Skip to main content

Retrospective analysis of arterial occlusive events in the PACE trial by an independent adjudication committee

A Correction to this article was published on 23 March 2022

This article has been updated

Abstract

Background

The phase 2 PACE (Ponatinib Ph+ ALL and CML Evaluation) trial of ponatinib showed robust long-term benefit in relapsed Philadelphia chromosome-positive (Ph+) leukemia; arterial occlusive events (AOEs) occurred in ≥ 25% of patients based on investigator reporting. However, AOE rates vary depending on the definitions and reporting approach used.

Methods

To better understand clinically relevant AOEs with ponatinib, an independent cardiovascular adjudication committee reviewed 5-year AOE data from the PACE trial according to a charter-defined process and standardized event definitions.

Results

A total of 449 patients with chronic myeloid leukemia (CML) or Ph+ acute lymphoblastic leukemia (ALL) received ponatinib (median age 59 y; 47% female; 93% ≥ 2 prior tyrosine kinase inhibitors (TKIs); median follow-up, 37.3 months). The adjudicated AOE rate (17%) was lower than the non-adjudicated rate (i.e., rate before adjudication; 25%). The only adjudicated AOE in > 2% of patients was peripheral arterial occlusive disease (4%). Exposure-adjusted incidence of newly occurring adjudicated AOEs decreased over time. Patients with multiple baseline cardiovascular risk factors had higher adjudicated AOE rates than those without risk factors.

Conclusions

This independent adjudication study identified lower AOE rates than previously reported, suggesting earlier overestimation that may inaccurately reflect AOE risk with ponatinib. This trial was registered under ClinicalTrials.gov identifier NCT01207440 on September 23, 2010 (https://clinicaltrials.gov/ct2/show/NCT01207440).

Background

Ponatinib, a pan-BCR::ABL1 inhibitor, is an orally active third-generation tyrosine kinase inhibitor (TKI) designed to potently inhibit BCR::ABL1 with or without any point mutation, including BCR::ABL1T315I [1]. In the pivotal phase 2 PACE (Ponatinib Ph+ ALL and CML Evaluation) trial, ponatinib demonstrated robust clinical activity with rapid, deep, and long-term responses, progression-free survival (PFS), and overall survival in patients with chronic-phase chronic myeloid leukemia (CP-CML), ≥ 90% of whom had failed treatment with ≥ 2 TKIs, regardless of the presence or absence of BCR::ABL1 mutations, including T315I [2, 3]. The 5-year results of the PACE trial confirmed the durability of these responses with a 5-year overall survival rate of 73% for CP-CML [3]. However, arterial occlusive events (AOEs) were reported by investigators in 25% in the overall population (serious AOEs, 20%) and 31% in the CP-CML population (serious AOEs, 26%) in the 5-year follow-up [3]. The exposure-adjusted incidence of newly occurring AOEs decreased from year 1 (15.8 patients with events per 100 patient-years in the total population) to year 5 (3.9 per 100 patient-years) [3]. The incidence of AOEs associated with ponatinib use has varied widely in subsequent reports. Two retrospective studies have reported an absence or very low incidence (6%) of AOEs [4, 5]. Other real-world studies have reported AOE rates ranging from 18 to 26% [6, 7]. Multiple factors may contribute to variability in reported AOE rates, including differences in patient populations, as well as differences in the clinical definitions used to identify and categorize vascular occlusive events. One of the most important factors is the lack of a standardized approach for defining and capturing AOEs with BCR::ABL1 TKIs.

The AOE incidence rate reported for PACE was based on a list of approximately 400 Medical Dictionary for Regulatory Activities (MedDRA) preferred terms developed by the sponsor. However, differences in the preferred terms used to define AOEs led to variability in AOE incidence rates. Some preferred terms included in the AOE analysis of PACE are highly sensitive for identification of potential AOEs but may not themselves indicate the occurrence of arterial occlusions, frequently including symptoms or descriptions rather than events; these include chest pain, cold hands, dysarthria, and poor peripheral circulation. This approach to characterize AOEs based on adverse event terms results in broadly capturing non-specific symptoms that may be associated with AOE rather than true AOEs and may thus overestimate the incidence of clinically meaningful events.

A clear understanding of clinically relevant AOE risk is imperative when characterizing the benefit-risk profile of ponatinib. Patients with CP-CML who become resistant to a second-generation BCR::ABL1 TKI, either with or without a BCR::ABL1 gene mutation, generally experience low response rates and poor survival if treated with another second-generation TKI [8, 9]. Importantly, ponatinib is the only currently available TKI effective in patients with the BCR::ABL1T315I mutation [3]. Therefore, the potential for improved survival and duration of response on ponatinib may outweigh the risk of AOEs [8, 9]. However, the lack of clear data regarding clinically meaningful AOEs has led to confusion about how to optimally use ponatinib to treat relapsed/refractory CML and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) and, in some instances, avoidance in patients who could potentially benefit. To provide a more accurate characterization of AOE incidence with ponatinib, an independent adjudication committee of experts was convened to retrospectively adjudicate all AOE reports in the PACE trial in a standardized, rigorous manner.

