Ponatinib vs Imatinib in Frontline Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia:A Randomized Clinical Trial


Ponatinib vs Imatinib in Frontline Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia

FullText

Key Points

Question  Is frontline ponatinib superior to imatinib when combined with reduced-intensity chemotherapy in adults with newly diagnosed Philadelphia chromosome–positive (Ph+) acute lymphoblastic leukemia (ALL)?

Findings  In this randomized clinical trial, ponatinib demonstrated a significantly higher minimal residual disease–negative complete remission rate at the end of induction (34.4% vs 16.7% with imatinib) and a comparable safety profile vs imatinib when combined with reduced-intensity chemotherapy in adults with newly diagnosed Ph+ ALL.

Meaning  These efficacy and safety results support consideration of ponatinib as a frontline tyrosine kinase inhibitor in combination with chemotherapy for adults with newly diagnosed Ph+ ALL.

Abstract

Importance  In newly diagnosed Philadelphia chromosome–positive (Ph+) acute lymphoblastic leukemia (ALL), disease progression due to acquired resistance to first- or second-generation BCR::ABL1 tyrosine kinase inhibitors is common. Ponatinib inhibits BCR::ABL1 and all single-mutation variants, including T315I.

Objective  To compare frontline ponatinib vs imatinib in adults with newly diagnosed Ph+ ALL.

Design, Setting, and Participants  Global registrational, phase 3, open-label trial in adults aged 18 years or older with newly diagnosed Ph+ ALL. From January 2019 to May 2022, eligible patients at 77 sites were randomized 2:1 to ponatinib (30 mg/d) or imatinib (600 mg/d) with reduced-intensity chemotherapy, followed by single-agent ponatinib or imatinib after the cycle 20 phase of the trial. The last date of follow-up for this analysis was August 12, 2022.

Intervention  Patients received ponatinib, 30 mg/d, or imatinib, 600 mg/d, with reduced-intensity chemotherapy, followed by single-agent ponatinib or imatinib after cycle 20. The ponatinib dose was reduced to 15 mg on achievement of minimal residual disease–(MRD) negative complete remission.

Main Outcomes and Measures  The primary end point of this interim analysis was MRD-negative complete remission (≤0.01% BCR::ABL1 [MR4] centrally assessed by reverse transcriptase–quantitative polymerase chain reaction), with complete remission maintained for at least 4 weeks at the end of cycle 3. The key secondary end point was event-free survival.

Results  Of 245 patients randomized (median age, 54 years; 133 [54.3%] female), 232 (ponatinib, n = 154; imatinib, n = 78) who had p190 or p210 dominant isoforms verified by the central laboratory were analyzed for the primary end point. The MRD-negative complete remission rate (primary end point) was significantly higher with ponatinib (34.4% [53/154]) vs imatinib (16.7% [13/78]) (risk difference, 0.18 [95% CI, 0.06-0.29]; P = .002). At the data cutoff, event-free survival had not met the prespecified number of events. Median event-free survival was not reached in the ponatinib group and was 29 months in the imatinib group. The most common adverse events were similar between treatment groups. Arterial occlusive events were infrequent and comparable between groups (ponatinib, 2.5%; imatinib, 1.2%).

Conclusions and Relevance  Ponatinib demonstrated a superior rate of MRD-negative complete remission at the end of induction vs imatinib when combined with reduced-intensity chemotherapy in adults with newly diagnosed Ph+ ALL. The safety profile of ponatinib was comparable with imatinib.

Dose-Adjusted EPOCH Plus Inotuzumab Ozogamicin in Adults With Relapsed or Refractory B-Cell ALLA Phase 1 Dose-Escalation Trial


Question  Does the addition of inotuzumab ozogamicin (InO) to dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin (DA-EPOCH) result in a safe and effective treatment for adults with relapsed or refractory B-cell acute lymphoblastic leukemia or lymphoma?

Findings  In this phase 1 dose-escalation trial of 24 adult participants receiving DA-EPOCH-InO, the highest studied dose level of InO (0.6 mg/m2 on days 8 and 15) was the maximum tolerated dose. Significant severe hepatic toxic effects were rare, and there was only 1 case of sinusoidal obstructive syndrome.

Meaning  The findings suggest that DA-EPOCH-InO is a safe, well-tolerated, and clinically active chemoimmunotherapy regimen warranting further clinical investigation.

Abstract

Importance  Options for adults with relapsed or refractory B-cell acute lymphoblastic leukemia or lymphoma (B-ALL) are limited, and new approaches are needed. Inotuzumab ozogamicin (InO) has been combined with low-intensity chemotherapy, with modest improvements over historical controls, and dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin (DA-EPOCH) treatment is safe and active for newly diagnosed ALL.

Objective  To assess the safety and clinical activity of DA-EPOCH and InO in adults with relapsed or refractory B-ALL.

Design, Setting, and Participants  This single-center, single-arm, nonrandomized, phase 1 dose-escalation trial included adults with relapsed or refractory CD22+ B-ALL and was conducted between September 2019 and November 2022. At least 5% blood or marrow blasts or measurable extramedullary disease (EMD) was required for enrollment.

Interventions  DA-EPOCH was given on days 1 to 5, while InO was given on day 8 and day 15 of a 28-day cycle. Three dose levels were studied using a bayesian optimal interval design.

Main Outcomes and Measures  The primary outcome was the maximum tolerated dose of InO when combined with DA-EPOCH, defined as the highest dose level that produced a rate of dose-limiting toxicity below 33%. Secondary objectives included response rates, survival estimates, and descriptions of toxic effects.

Results  A total of 24 participants were screened and enrolled (median age, 46 [range, 28-76] years; 15 [62%] male). The median number of lines of prior therapy was 3 (range, 1-12). Three of 11 participants (27%) treated at the highest dose level (InO, 0.6 mg/m2, on day 8 and day 15) experienced dose-limiting toxicity, making this the maximum tolerated dose. No deaths occurred during the study, and only 1 patient (4%; 95% CI, 0.1%-21%) developed sinusoidal obstructive syndrome after poststudy allograft. The morphologic complete response rate was 84% (95% CI, 60%-97%), 88% (95% CI, 62%-98%) of which was measurable residual disease negative by flow cytometry. Five of 6 participants with EMD experienced treatment response. The overall response rate was 83% (95% CI, 63%-95%). Median overall survival, duration of response, and event-free survival were 17.0 (95% CI, 8.4-not reached), 15.0 (95% CI, 6.7-not reached), and 9.6 (95% CI, 4.5-not reached) months, respectively.

