honoraria; Genentech: Research Funding; Pharmacyclics: Consultancy, Honoraria, Research Funding; Abbvie: Research Funding; BMS: Research Funding; Servier: Consultancy, Honoraria; Novimmune: Consultancy, Honoraria. Iizuka:Delta Fly Pharma, Inc.: Employment. Jin:Delta Fly Pharma, Inc.: Employment. Zhang:Delta-Fly Pharma, Inc.: Employment.


Sign in / Sign up

Export Citation Format

Share Document