Methods

PACE trial design

The phase 2 PACE trial (ClinicalTrials.gov identifier: NCT01207440) enrolled adults with CML or Ph+ ALL whose disease was resistant or intolerant to dasatinib or nilotinib, or who had the BCR::ABL1T315I mutation regardless of prior TKI use [3]. All patients received ponatinib at a starting dose of 45 mg once daily (qd); dose reductions to 30 or 15 mg qd were applied per protocol (Table 1) to manage adverse events (AEs), or implemented proactively following recommendations from the sponsor in October 2013 in response to AOEs emerging as notable AEs. The trial has been completed; detailed methods are published [2, 3].

Table 1 Dose reduction recommendations (as of 2013)

Adjudication methods

All activities related to the adjudication of AOEs were conducted by ACI Clinical (Bala Cynwyd, PA), including the identification of an independent adjudication committee. ACI Clinical is a clinical research organization with expertise in Endpoint Adjudication and Data Monitoring Committees to support safety decisions around clinical development programs. ACI Clinical was contracted by the sponsor; adjudication activities were not part of the PACE trial.

Identification of AEs for adjudication

To ensure all relevant potential events were captured, the PACE AE dataset (449 patients with 12,224 AE records; extraction date: May 9, 2018) was searched using a comprehensive set of 604 preferred terms potentially relevant to AOEs that was developed by the sponsor (Table 2). This search strategy, which was more comprehensive than that used in initial analyses of the PACE trial, identified 181 patients and 455 AE records for adjudication (Fig. 1A). In addition, all patient deaths not attributable to disease progression by the clinical investigator were reviewed by the chair of the adjudication committee (described below) for identification of potential fatal AOEs. The adjudication committee identified 45 fatal events for review. In total, 202 patients and 490 events were submitted to the independent adjudication committee for review (Fig. 1A).

Table 2 List of 604 preferred terms used to identify AEs for adjudication
Fig. 1
figure 1

CONSORT flow diagram and process for adjudication of arterial occlusive events (AOEs). A CONSORT diagram: Identification of AOEs for review by the adjudication committee. B Adjudication process flow charts. AE adverse event, AC adjudication committee, AIM Applied Clinical Intelligence Information Management System, MedDRA Medical Dictionary for Regulatory Activities, PACE Ponatinib Ph+ ALL and CML Evaluation, PE pulmonary embolism, PVD peripheral vascular disease, VTE venous thromboembolism. aThe Adjudication Committee also reviewed any events included in the Cardiac Failure Standard MedDRA Query (SMQ) to determine whether any heart failure events were AOEs. bAOEs evaluated on the left panel excluded events evaluated in the right panel (stroke, DVT, and PE). cPer the charter, panel meetings were convened to discuss events for which a decision was not reached via independent voting. The quorum for panel meeting attendance was dependent on the type of event(s) to be discussed (i.e., cardiologist, neurologist, or vascular specialist)

An individual case package containing all available clinical information (including medical history) was created for each event and provided to the adjudication committee members for their review. If a patient experienced more than 1 event within 48 h, these events were adjudicated as potentially representing a single clinical event, unless the case evidence suggested they were independent events. Individual events occurring > 48 h apart were adjudicated as independent events. All data were from the clinical trial database that was in SAS format and structured in conformance to CDISC SDTM format; no other source material was available.

Adjudication procedure

An adjudication committee of academic research clinicians who are highly experienced in adjudication activities in cardiovascular trials was appointed by ACI Clinical. The adjudication committee of 5 independent academic experts (3 cardiologists, 1 vascular medicine specialist, and 1 vascular neurologist) retrospectively adjudicated suspected cases of arterial occlusive events in the PACE study. The committee followed a predefined process outlined in the adjudication charter developed by ACI clinical. The charter defined the responsibilities of the adjudication committee and the adjudication endpoints using established definitions developed by the 2014 American College of Cardiology (ACC)/American Heart Association (AHA) guideline [10], and the definitions for cardiovascular and stroke outcomes developed by the Standardized Data Collection for Cardiovascular Trials Initiative (SCTI) and the US Food and Drug Administration [11, 12]. All suspected AOEs identified in the PT search were assessed using the charter definitions (Table 3) for myocardial infarction; heart failure if attributed to an AOE, which may include coronary artery disease, arterial hypertension, cardiomyopathy, or myocardial infarction; hospitalization for unstable angina; stroke and other cerebrovascular events; and peripheral vascular disease. Any events meeting the criteria of these endpoints were considered adjudicated AOEs. Specific criteria were required (e.g., revascularization, change in cardiac biomarkers, diagnostic evidence as shown by computerized tomography scan, magnetic resonance imaging, etc.) to determine the presence of a clinical endpoint. The adjudication committee members were blind to ponatinib dose at the time of the event, whether dose modifications were made, and the investigator’s opinion on AE causality.