Conclusions  In this study, adding InO to DA-EPOCH in adults with relapsed or refractory B-ALL was feasible, with high response rates and sinusoidal obstructive syndrome occurring rarely in a heavily pretreated population. Many patients were able to proceed to poststudy consolidative allogeneic hematopoietic cell transplant and/or chimeric antigen receptor T-cell therapy. Further investigation of this combination is warranted.

CAR T Cell Therapies Last Longer, Work Better with FOXO1 Protein.


Car t cells

These days, lymphomas, leukemias, and other kinds of cancers can be effectively treated using chimeric antigen receptor T cell (CAR T) therapies. Now, new research from scientists at Children’s Hospital of Philadelphia (CHOP) and Stanford University School of Medicine helps explain why these therapies work and why they get better the longer the cells are in the body. Full details of the findings were published in Nature in a paper titled, “FOXO1 is a master regulator of memory programming in CAR T cells.”

Fewer than 50% of patients who are treated with CAR T therapies remain cured after one year. One of the reasons for this is that CAR T cells often don’t survive long enough in patients to completely eradicate their cancer. However, other studies showed that patients who were cured by CAR T therapy had cells that were more durable and able to fight the cancer longer. Results from the Nature study indicate that longer lasting cells benefit from a protein called FOXO1, which improves the survival and function of CAR T cells.

“By studying factors that drive memory in T cells, like FOXO1, we can enhance our understanding of why CAR T cells persist and work more effectively in some patients compared to others,” said Evan Weber, PhD, an assistant professor of pediatrics at the University of Pennsylvania Perelman School of Medicine and senior author on the study. 

FOXO1 is a protein that activates genes associated with T cell memory. It has previously been studied in mice but remains under-researched in human T cells or CAR T cells. To learn more about its role in human CAR T cells, the researchers used CRISPR to delete FOXO1. They found that in its absence, human CAR T cells lost their ability to form healthy memory cells or to protect against cancer in animal models.

Conversely, when researchers forced CAR T cells to overexpress FOXO1, they observed that memory genes were turned and that the T cells have an enhanced ability to persist and fight cancer. In contrast, when the researchers overexpressed a different memory-promoting factor, there was no improvement in CAR T cell activity, suggesting that FOXO1’s role is unique. The researchers also found evidence that FOXO1 activity in patient samples correlates with persistence and long-term disease control, thereby implicating FOXO1 in clinical CAR T cell responses.

“These findings may help improve the design of CAR T cell therapies and potentially benefit a wider range of patients,” Weber said. “We are now collaborating with labs at CHOP to analyze CAR T cells from patients with exceptional persistence to identify other proteins like FOXO1 that could be leveraged to improve durability and therapeutic efficacy.”

Impact of Pain on Symptom Burden in Chemotherapy-Induced Peripheral Neurotoxicity


Background

Chemotherapy-induced peripheral neurotoxicity (CIPN) is associated with treatment with neurotoxic chemotherapies, including platinum-based agents, taxanes, vinca-alkaloids, bortezomib, and thalidomide.1 Because there are no preventive or treatment measures for CIPN, symptoms may require chemotherapy dose modification, which could reduce effectiveness.2 Furthermore, CIPN symptoms affect the quality of life (QoL) of cancer survivors, often producing long-term disability, including an impact on fine motor skills, walking, and gait.3

The core symptoms associated with CIPN are sensory disturbances, including numbness and tingling.46 Neuropathic pain, often described as shooting or burning pain, is less common,7 with only 33% of patients with CIPN reporting burning pain, compared with 77% reporting severe numbness and tingling.7 This discrepancy occurs for multiple neurotoxic drugs, with patients treated with taxanes,5,8,9 bortezomib,10 and platinum-based agents2,5,9,11 all reporting more severe tingling and numbness compared with neuropathic pain.

However, our understanding of the impact of painful CIPN on patients treated with chemotherapy is inadequate. In limited previous studies, participants with painful CIPN reported worse health-related QoL than those with nonpainful CIPN.8 Furthermore, painful CIPN may be associated with comorbidities, including fatigue, anxiety, and sleep impairments.12 However, the assessment of neuropathic pain in the context of CIPN remains a challenge. There is a lack of validated diagnostic tools that address pain and its impacts separately from nonpainful CIPN symptoms. Multiple studies use the clinically graded NCI-CTCAE scale for the quantification of neuropathy severity,13 which does not include neuropathic pain. Further, few patient-reported outcome measures (PROMs) for CIPN evaluation focus on identifying pain and its impact.12 The aim of this study was to understand the differences in prevalence and symptom burden of painful versus nonpainful CIPN.

Patients and Methods

Participants

Eligible participants were cancer survivors aged ≥18 years and were 3 to 12 months post treatment with neurotoxic chemotherapy (including taxanes, platinum-based agents, bortezomib, thalidomide, and vinca alkaloids). Participants were assessed cross-sectionally on a single occasion. Relevant clinical data were retrieved from medical records, including sex and age. This study was approved by Sydney Local Health District (Royal Prince Alfred Hospital [RPAH] zone) and South Eastern Sydney Local Health District Human Research Ethics Committees, and informed consent was obtained from each participant.

CIPN and Pain Assessment: PROMs

Assessment tools are briefly described in the following sections, with further details available in Appendix 1 in the supplementary materials (available online with this article).

The validated 20-item EORTC Quality of Life Questionnaire–Chemotherapy-Induced Peripheral Neuropathy (EORTC QLQ-CIPN20) was used to assess CIPN.14 The total score as well as individual item scores assessing the impact of CIPN symptoms on patient function were investigated.

The Pain Numeric Rating Scale (PNRS) was used to assess the intensity of nerve pain experienced by participants in the 24 hours prior to testing.15 A modified Douleur Neuropathique 4 (DN4) was used to report the most common descriptors of pain in participants who had neuropathic pain, including a comparison of pain descriptors reported across different chemotherapy types.16

A semistructured qualitative interview was conducted to collect information about participant symptoms and their impact, similar to previously conducted interviews.17

Clinical Neuropathy Assessment

Trained researchers undertook a comprehensive neuropathy assessment protocol to grade CIPN severity, including clinical neuropathy grading scales and functional assessments.

The NCI-CTCAE sensory neuropathy subscale version 4.018 and the Total Neuropathy Score–clinical version (TNSc; John Hopkins University) were undertaken.19,20 Nerve conduction studies (NCS) were undertaken in the lower-limb sural and tibial nerves as per previous studies.21

Functional assessments on participants’ dominant hand assessed sensory perception via Von-Frey monofilaments22 and grating orientation task (GOT),23 as well as fine motor skills via the grooved pegboard task.24

Participant Classification

Participants were classified based on CIPN symptoms reported in the PROM (EORTC QLQ-CIPN20) as in previous studies.2 Participants who did not report any painful or nonpainful CIPN symptoms were placed in the “no CIPN” group and were excluded from further analyses. From the remaining cohort, the presence of painful CIPN was characterized using either EORTC QLQ-CIPN20 items or PNRS score (detailed in Appendix 1).

Statistical Analyses

Data analysis was undertaken using SPSS Statistics, version 27 (IBM Corp). Data were assessed for normality using the Shapiro-Wilk test, and nonnormally distributed data (P<.05) were presented as median (IQR). Mann-Whitney U tests were used to explore differences between CIPN outcome measure scores, clinical characteristics, neurophysiological measurements, and treatment factors of painful and nonpainful CIPN cohorts. Chi-square tests were used to explore group differences between participants treated with taxane and platinum-based chemotherapy in the painful CIPN cohort and to investigate behavioral changes associated with CIPN subgroups. Statistical significance was considered when P<.05.

Results

Demographics and Clinical History

A total of 579 participants with a median age of 59 years (IQR, 19 years) were assessed cross-sectionally 6 months post neurotoxic chemotherapy treatment. In total, 66% of the cohort were female (n=384). The most common cancer types were breast (32%; n=184), gastrointestinal (28%; n=162), and gynecologic (18%; n=102). The most common chemotherapy types were taxanes (57%; n=329) and platinum-based (33%; n=194).

Overall, 28% (n=159) reported no CIPN, 24% (n=140) reported painful CIPN, and 48% (n=280) reported nonpainful CIPN. Participants not reporting CIPN at the time of assessment were excluded from the analysis (n=159). Of those with CIPN (n=420), females were more likely to report painful CIPN than males (P=.02). However, there were no differences in cancer type, cancer stage, or chemotherapy type between patients with painful versus nonpainful CIPN (all P>.05) (Table 1).

Table 1.

Clinical and Demographic Characteristics of Participants With CIPN

Table 1.VIEW TABLE

There were no demographic differences between both groups in terms of age and body mass index (both P>.05). However, participants reporting painful CIPN were significantly farther from treatment completion (6 [IQR, 5] months) than participants with nonpainful CIPN (4 [IQR, 3] months) (P=.02) (Table 1).

Neuropathy Profiles and Subgroups

Participants with painful CIPN had a greater symptom burden than those with nonpainful CIPN across multiple measures, including the clinically graded scale (NCI-CTCAE; P<.001) (Table 2) and the PROM (EORTC QLQ-CIPN20; P<.001) (Figure 1A and Table 2, and Supplementary Table S1). Participants with painful CIPN also had worse neurologic examination scores (TNSc; P<.001) (Figure 1B), including higher report of sensory (P=.003) and motor (P=.001) symptoms in the extremities (Table 2). However, there were no significant differences in pinprick or vibration scores, functional assessments (all P>.05), or sural (P=.10) or tibial amplitudes (P=.30) between the groups (Table 2).

Table 2.

Comparison of Neuropathy Outcomes Between Participants With Painful and Nonpainful CIPN

Table 2.VIEW TABLE

Figure 1.
Figure 1.

Items assessing the impact of symptoms on function on the PROM (EORTC QLQ-CIPN20) were investigated. Participants with painful CIPN reported significantly more functional impairments across all of these items compared with participants with nonpainful CIPN (all P≤.003) (Figure 2).

Figure 2.
Figure 2.

The severity of neuropathic pain was reported by participants with painful CIPN on the PNRS, focused on the shorter recall period of 24 hours. The median PNRS score was 4 (IQR, 6) out of 10, with 31% (n=44) reporting no pain in the 24 hours prior to testing (score, 0/10). Overall, the level of neuropathic pain on PNRS was significantly correlated with CIPN severity across all measures (all P<.05), including the PROM (Figure 3A), the neurologic examination score (Figure 3B), and the clinically graded scale (r=0.3; P=.002). Similarly, those who reported more severe pain in the past week (EORTC QLQ-CIPN20) had worse CIPN severity across all measures compared with those reporting lower pain severity (all P<.05). Common descriptors of pain are reported in Appendix 2 in the supplementary materials.

Figure 3.
Figure 3.

Subgroups Within Moderate-to-Severe CIPN Cohort

To examine further subgroup differences, comparisons were made between participants with moderate-to-severe CIPN symptoms with pain (n=102) and without pain (n=121). Even within the moderate-to-severe CIPN cohort, those with painful CIPN symptoms had worse impairments across all CIPN severity measures, including the PROM, the clinically graded scale, and the neurologic examination score (all P<.05) (Table 3). However, there were no demographic, neurophysiological, or functional differences between the groups (P>.05) (Table 3).

Table 3.

Comparison Between Participants With Worse CIPN (NCI-CTCAE ≥2) and No Pain Versus With Pain

Table 3.VIEW TABLE

Comparison of CIPN Subgroups Among Different Chemotherapy Types

The 2 largest chemotherapy-type cohorts (paclitaxel and oxaliplatin) were selected for group comparisons. There were significant differences in the prevalence of pain between the paclitaxel and oxaliplatin chemotherapy cohorts (28% vs 39%, respectively; P=.03) (Supplementary Figure S1). Group comparisons between paclitaxel-treated and oxaliplatin-treated participants can be found in Appendix 2 and Supplementary Tables S2 and S3.