Table 3 Adjudication committee prespecified definitions of events

During the adjudication process, the committee reviewed all potential AOEs, as well as any AEs identified in a Cardiac Failure Standard MedDRA Query (SMQ), to determine whether any heart failure events were AOEs. Two members of the adjudication committee independently evaluated whether an individual case met the prespecified event definitions (Fig. 1B). If agreement between 2 members was not reached for cases of AOEs or heart failure, the case was reviewed by a third cardiologist adjudication committee member; if agreement was not reached with 3 votes, the case was reviewed at a panel meeting. If agreement was not reached for cases of stroke, deep vein thrombosis, pulmonary embolism, and peripheral vascular disease, the case was discussed at a panel meeting with the appropriate neurologist and/or vascular specialist member(s). All fatal events were decided by consensus of adjudicators.

Events that met one of the charter-defined endpoint definitions were further categorized depending on the event type (e.g., myocardial infarction, peripheral arterial occlusive disease, deep vein thrombosis, etc.). Non-adjudicated AOEs that were recorded as symptoms (e.g., "non-cardiac chest pain" or "claudication") with a low severity level and no accompanying changes in medication or hospitalization were adjudicated to not be AOEs unless they had an anatomic diagnosis provided (e.g., "severe superficial femoral artery stenosis"). If the term "infarction" was provided for stroke events, the adjudicators categorized the event as ischemic stroke. Revascularization was not always clearly reported by investigators.

Statistics

Exposure-adjusted AOE rates were calculated as: (number of first events in interval)/(total exposure for interval in patient-years) × 100. The relative risk of serious AOEs was analyzed by baseline risk category in patients from the safety population for whom all baseline risk categories were available. Risk categories included commonly recognized cardiovascular risk factors for which data were collected (arterial hypertension, hypercholesterolemia, diabetes mellitus, and obesity), and history of heart disease (non-ischemic or ischemic).

Results

Patient disposition and baseline characteristics

Patient disposition and baseline characteristics in the PACE trial have been published [2, 3]. A total of 449 patients, including 270 CP-CML patients, 85 accelerated-phase (AP) CML patients, 62 blast-phase (BP) CML patients, and 32 Ph+ ALL patients, were enrolled between September 2010 and October 2011. Baseline characteristics are summarized in Table 4. Among all 449 patients, the median age was 59 years and 53% of patients were male. Most (93%) patients had received 2 or more prior TKIs. At baseline, 53% of patients had arterial hypertension, 49% had hypercholesterolemia, and 24% had BMI ≥ 30 kg/m2. Forty-three percent of patients had a baseline history of non-ischemic cardiac disease, and 23% had a history of ischemic cardiovascular disease. Safety data reviewed by the adjudication committee reflect data collected as of February 6, 2017, with median follow-up of 37.3 months for all patients and 56.8 months (range 0.1–73.1 months) for CP-CML patients.

Table 4 Baseline characteristics and disposition at end-of-study3

Adjudication results

Rates of adjudicated AOEs were lower than rates of non-adjudicated AOEs (Fig. 2A). Overall, 17% (78/449) of patients had adjudicated AOEs compared with 25% (111/449) with non-adjudicated AOEs. Most patients with serious AOEs were adjudicated as having serious AOEs (20% [90/449] non-adjudicated vs. 16% [74/449] adjudicated). Most (95% [74/78]) patients with adjudicated AOEs had serious AOEs. In CP-CML patients, rates of adjudicated AOEs (21% [57/270]) were also lower than rates of non-adjudicated AOEs (31% [84/270]); 95% [54/57] of CP-CML patients with adjudicated AOEs had serious AOEs. The rates of AOEs by AOE type (i.e., cardiovascular, cerebrovascular, and peripheral vascular) are presented for all patients in Table 5 and for CP-CML patients in Table 6.

Fig. 2
figure 2

Arterial occlusive event (AOE) rates with ponatinib. A Rates of non-adjudicated and adjudicated AOEs. B Exposure-adjusted incidence of newly occurring arterial occlusive events (AOEs) by year (all patients). Later intervals excluded patients with prior events. Non-adjudicated values were published previously [3]. C Incidence of AOEs (adjudicated) by number of baseline risk factors (all patients). Risk factors included arterial hypertension, hypercholesterolemia, obesity, diabetes mellitus, non-ischemic cardiac disease, and ischemic disease. D Overall survival (OS) in chronic-phase chronic myeloid leukemia (CP-CML) patients with and without AOEs

Table 5 Rates of non-adjudicated and adjudicated AOEs by type in the total population (n = 449)
Table 6 Rates of AOEs non-adjudicated and adjudicated AOEs in CP-CML patients (n = 270)

The most common non-adjudicated and adjudicated AOEs and serious AOEs are summarized in Table 7. The most common (> 2%) non-adjudicated AOEs were angina pectoris (6%; 28/449), peripheral arterial occlusive disease (5%; 22/449), MI (4%; 18/449), coronary artery disease (3% [14/449]). The only adjudicated AOE reported in > 2% of patients was peripheral arterial occlusive disease (4% [16/449]). Non-adjudicated AOEs that were most commonly adjudicated as not AOEs were angina pectoris, non-cardiac chest pain, and chest pain, as these events were often recorded as symptoms (e.g., "non-cardiac chest pain" or "claudication") or presumptive diagnoses with a low severity level and no accompanying changes in medication or hospitalization.

Table 7 Arterial occlusive events (AOEs) in ≥ 2.0% of patients (n = 449)

The exposure-adjusted incidence of adjudicated AOEs (8.9 patients with events per 100 patient-years) and serious AOEs (8.4 patients with events per 100 patient-years) was lower than the exposure-adjusted incidence of non-adjudicated AOEs (11.3 and 9.2 per 100 patient-years, respectively). The exposure-adjusted incidence of newly occurring AOEs decreased over time (Fig. 2B). The median time to onset of the first adjudicated AOE was 14.1 months (range: 0.1 to 49.5; Table 8).

Table 8 Time to onset of adjudicated AOEs

Resolution of AOEs, dose modifications, and discontinuations

Among the 78 patients with an adjudicated AOE, events resolved in 51 patients. Among 43 patients with just one AOE, 74% (32/43) had resolution of the event; 35 patients had multiple AOEs recorded, with 54% (19/35) patients having resolution of all the events. Most patients continued ponatinib after the AOE, including 36 patients (46%) who continued ponatinib without dose modification and 27 patients (35%) who had their doses reduced and/or interrupted after the event (Table 9). Seven patients (9%) discontinued ponatinib due to an adjudicated AOE. Rates of dose modifications following AOEs are summarized in Table 9.

Table 9 Ponatinib dose modifications following non-adjudicated and adjudicated arterial occlusive events (AOEs)a

Risk factor analysis

The most common baseline risk factors in patients who developed an AOE were arterial hypertension and hypercholesterolemia (Table 10). Patients with adjudicated AOEs also had higher rates of concomitant use of antihypertensive medications, platelet aggregation inhibitor medications, and anti-diabetic agents compared with patients who did not have AOEs (Table 11).

Table 10 Prevalence of baseline risk factors by adjudicated AOE and serious AOE status
Table 11 Concomitant medication use by adjudicated AOE and serious AOE status

The incidence of adjudicated AOEs by number of baseline risk factors (including arterial hypertension, hypercholesterolemia, obesity, diabetes mellitus, non-ischemic cardiac disease, and ischemic disease) is shown in Fig. 2C. The rate of adjudicated AOEs was 13% (24/189) among patients with 1–2 risk factors, and 29% (52/180) among patients with 3 or more risk factors. Of the 80 patients without any risk factors at baseline, only 2 (3%) had an AOE.

Fatal AOEs

Separate adjudication of deaths revealed that 11 adjudicated AOEs were associated with death. These included 2 cases of cardiac arrest and 1 each of the following: bradycardic arrest, cardiac failure, intracranial hemorrhage, worsening of congestive heart failure, superior mesenteric artery occlusion, hemorrhagic cerebral infarction, congestive heart failure, ischemic stroke, and acute anterior myocardial infarction. Nine of the 11 patients with AOEs associated with death had a history of cardiovascular events and/or cardiovascular risk factors recorded at baseline (Table 12). The long-term survival of patients with adjudicated AOEs was similar to survival of patients without AOEs (Fig. 2D).

Table 12 Fatal AOEs and patient baseline characteristics

Discussion

In this study, adjudication of AOEs by an independent committee of experts allowed for a clinically meaningful description of AOEs associated with ponatinib, which can help to inform health care providers and patients of safety risks in an accurate and objective manner. The search that identified potential AOEs for adjudication was broader (based on 604 MedDRA terms related to vascular ischemia or thrombosis) than that initially used to calculate non-adjudicated AOE rates in the PACE trial (400 MedDRA terms) [3]. Based on 5-year follow-up of the PACE trial, the adjudicated AOE rate (17%) was lower than the non-adjudicated AOE rate (25%) [3]. Although the majority of adjudicated AOEs were serious, 81% of patients with AOEs continued on ponatinib (35% with dose modifications), the benefit of the drug was felt to outweigh the risk of the AOEs. Although vascular occlusive events were rarely reported during the initial development of second-generation BCR::ABL1 TKIs, a meta-analysis found that these events occurred in 5.9% of patients with CML treated with these agents, including bosutinib, dasatinib, nilotinib, and ponatinib [13]. In another review of prospective trials of patients treated with TKIs, including imatinib, nilotinib, dasatinib, and ponatinib, overall incidence of CV events was 45% (range, 41–63%) [14]. Accordingly, a high level of vigilance is indicated to recognize this potential complication of TKI therapy.

Notably, although concern existed around the potential for increasing AOE rates with long-term dosing, as seen with AEs related to other TKIs [15,16,17,18] the exposure-adjusted incidence of newly occurring adjudicated AOEs decreased over time on ponatinib, suggesting that the toxicity of ponatinib may not increase with longer treatment duration.