Impact of Painful CIPN on Sleep, Exercise, and Treatment-Seeking Behavior

To characterize the impact of painful CIPN on behavior and patient function, we compared the painful (n=87) and nonpainful CIPN (n=193) cohorts who completed the structured interview in terms of self-reported sleep dysfunction, exercise impairment, and treatment-seeking behavior.

Participants with painful CIPN were more likely to report that their symptoms affected their ability to exercise (odds ratio [OR], 2.1; P=.007) than those without pain, with 43% (n=37) in the painful CIPN cohort reporting their exercise ability being affected by CIPN compared with 26% (n=51) of those without pain. Similarly, participants with painful CIPN were more likely to report that they had trouble sleeping (OR, 2.8; P<.001), with 47% (n=41) in the painful CIPN cohort reporting sleep dysfunction due to CIPN compared with 24% (n=46) of those without pain (Supplementary Table S4).

In addition, participants with painful CIPN were more likely to report seeking treatment of their symptoms than those without pain (OR, 3.2; P<.001) (Supplementary Table S4), with 69% (n=60) of the painful CIPN cohort reporting trying to find treatment options compared with 41% (n=79) of those with nonpainful CIPN. Furthermore, participants with painful CIPN were 4 times as likely to report the use of medication to ameliorate neuropathy than those with nonpainful CIPN (P<.001; Supplementary Table S5). These medications included anticonvulsants (pregabalin and gabapentin) and antidepressants (duloxetine and amitriptyline). In total, 12% (n=17) of the painful CIPN cohort were receiving medication for CIPN at the time of assessment, compared with 3% (n=9) of the nonpainful CIPN cohort.

Discussion

This study investigated neuropathic pain and its impact on symptom severity, sensory function, and behavior in participants with CIPN. Overall, 33% of participants with CIPN reported painful CIPN, which was associated with higher symptom severity across all CIPN outcome measures. The participants with painful CIPN reported more functional consequences than those without pain and were more likely to take neuropathy medications and report sleep dysfunction and exercise intolerance. Pain descriptors were similar between paclitaxel-treated and oxaliplatin-treated cohorts; however, pain was more prevalent in the oxaliplatin-treated cohort.

Other cohort studies have reported a similar prevalence of neuropathic pain, ranging between 20% and 33% of patients,2527 in line with our results. In this study, participants with painful CIPN had worse global CIPN severity across all measures compared with participants with nonpainful CIPN. The presence of painful CIPN was associated with worse impairment of activities of daily living, a particularly reduced ability to distinguish temperature, more instability when standing or walking, and difficulty writing and manipulating small objects with fingers.

However, there were no group differences in performance on functional assessments and NCS. This suggests a potential separation between the perception of overall symptom burden and objective measures of neuropathy severity. Discrepancies between patient-reported symptoms of CIPN and neurologic examination have been previously identified,28 suggesting that these assessment tools address different aspects of CIPN.29 Patient reports of CIPN symptom severity and impact often provide a broader perspective compared with focal quantification of neurologic status. In addition, most neurophysiological measures of CIPN, including NCS, measure large nerve fiber function, whereas small nerve fiber dysfunction is less accessible to measure, presenting a potential limitation in fully capturing objective deficits.30 Importantly, patient report remains a key metric of CIPN severity, particularly given the lack of efficacy of measures such as NCS to identify differences between CIPN cohorts.

Participants with painful CIPN may be higher symptom reporters due to their increased symptom severity. A previous study found that participants who reported painful CIPN also reported higher anxiety and depression,31 suggesting that there may be a modulating effect of psychological factors on pain perception in patients with CIPN. However, the direction of this association remains uncertain, given that patients with painful CIPN were more likely to have persisting anxiety and depression following treatment, in contrast to patients with nonpainful CIPN, who demonstrated greater improvements in anxiety and depression following treatment cessation.8

The presence of neuropathic symptoms, including pain, negatively impacts the QoL of cancer survivors. In this study, the presence of painful CIPN affected patient-reported sleep, exercise, treatment-seeking behavior, and functional capacity. Although one previous study also highlighted the association between painful CIPN and comorbidities, including increased sleep dysfunction, fatigue, anxiety, and depression,32 research on the comorbidities associated with painful CIPN remains limited12 and represents a gap in enabling personalized management strategies for people with CIPN.

This study also found that oxaliplatin-treated patients had overall worse CIPN symptoms, significantly lower sural amplitudes, and greater functional changes in sensory perception and fine motor skills than those treated with paclitaxel. The different CIPN profiles of taxane and platinum-based chemotherapies have been previously reported,3335 with reports at 1-year follow-up being similar to the current study.33 On the contrary, there were no differences in subjective or objective measures of CIPN between taxane-treated or platinum-treated patients at 5-year follow-up.35 With regard to pain, in this current study it was significantly more prevalent in oxaliplatin-treated patients than paclitaxel-treated patients. Interestingly, oxaliplatin-treated patients with painful CIPN also benefitted the most from duloxetine treatment in clinical trials, suggesting that different pain phenotypes may guide treatment responsiveness between chemotherapy types.36,37 Understanding differences in the chemotherapy-specific profiles of CIPN is important to guide patients and clinicians in understanding the likelihood of symptom recovery and adaptation over time.29

To date, treatment options recommended for the management of painful CIPN remain limited.36 In this study, only 12% of participants with painful CIPN reported taking anticonvulsants or antidepressants for neuropathy treatment. This reflects similar experiences with low medication uptake in Australian3 and international settings.38,39 Although duloxetine is recommended for the treatment of painful CIPN by international guidelines,40 in real-world practice, duloxetine treatment of painful CIPN is limited, with high rates of nonresponse and side effects leading to lack of tolerability.38 Better phenotyping of patients to determine who is most responsive to duloxetine and other therapies will likely improve real-world outcomes.36,37

Although there are emerging strategies for the management of neuropathic pain, including both pharmacologic and nonpharmacologic approaches,41,42 both will require the identification of patient subgroups likely to benefit most. Novel therapies, such as targeted drug delivery and neurostimulation methods, hold promise for reducing neuropathic pain but require additional testing and validation.41 Preliminary findings suggest that nonpharmacologic approaches, such as cognitive behavioral therapy, might improve QoL in patients with neuropathic pain42 and be more acceptable to patients.