Patients with adjudicated AOEs were more likely to have multiple baseline cardiovascular risk factors (e.g., ischemic cardiac disease, arterial hypertension, hypercholesterolemia, and diabetes mellitus), and only 2 patients had an adjudicated AOE without any cardiovascular risk factors. These observations align with those of previous studies [6, 19]. It is important to identify and manage cardiovascular risk factors before and during therapy with ponatinib or other TKIs [20,21,22]. In PACE, 80% of CP-CML patients were resistant to dasatinib or nilotinib, and 24% had the BCR::ABL1T315I resistance mutation [3]. Among CP-CML patients, estimated 5-year PFS and OS rates were 53% and 73%, respectively [3]. Data for overall survival in patients with and without adjudicated AOEs suggest that the risk of AOE-related death did not substantially impact survival, with disease-related death being the main driver of the OS curve. This underscores the need for providers to fully understand the therapeutic profile of ponatinib and consider its use when the potential benefits outweigh the risks for a given patient.

This study reinforces the importance of proper assessment of cardiovascular AEs to ensure accurate estimation of cardiovascular risk. The conventional processes of AE reporting and causality assessment may need to be re-assessed to avoid pitfalls associated with over- or under-reporting of AOEs, both of which may adversely affect patient care [23, 24]. Formal adjudication of events is a mainstay for development programs in other therapeutic areas such as diabetes mellitus [25, 26] and cardiology. A better understanding of the AOE risk associated with TKI therapy is a prime example of where formal adjudication is critical because accurate knowledge of risks is crucial before prescribing any TKI. The potential benefits of effective BCR::ABL1 TKI treatment, even with accompanying AEs, may outweigh the potential risks of progression-related mortality in patients with CP-CML and Ph+ ALL receiving second- or third-line therapy. This is particularly true for patients such as those with the BCR::ABL1T315I mutation who may have limited treatment options [27]. Understanding the true incidence of the most significant events is a central element in properly assessing the benefit-risk ratio of an intervention. All later-generation TKIs are associated with risk of cardiovascular AEs [28], and the results of the formal adjudication process suggest the risk of these events with ponatinib may not be dissimilar to the event rates seen with some second-generation BCR::ABL1 TKIs [16,17,18].

A noteworthy finding in our analysis is that the exposure-adjusted incidence of newly occurring adjudicated AOEs decreased over time on ponatinib. These results are reassuring that the rate of new AOEs may not increase with longer duration of ponatinib treatment. Furthermore, patients with positively adjudicated AOEs were much more likely to have baseline cardiovascular risk factors (e.g., arterial hypertension, hypercholesterolemia, diabetes mellitus) or established cardiovascular disease; of those patients without any cardiovascular risk factors only 2 had a subsequent AOE. These results may provide clinical guidance with respect to the approach to use of ponatinib in patients at risk for an AOE. The ongoing phase 2 OPTIC trial (ClinicalTrials.gov Identifier: NCT02467270) is using a response-based dose reduction protocol approach to evaluate the optimal ponatinib dosing regimen for maximizing efficacy while mitigating toxicity. Results show that higher doses of ponatinib were associated with increased incidence of AOEs, with exposure-adjusted treatment-emergent AOE rates of 5.6%, 3.6%, and 2.1% for the 45-mg, 30-mg, and 15-mg cohorts, respectively [29]. However, the benefit differential was considerably larger with a starting dose of 45 mg, which was associated with a 26.3 percentage-point improvement in the response rate compared with a 15-mg starting dose (51.6% vs. 25.3%) [29]. Overall, the study indicated the best risk/benefit ratio when the 45-mg starting dose was reduced to 15 mg upon achievement of response (BCR::ABL1IS transcript levels ≤ 1%) [29].

This retrospective study has strengths and limitations. The adjudication methodology provided a comprehensive and objective approach for characterizing AOE risk. A limitation is that only data from the clinical trial database were available. Prospective implementation of this strategy, as is being done in 2 ongoing trials, OPTIC and Ponatinib-3001 (NCT03589326), will overcome this challenge and add further value to the methodology and strength to the conclusions. In OPTIC, an independent cardiovascular endpoint adjudication committee is reviewing AOEs as they are reported using source documentation including cardiovascular workup (e.g., echocardiograms, electrocardiograms, biomarkers), hospitalization records, and any cardiovascular examinations performed.

Conclusions

Independent reconsideration of AOEs by an expert adjudication committee showed lower rates of clinically relevant AOEs overall (17% vs. 25%) and serious AOEs (16% vs. 20%) than were originally reported in the PACE trial, suggesting an earlier possible overestimation that may not accurately reflect the AOE risk with ponatinib. The incidence of exposure-adjusted newly occurring AOEs decreased over time during ponatinib treatment. Improved understanding of the AOE profile with ponatinib and risk factors for AOEs can help guide decisions around TKI treatment. Results from the OPTIC study support a novel ponatinib treatment regimen of a 45-mg starting dose reduced to 15 mg upon achievement of response, maximizing response while minimizing toxicity [29].

Availability of data and materials

The data sets, including the redacted study protocol, redacted statistical analysis plan, and individual participants data supporting the results reported in this article, will be made available within three months from initial request, to researchers who provide a methodologically sound proposal. The data will be provided after its de-identification, in compliance with applicable privacy laws, data protection and requirements for consent and anonymization.

Change history

Abbreviations

ACC:

American College of Cardiology

AE:

Adverse events

AHA:

American Heart Association

ALL:

Acute lymphoblastic leukemia

AOE:

Arterial occlusive event

AP:

Accelerated-phase

BP:

Blast-phase

CML:

Chronic myeloid leukemia

CP-CML:

Chronic-phase chronic myeloid leukemia

MeDRA:

Medical Dictionary for Regulatory Activities

PACE:

Ponatinib Ph+ ALL and CML Evaluation

PFS:

Progression-free survival

Ph+:

Philadelphia chromosome positive

qd:

Once daily

SCTI:

Standardized Data Collection for Cardiovascular Trials Initiative

SMQ:

Cardiac Failure Standard MedDRA Query

TKI:

Tyrosine kinase inhibitor

References

  1. O’Hare T, Shakespeare WC, Zhu X, et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell. 2009;16:401–12.

    Article  CAS  Google Scholar 

  2. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. N Engl J Med. 2013;369:1783–96.

    Article  CAS  Google Scholar 

  3. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. Ponatinib efficacy and safety in Philadelphia chromosome–positive leukemia: final 5-year results of the phase 2 PACE trial. Blood. 2018;132:393–404.

    Article  CAS  Google Scholar 

  4. Breccia M, Abruzzese E, Castagnetti F, et al. Ponatinib as second-line treatment in chronic phase chronic myeloid leukemia patients in real-life practice. Ann Hematol. 2018;97:1577–80.

    Article  CAS  Google Scholar 

  5. Shacham-Abulafia A, Raanani P, Lavie D, et al. Real-life experience with ponatinib in chronic myeloid leukemia: a multicenter observational study. Clin Lymphoma Myeloma Leuk. 2018;18:e295–301.

    Article  Google Scholar 

  6. Caocci G, Mulas O, Abruzzese E, et al. Arterial occlusive events in chronic myeloid leukemia patients treated with ponatinib in the real-life practice are predicted by the Systematic Coronary Risk Evaluation (SCORE) chart. Hematol Oncol. 2019;37:296–302.

    Article  CAS  Google Scholar 

  7. Heiblig M, Rea D, Chretien ML, et al. Ponatinib evaluation and safety in real-life chronic myelogenous leukemia patients failing more than two tyrosine kinase inhibitors: the PEARL observational study. Exp Hematol. 2018;67:41–8.

    Article  CAS  Google Scholar 

  8. Ashaye AO, Thomas C, Dalal M, et al. Treatment of newly diagnosed Philadelphia chromosome positive acute lymphoblastic leukemia using tyrosine kinase inhibitors in combination with chemotherapy: a patient-centered benefit-risk assessment [abstract]. Blood. 2020;136(Suppl 1):20–1.

    Article  Google Scholar 

  9. Kantarjian HM, Deininger MW, Abruzzese E, et al. Efficacy and safety of ponatinib (PON) in patients with chronic-phase chronic myeloid leukemia (CP-CML) who failed one or more second-generation (2G) tyrosine kinase inhibitors (TKIs): analyses based on PACE and Optic [abstract]. Blood. 2020;136(suppl 1):43–4.

    Article  Google Scholar 

  10. Hicks KA, Hung HMJ, Mahaffey KW, et al. Standardized definitions for cardiovascular and stroke endpoint events in clinical trials. 2014. Available at https://www.cdisc.org/system/files/all/standard/Draft%20Definitions%20for%20CDISC%20August%2020%2C%202014.pdf. Accessed 14 Nov 2019.

  11. Hicks KA, Tcheng JE, Bozkurt B, et al. 2014 ACC/AHA key data elements and definitions for cardiovascular endpoint events in clinical trials: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Data Standards (writing committee to develop cardiovascular endpoints data standards). J Am Coll Cardiol. 2015;66:403–69.

    Article  Google Scholar 

  12. Hicks KA, Mahaffey KW, Mehran R, et al. 2017 cardiovascular and stroke endpoint definitions for clinical trials. Circulation. 2018;137:961–72.

    Article  Google Scholar 

  13. Douxfils J, Haguet H, Mullier F, Chatelain C, Graux C, Dogne JM. Association between BCR-ABL tyrosine kinase inhibitors for chronic myeloid leukemia and cardiovascular events, major molecular response, and overall survival: a systematic review and meta-analysis. JAMA Oncol. 2016;2:625–32.

    Article  Google Scholar 

  14. Jain P, Kantarjian H, Boddu PC, et al. Analysis of cardiovascular and arteriothrombotic adverse events in chronic-phase CML patients after frontline TKIs. Blood Adv. 2019;3:851–61.

    Article  CAS  Google Scholar 

  15. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib versus imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30:1044–54.