Overall, this study provides a clearer understanding of differing symptom patterns in a large cohort. We used a combination of subjective and objective measures of CIPN as well as the combination of patient-reported and clinically graded outcome measures of CIPN. An issue limiting the previous understanding of painful CIPN is the use of combined descriptors of CIPN, collapsing sensory and painful symptoms of CIPN into a singular measure.43 For this reason, we used specific PROM items that assessed numbness, tingling, and shooting or burning pain in the last 7 days to capture a broader recall period and separate patients into groups according to neuropathic pain.2,7,33 However, these measures are not specifically validated to identify neuropathic pain. Although we did use validated neuropathic pain tools to assess pain intensity and descriptors, we did not use them for the purpose of participant classification due to the recall period only pertaining to the last 24 hours prior to participant testing.

Given that prior studies have demonstrated a “coasting” effect for up to 3 months post treatment completion,44 we chose to examine a cross-sectional cohort between 3 and 12 months post treatment completion. However, the cross-sectional nature of this study may be a limitation, and future prospective analyses will provide insights into the development of painful CIPN over time. Furthermore, we included multiple cancer and chemotherapy types in the analysis and did not control for all preexisting conditions that cause peripheral neuropathy or pain. However, >70% of this cohort developed neuropathy during treatment, suggesting that the CIPN symptoms identified were related to treatment-emergent toxicity rather than other factors. Furthermore, the inclusion criteria were deliberately broad to capture a naturalistic cohort reflecting patients receiving chemotherapy in a clinical setting. Although participants were asked about functional limitations, this study did not quantify the number of falls or participant balance performance. In addition, participants did not report whether they had tried medications to treat neuropathic symptoms previously and why these were discontinued.

Overall, given the outcomes of this study, we recommend that neuropathic pain be assessed in research and clinical settings as part of a comprehensive CIPN assessment. The tools used for this purpose should use a recall period longer than 24 hours, such as the EORTC QLQ-CIPN20. Other CIPN outcome measures, including the clinically graded scale (NCI-CTCAE) and the neurologic examination score (TNSc), which are used to assess CIPN severity, do not include questions to address neuropathic pain severity, and additional tools are required to address this. Finally, we recommend assessment of the impact of neuropathic pain on patient function and behavior, because our study has highlighted the long-term and deleterious consequences of pain on cancer survivors with CIPN.

Current guidelines for CIPN discuss the assessment of CIPN and include potential treatment options.40,45 However, these guidelines lack information relevant to patient subgrouping and phenotyping, particularly patients with neuropathic pain. The use of PROMs remains important in identifying clinically relevant symptom patterns, whereas NCS may not provide useful information in the classification of CIPN subgroups. Critically, the lack of accurate assessment of painful and nonpainful CIPN symptoms in clinical trials may lead to inaccurate results regarding intervention efficacy. Accordingly, it is essential that appropriate outcome measures be used to enable differentiation of painful and nonpainful symptoms.

Conclusions

Although the pathophysiological mechanisms underlying the differences in symptom expression within CIPN remains unclear, improved screening for pain and associated functional changes will allow a better appreciation of symptom burden and encourage more tailored intervention strategies to improve the QoL of cancer survivors.

Navigating the Management of Chronic Phase CML in the Era of Generic BCR::ABL1 Tyrosine Kinase Inhibitors


The introduction of the BCR::ABL1 tyrosine kinase inhibitors (TKIs) in 2000 changed the treatment landscape and outcome of patients with chronic myeloid leukemia (CML).1 While undergoing TKI therapy, patients are now anticipated to have a near-normal life expectancy, if they can afford the treatment, are optimally managed, are compliant, and are salvaged with later-line TKI therapy if they manifest TKI resistance (defined as BCR::ABL1 transcripts on the International Scale [IS] >1% after >12 months of TKI therapy).24 The 4 FDA-approved TKIs for frontline chronic phase CML (CML-CP) therapy are imatinib (first-generation TKI), dasatinib, nilotinib, and bosutinib (second-generation TKIs).58 All can be used as later-line therapies depending on the reasons for changing (eg, resistance or intolerance, prior TKIs used, presence of ABL1 kinase domain mutations, patient comorbidities, cost). Ponatinib and asciminib are third-generation TKIs approved for patients who do not experience a response to prior second-line TKIs or those with T315I mutation.9,10 This review discusses the approach to treatment of CML-CP in the era of the generic TKIs to quantify the treatment value, or cost/benefit ratio, of such strategies.

A Primer on the Price of the BCR::ABL1 TKIs

The first BCR::ABL1 TKI, imatinib, demonstrated remarkable efficacy and a favorable safety profile compared with the standards of care—interferon alpha and allogeneic stem cell transplantation (SCT)—justifying its widespread use and initial price of $26,400/year when it was introduced in 2000 (inflation-adjusted estimate $48,000/year in 2023). The initial proposed price was comparable to that of interferon and was estimated by the company to cover the costs of its research and development (originally funded by investigator-awarded grants from the National Institutes of Health) and generate generous profits.11 However, by 2012, the price had increased to $120,000/year, even though the number of patients being treated was increasing (from 5,000 to 9,500 new cases per year between 2000 and 2020; mortality only 1% annually), with the CML prevalence in the United States increasing from 30,000 cases in 2000 to an estimated 120,000 to 150,000 cases between 2020 and 2030.12 The price of brand imatinib increased further, to approximately $160,000/year by 2020, and newer-generation TKIs were all priced at a range of $200,000 to $300,000/year.1323

Imatinib lost its patent in July 2015, and generics became available outside the United States at a price range of $500 to $5,000/year.17,24 In the United States, because of distorted market forces and even with the availability of 4 to 5 generics by 2020 and ≥15 generics today, the average annual price of imatinib decreased to only $5,000/year when leaving the manufacturer (wholesale acquisition cost [WAC]), but remained at approximately $132,000/year by the time it reached the patient (average wholesale price [AWP]) because of the profits added by the various intermediaries (eg, wholesalers, pharmacies, hospitals, group purchasing organizations, pharmacy benefit managers).14,17 This is until the Mark Cuban Cost Plus Drug Company and similar organizations started offering generic imatinib directly to patients (with a prescription) (Table 1).17

Table 1.

Average Wholesale Price of BCR::ABL1 Tyrosine Kinase Inhibitors in 2023

Table 1.VIEW TABLE

Generic dasatinib is available outside the United States and is expected to be available in the United States by 2024. Generic formulations of bosutinib and nilotinib are expected to become available in the United States by 2027. This would then fulfill the 3 key aims of TKI therapy in CML: (1) survival normalization; (2) achievement of a durable deep molecular response (DMR; BCR::ABL1 transcripts [IS] ≤0.01%; MR4+), which translates into a treatment-free remission (TFR) status in 40% to 80% of patients, depending on whether the DMR duration before TKI discontinuation is 1 year, 2 years, or ≥5 years25,26; and (3) making the treatment available and affordable to 100% of patients with CML, hence the discussion of the treatment value of different TKIs.

Treatment of CML-CP in the Frontline Setting

The choice of frontline TKI depends on several factors: (1) the goal of CML therapy (survival vs treatment discontinuation; potentially linked to the patient’s age); (2) the cost and affordability of TKIs; and (3) the patient’s age and comorbidities. When overall survival (OS) is the endpoint of therapy, imatinib provides a good treatment value.20,27 Cost Plus Drugs offers generic drugs at cost plus 15%; for instance, 1 month of imatinib at 400 mg daily costs $34.70 per month, or $416.40/year.17 For a patient aged ≥60 years and considering a life expectancy of approximately 80 years in the United States, an additional 20 to 30 years of therapy at today’s price would cost approximately $8,300 to $12,500. The survival benefit with imatinib is similar to that obtained with second-generation TKIs priced at >$200,000/year (Table 1).17 Multiple other generic companies (eg, Civica Rx, ScriptCo Pharmacy) are working on providing more affordable generics to the patients, which may further reduce costs.17 Today’s issues with such generic companies are that (1) they bill the patients directly, although the cost may still be lower than the out-of-pocket expenses for drugs covered by insurers; 2) the range of medications offered is limited; and (3) all drugs that are offered are generics. However, encouraging and publicizing these new sources of affordable drugs could result in such companies broadening the range of drugs offered, bypassing intermediaries to expand to brand drugs, and billing insurers. This would be a major disruption of the distorted market forces in the United States, normalizing them to the original free market/capitalistic intent, and lowering significantly the drug prices and cost of care in the United States, similar to the situation existing today in Europe, Canada, Australia, Japan, and the rest of the world.

A recent analysis of patients with CML in the SEER database reported a 5-year OS rate of 73%, approximately 15% to 20% lower than reported from national studies in countries with universal health care systems (Sweden, Germany, France) and from company-sponsored trials in which 100% of patients have continuous access to the TKIs. Survival was also worse among patients with lower income, suggesting reduced access to TKI therapy among those with poorer financial status.28

With the availability of more and probably cheaper generics, all patients with newly diagnosed CML-CP should have access to TKI therapy. Hematologists and oncologists should take into consideration their patients’ financial means when selecting a TKI.

If TFR is the goal of therapy, and among patients presenting with high disease burden (high Sokal risk), second-generation TKIs may be the best frontline strategy.20,21,27 Outside the United States, generic dasatinib 50 mg daily provides the best treatment value (discussed later). However, in the United States, the benefit of second-generation TKIs may be offset by their higher total cost. A good treatment value, based on the Institute for Clinical and Economic Review (ICER), should not exceed $50,000/year, and should in time not exceed $5,000/year as generic formulations are available.21 Generic versions of dasatinib, bosutinib, and nilotinib will make them a good treatment value as frontline therapy to nearly all patients with CML, whether the aim of therapy is OS or TFR. This will also create more incentive for providers and patients to favor second-generation TKIs in the frontline setting. Faster and earlier achievement of deeper molecular responses will prevent the rare occurrence of early lymphoid blastic transformation with imatinib (approximately 2%–5% in the first 2 years) and will allow patients to reach the goal of durable DMR 1 to 2 years earlier than with imatinib therapy. This will improve overall outcome, particularly in patients with high Sokal risk; reduce the cost of care; and reduce the potential occurrence of long-term toxicities.

However, as with the experience with imatinib generics in the United States, one cannot assume that the AWP of generic second-generation TKIs will fall to reasonable levels when 4 to 5 generics of each become available. Brand drug companies often maneuver to extend patent durations, which delays the availability of generics, and then when they are available, the intermediaries often intervene to keep the AWP inflated.14

Lowering TKI Doses to Reduce Cost and Improve Affordability

To improve the affordability and lower the cost and toxicities of TKIs, their dose schedules can be safely reduced without compromising efficacy. Studies have shown that, in newly diagnosed CML-CP (mostly not high-risk Sokal), dasatinib 50 mg daily was associated with high rates of major molecular response (MMR; BCR::ABL1 transcripts [IS] ≤0.1%) and DMR with a low incidence of adverse events.29,30 In a propensity score matching analysis, dasatinib 50 mg daily was at least as effective as 100 mg daily with a better safety profile, probably due to fewer treatment interruptions.31 In a Japanese trial of older patients with newly diagnosed CML-CP, dasatinib 20 mg daily showed encouraging efficacy and safety profiles, although the risk of early failure was higher than expected with the higher doses of dasatinib. Dasatinib 20 mg daily may be worth considering in patients aged ≥70 years in whom a higher incidence of pulmonary toxicities is anticipated.32