    Article  CAS  Google Scholar 

  16. Kantarjian HM, Hughes TP, Larson RA, et al. Long-term outcomes with frontline nilotinib versus imatinib in newly diagnosed chronic myeloid leukemia in chronic phase: ENESTnd 10-year analysis. Leukemia. 2021;35:440–53.

    Article  CAS  Google Scholar 

  17. Rea D, Mirault T, Raffoux E, et al. Identification of patients (pts) with chronic myeloid leukemia (CML) at high risk of artery occlusive events (AOE) during treatment with the 2nd generation tyrosine kinase inhibitor (TKI) nilotinib, using risk stratification for cardiovascular diseases (CVD) [abstract]. Blood. 2013;122:2726.

    Article  Google Scholar 

  18. Valent P, Hadzijusufovic E, Schernthaner GH, Wolf D, Rea D, le Coutre P. Vascular safety issues in CML patients treated with BCR/ABL1 kinase inhibitors. Blood. 2015;125:901–6.

    Article  CAS  Google Scholar 

  19. Dorer DJ, Knickerbocker RK, Baccarani M, et al. Impact of dose intensity of ponatinib on selected adverse events: multivariate analyses from a pooled population of clinical trial patients. Leuk Res. 2016;48:84–91.

    Article  CAS  Google Scholar 

  20. Cortes J. How to manage CML patients with comorbidities. Blood. 2020;136:2507–12.

    Article  CAS  Google Scholar 

  21. Hochhaus A, Baccarani M, Silver RT, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 2020;34:966–84.

    Article  CAS  Google Scholar 

  22. Steegmann JL, Baccarani M, Breccia M, et al. European LeukemiaNet recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukaemia. Leukemia. 2016;30:1648–71.

    Article  CAS  Google Scholar 

  23. Russo Rossi A, Breccia M, Abruzzese E, et al. Outcome of 82 chronic myeloid leukemia patients treated with nilotinib or dasatinib after failure of two prior tyrosine kinase inhibitors. Haematologica. 2013;98:399–403.

    Article  Google Scholar 

  24. Ongoren S, Eskazan AE, Suzan V, et al. Third-line treatment with second-generation tyrosine kinase inhibitors (dasatinib or nilotinib) in patients with chronic myeloid leukemia after two prior TKIs: real-life data on a single center experience along with the review of the literature. Hematology. 2018;23:212–20.

    Article  CAS  Google Scholar 

  25. Guidance for Industry Diabetes Mellitus—evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes. 2008. Available at https://www.fda.gov/media/71297/download. Accessed 1 Apr 2021.

  26. US Food and Drug Administration. Type 2 diabetes mellitus: evaluating the safety of new drugs for improving glycemic control; draft guidance for industry; availability. Fed Regist. 2020;85:13903–5.

    Google Scholar 

  27. Moslehi JJ, Deininger M. Tyrosine kinase inhibitor-associated cardiovascular toxicity in chronic myeloid leukemia. J Clin Oncol. 2015;33:4210–8.

    Article  CAS  Google Scholar 

  28. Medeiros BC, Possick J, Fradley M. Cardiovascular, pulmonary, and metabolic toxicities complicating tyrosine kinase inhibitor therapy in chronic myeloid leukemia: strategies for monitoring, detecting, and managing. Blood Rev. 2018;32:289–99.

    Article  CAS  Google Scholar 

  29. Cortes J, Apperley JF, Lomaia E, et al. Ponatinib dose-ranging study in chronic-phase chronic myeloid leukemia: a randomized, open-label phase 2 clinical trial. Blood. 2021;138:2042–50.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the patients, their families, and their caregivers, and the study investigators and their team members at each site for participation in the PACE trial. Professional medical writing assistance was provided by Peloton Advantage, LLC, an OPEN Health company, Parsippany, NJ, USA, and funded by Millennium Pharmaceuticals, Inc., Cambridge, MA, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited.

Presented in part at Januzzi, J.L., Garasic, J., Kasner, S. et al. (2020). "An independent review of arterial occlusive events (AOEs) in the ponatinib (PON) phase II PACE trial (NCT01207440) in patients (pts) with Ph+ leukemia [abstract]." Journal of Clinical Oncology 38(15 suppl): 7550. Januzzi, J., Garasic, J., Kasner, S. et al. (2020). "Retrospective independent review of arterial occlusive events (AOES) in the phase 2 pace trial of ponatinib in Philadelphia chromosome-positive (PH+) leukemia [abstract]." HemaSphere 4(Suppl 1): 338; Januzzi, J., Garasic, J., Kasner, S. et al. (2020). "Retrospective independent review of arterial occlusive events in the phase 2 pace trial of ponatinib in Philadelphia chromosome-positive leukemia [abstract]." Presented at the 8th Annual Meeting (Virtual) of the Society of Hematologic Oncology (SOHO), September 9–12, 2020; Januzzi, J., Garasic, J., Kasner, S. et al. (2020). "Retrospective independent review of arterial occlusive events in the phase 2 pace trial (NCT01207440) of ponatinib in Philadelphia chromosome-positive leukemia [abstract]." Presented at the John Goldman E-Conference on Chronic Myeloid Leukemia: Biology and Therapy (iCMLf) October 1–4, 2020.xxx

Funding

The PACE study is sponsored by ARIAD Pharmaceuticals, Inc., Cambridge, MA, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited.