Other studies have reported similar observations with bosutinib, nilotinib, and ponatinib, where lower doses were associated with good efficacy, fewer toxicities, and lower cost.3339 In a retrospective analysis of the BFORE randomized trial of bosutinib versus imatinib in newly diagnosed CML-CP, 103 of the 268 patients treated with bosutinib had dose reductions from 400 mg to 300 mg (n=80) and 200 mg (n=23). Of them, 41 (42%) achieved their first complete cytogenetic response following dose reduction, and 40 (39%) achieved their first MMR following dose reduction. This strategy reduced the incidence of most adverse events.33 Another study of bosutinib dose optimization in the second-line setting with elderly patients showed high response rates and a favorable safety profile when bosutinib was given in a stepwise approach (starting at 200 mg daily and increasing by 100 mg increments as tolerated).34 In the OPTIC trial, a response-based dose-reduction strategy of ponatinib was evaluated in 283 patients with CML-CP resistant to ≥2 TKIs or with T315I mutation. Patients received ponatinib 45, 30, or 15 mg daily, with dose reduction to 15 mg daily once the BCR::ABL1 transcripts (IS) were ≤1%. This approach resulted in higher response rates with ponatinib 45 mg daily in patients with T315I-mutated CML, but similar response rates among the 3 dose levels in patients without the mutation. The 4-year OS rates of 86% to 88% were similar with the 3 dose levels across all CML subsets.38 A pooled analysis from the PACE and OPTIC trials showed that adopting a dose-optimization strategy with ponatinib resulted in fewer adverse events and similar or better outcomes.39

Dose-reduction strategies were also evaluated among patients in MMR. In one retrospective analysis, 298 cases (in 246 patients) of CML-CP with TKI dose de-escalation due to adverse events were analyzed: imatinib (n=90), dasatinib (n=88), nilotinib (n=81), and bosutinib (n=39). The dose of imatinib was reduced to 200 to 300 mg daily, dasatinib to ≤20 to 70 mg daily, nilotinib to ≤200 to 400 mg daily, and bosutinib to <200 to 300 mg daily.36 After dose reduction, MMR was maintained in 274 (92%) cases. Of 204 patients in MR4 at the time of dose reduction, 171 (84%) maintained or deepened their molecular responses. Of 94 cases in MMR at the time of dose reduction, 51 (54%) had improved molecular response. Of 13 cases who lost MMR, 11 regained response at a median of 3 months from restarting either the same TKI (at a higher dose) or a different TKI. Seventy-six patients eventually discontinued the low-dose TKI; their 2-year TFR rate was 74%.36 Another phase II study examined the role of TKI dose de-escalation before treatment discontinuation among 174 patients treated for ≥3 years, and with stable MMR or MR4 for ≥12 months. They received their initial TKI with 50% dose reduction for 12 months: imatinib 200 mg daily (n=148), dasatinib 50 mg daily (n=10), or nilotinib 200 mg twice daily (n=16). During the 12 months of dose de-escalation, 12 (7%) patients lost MMR but regained it within 4 months of resuming full-dose TKI. This strategy was associated with fewer adverse events (lethargy, diarrhea, rash, and nausea) after dose reduction.35

When considering dose reductions, we should note that lower doses of some TKIs (imatinib, dasatinib, nilotinib, bosutinib) translate into lower costs (Table 1). The unfortunate exception is ponatinib, where the manufacturer decided to price 45-, 30-, and 15-mg tablets at the same annual price of $271,000. This exception should be remedied.

Whether using generic or brand formulations, physicians should consider tailoring the choice of TKI to the patients’ comorbidities.27 Common adverse events observed with imatinib include fluid retention, periorbital edema, and bone and muscle aches. Dasatinib therapy is mostly associated with myelosuppression, pleural effusions, and rarely, pulmonary hypertension (1%–2%).40,41 Nilotinib is associated with an increased risk of diabetes and dyslipidemia, and a higher incidence of arterial and venous occlusive events as reported after a follow-up of 10 years.42,43 Bosutinib can result in gastrointestinal toxicities, mostly diarrhea, as well as hepatic and renal dysfunction.44,45

The Two Causes of Treatment Failure in CML

Treatment failure can be due either to CML resistance or TKI intolerance. The rate of true CML resistance, defined as BCR::ABL1 transcripts (IS) >1% (loss of cytogenetic or hematologic response) or CML transformation, is approximately 10% at 10 years of treatment. TKI intolerance (toxicities) occurs in 15% to 25%.20,27,4648 The most common reason for changing TKI therapy is toxicity, which occurs in 15% to 25% of patients at 10 years, depending on the physician and patient thresholds. The choice of second-line therapy and beyond depends on the previous TKIs used, the cause(s) of failure (resistance or intolerance), the presence of ABL1 kinase domain mutations, the patient’s comorbidities, and the cost of therapy.17

Management of TKI Intolerance/Toxicities

For this discussion, TKI cross-intolerance is defined as the occurrence of similar toxicities with different TKIs, or the occurrence of different toxicities more often in a patient with a TKI toxicity. In the past, TKI cross-intolerance was considered uncommon when changing TKIs because the agents had very different chemical structures, but recently the phenomenon has been clearly recognized. For example, a patient who develops pleural effusions on dasatinib may be at a higher risk of developing pleural effusions on bosutinib, but less so on imatinib or nilotinib. A patient who develops arterial occlusive events (AOEs) on nilotinib or ponatinib would be more likely to develop AOEs on dasatinib (100 mg daily), but less so on imatinib or bosutinib. In a recent analysis, Busque et al49 showed that the primary driver for switching TKIs was intolerance in all lines of treatment. Overall, serial intolerance was 6.6 times more frequent than serial resistance, suggesting a class effect for intolerance in some patients (20 patients switched serially across all lines due to intolerance).