Author information

Authors and Affiliations

Authors

Contributions

KC, EB, DN, JX, SS, JLJ, TH, and JC were involved in the conception and design. JC, MM, MD, AH, JP-I, FN, D-WK, DJD, and HK contributed to the provision of study material or patients. JMG, SEK, VM, MCP, JC, MM, MD, AH, JP-I, FN, D-WK, DJD, HK, SS, TH, JX, and DN contributed to the collection and assembly of data. All authors contributed to the data analysis and interpretation. All authors wrote the manuscript. All authors were involved in the final approval of manuscript. All authors are accountable for all aspects of the work. All authors performed data analysis and interpretation, had full access and verified all the data in the study, and had final responsibility for the decision to submit for publication. All authors were involved in drafting and providing critical revision of the article. All authors read and approved the final manuscript.

Corresponding author

Correspondence to James L. Januzzi.

Ethics declarations

Ethics approval and consent to participate

PACE was approved by local ethics committees and was conducted in accordance with the Declaration of Helsinki and the International Council for Harmonisation guidelines for good clinical practice. All patients provided written informed consent.

Consent for publication

All authors have critically reviewed the manuscript and consent to publication.

Competing interests

J.L.J.: Consulting/advisory role (Takeda), research funding (Novartis); J.M.G.: Employment and stock/other ownership (family member: Vertex Pharmaceuticals), consulting/advisory role (Clinical Events Committee, ACI; AbbVie; Baim Institute; Parexel), research funding (ReCor Medical); S.E.K.: research funding (Bristol Myers Squibb, Genentech, Medtronic), consulting/advisory role (AbbVie, Abbott, AstraZeneca, BMS, Janssen, Takeda, Medtronic); V.M.: Consulting/advisory role (Takeda, Novartis, Amgen, Bayer), honoraria (Amgen, Novartis), research funding (Novartis, Rigel); M.C.P.: Endpoint adjudication committee (Takeda); J.S.: Employment, leadership role, stock/other ownership (WCG Clinical); M.M.: Consulting/advisory role, travel/accommodations/expenses, and honoraria (Novartis, BMS, Pfizer, Takeda), research funding (all to institution: Sun Pharma, Novartis, BMS); K.C.: Honoraria, consulting/advisory role, travel/accommodations/expenses (Takeda, Abbott, CSI, Philips, Abiomed, Cordis, Boston Scientific), research funding (Abbott, ARIAD, Takeda); E.B.: Honoraria, consulting/advisory role, travel/accommodations/expenses (ARIAD); MD: Consulting/advisory role (Blueprint, Fusion Pharma, Takeda, Humana, Ascentage Pharma, Adelphi, Medscape, Novartis), research funding (Takeda, Pfizer, Novartis, Incyte, SPARC, Blueprint, Leukemia & Lymphoma Society); A.H.: Research funding (Incyte, BMS, Novartis, Pfizer); J.P.I.: Consulting/advisory role (AbbVie, Janssen, AstraZeneca, Novartis, TG Therapeutics, Takeda), speakers bureau (AbbVie, Janssen, AstraZeneca, Takeda), research funding (MEI, Sunesis), patents/royalties/other intellectual property (Sellas); F.N.: Honoraria, speakers bureau, travel/accommodations/expenses (Novartis, Incyte Biosciences), consulting/advisory role (Sun Pharma), research funding (Incyte Biosciences); D.W.K.: Research funding (Novartis, BMS, Pfizer, Takeda, Sun Pharma, Il-Yang Pharm. Co., Ltd.), advisory board (Novartis, BMS); D.J.D.: Consulting/advisory role (Incyte, Pfizer, BMS, Amgen, Novartis, Celgene, Immunogen, Takeda, Blueprint Medicines), research funding (all to institution: Novartis, AbbVie, GlycoMimetics, Blueprint Medicines); H.K.: Honoraria (AbbVie, Amgen, ARIAD, BMS, Immunogen, Orsenix, Pfizer, Agios, Takeda, Actinium Pharmaceuticals), research funding (all to institution: Pfizer, Amgen, BMS, Novartis, ARIAD, Astex Pharmaceuticals, AbbVie, Agios, Cyclacel, Immunogen, Jazz Pharmaceuticals); J.X.: Employment (Takeda); T.H.: Employment (Takeda); S.S.: Employment (Takeda); D.N.: Employment (Takeda); J.C.: Consulting/advisory role (BMS, Novartis, Pfizer, Takeda), research funding (Novartis, Pfizer, Takeda, Sun Pharma).

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Januzzi, J.L., Garasic, J.M., Kasner, S.E. et al. Retrospective analysis of arterial occlusive events in the PACE trial by an independent adjudication committee. J Hematol Oncol 15, 1 (2022). https://doi.org/10.1186/s13045-021-01221-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13045-021-01221-z

Keywords