Physicians and patients favored changing TKIs when intolerance occurred, because they assumed that a lower dose would have a lower efficacy, hence the term “failure.” This notion was also favored in the original studies (the only TKI available was imatinib, thus the need to include patients with imatinib toxicities) and current studies of second- and newer-generation TKIs that lumped together the 2 conditions (resistance and intolerance). This concept that a lower dose would have a lower efficacy was also publicized by the pharmaceutical companies, eager to promote the use of newer-generation TKIs over imatinib. Today, we advocate separating the 2 “failure” conditions, failure-resistance and failure-toxicity, and restricting the term failure to true resistance. This is how it is defined in the European LeukemiaNet (ELN) recommendations50 and NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for CML.51

In patients with TKI intolerance, the dose should always be reduced before a change of therapy is considered, in the absence of a prohibitive toxicity. A proposed strategy is summarized in Table 2. The prohibitive toxicities include (1) recurrent (>1 episode despite dose reduction) pleural effusions (occurring primarily with dasatinib, less commonly with bosutinib); (2) pulmonary hypertension (usually secondary to dasatinib; reversible in some cases after treatment with sildenafil and steroids); (3) arterio-occlusive or vaso-occlusive events (more common with nilotinib and ponatinib); (4) pancreatitis (possible with all TKIs, but mostly seen with nilotinib and ponatinib); (5) severe hypertension with ponatinib not responsive to antihypertensive therapies; (6) enterocolitis with bosutinib; (7) neurotoxicity (including dementia-like condition, parkinsonism, and intracranial hypertension; rarely seen with imatinib and dasatinib; can be reversible after several weeks or months from treatment discontinuation); and (8) immune-mediated adverse events, such as pneumonitis, hepatitis, myocarditis, pericarditis, or nephritis.27,52 Notably, the cost of treating adverse events resulting from the long-term use of newer-generation TKIs adds to the original price of the drug. This results in an incremental cost of care and a reduction in the treatment value of second- and third-generation TKIs.21

Table 2.

Management of TKI Intolerance or Toxicities in Patients With No Resistance

Table 2.VIEW TABLE

In the era of generic TKIs, we recommend using second-generation TKIs in patients responding to treatment who have intolerance to frontline imatinib: generic dasatinib 20 to 50 mg outside the United States, and lower dose schedules of any second-generation TKI in the United States (generic dasatinib is expected to be available in the United States in 2024, and generic nilotinib and bosutinib in 2027). Patients with intolerance to frontline second-generation TKIs can be switched to an alternative second-generation TKI that has a better safety profile tailored to the patient’s comorbidities. Generic imatinib could also be considered as a second-line therapy in selected patients who discontinue frontline second-generation TKIs due to toxicity (but not resistance). In responding patients with intolerance to imatinib and all 3 second-generation TKIs (very uncommon), lower dose schedules of ponatinib (15 mg daily) or asciminib (lower dose schedules not studied yet) may be used. For treatment intolerance, and assuming the patient does not have resistant disease (BCR::ABL1 transcripts [IS] ≤1%), we favor starting the new TKI at a lower dose schedule.

Management of TKI Resistance

Management of TKI resistance is guided by the type of frontline TKI used and the presence or absence of ABL1 kinase domain mutations (Table 3). In patients who develop resistance to frontline imatinib, and in the absence of resistance ABL1 mutations, choosing a second-generation TKI (dasatinib, bosutinib, or nilotinib) depends on the patient’s comorbidities, as discussed earlier. In patients who develop resistance to frontline second-generation TKIs, treatment is tailored according to the ABL1 mutational profile. In the presence of a resistance mutation other than the T315I, it is feasible to switch to an alternative second-generation TKI to which the detected mutation is sensitive. In patients without a guiding mutation and who have CML resistance to a second-generation TKI, switching to a third-generation TKI is better than rotating second-generation TKIs due to the low response rates observed with this strategy.53 In patients with no detectable ABL1 mutation, or in those with the T315I gatekeeper mutation, we recommend changing treatment to ponatinib. Ponatinib is approved by the FDA at a starting dose of 45 mg daily, and upon achieving BCR::ABL1 transcripts (IS) ≤1%, a dose reduction to 15 mg is recommended. However, based on data available from the OPTIC trial, we recommend a starting dose of 45 mg daily for T315I-mutated disease and 30 mg daily for non–T315I-mutated disease, with a subsequent dose reduction to 15 mg daily once BCR::ABL1 transcripts (IS) are ≤1%.54 Patients with ponatinib resistance should be considered for allogeneic SCT.

Table 3.

Management of TKI Resistance

Table 3.VIEW TABLE

Of note, the long-term follow-up results indicate that failure to achieve the ELN milestones (resistance) with imatinib do not appear to be associated with such a dire prognosis (as thought previously), and that the term “failure” may perhaps be replaced with “caution” and may not necessarily indicate a need to change TKI therapy.47,48 Also, the prognosis of T315I-mutated CML is not that adverse when it occurs in chronic phase (5-year OS of 70% in CML-CP, 37% in accelerated phase, and 10% in blastic phase). In such instances, allogeneic SCT, a one-time procedure, may be as effective (and curative) and less costly than using third-generation TKIs.55

Ponatinib and Asciminib: Third-Generation TKIs

Ponatinib is a third-generation TKI approved by the FDA for patients in whom ≥2 prior TKIs failed or those harboring the T315I mutation based on the results from the PACE trial showing improvement in the rates of major cytogenetic response.56 However, ponatinib 45 mg daily was associated with a high rate of adverse events, in particular AOEs (15%–20%). Findings from the OPTIC dose-optimization trial showed that using ponatinib 30 mg daily and decreasing the dose to 15 mg daily once BCR::ABL1 transcripts (IS) ≤1% is as effective and significantly less toxic than ponatinib 45 mg daily; the higher dose is most useful in patients with T315I mutation.54 Asciminib, a newer third-generation TKI was also approved by the FDA as third-line therapy and for T315I-mutated disease based on the results of ASCEMBL trial showing higher rates of MMR at 6 months (26% vs 13%) compared with bosutinib.10

Findings from the PACE and OPTIC trials showed 2-year OS rates of 85% and 91%, respectively, with ponatinib given to patients with CML-CP resistant to prior second-generation TKIs. With longer-term follow-up, the 5-year OS rate was 73%.9,54,56 This highlights the improved survival with ponatinib compared with second-generation TKIs in patients with resistant CML-CP.52,5759 In contrast, with longer follow-up of the ASCEMBL trial in patients with CML-CP and ≥2 prior TKIs, no survival difference was noted between asciminib and bosutinib, with 2-year OS rates of 97% and 99%, respectively.60

Treating patients with ponatinib or asciminib is expensive. The cost of ponatinib therapy is approximately $271,000/year regardless of the dose. The cost of asciminib therapy is approximately $290,000/year for patients without T315I mutation (40 mg twice a day) and $1,448,000/year for those with T315I mutation (200 mg twice a day). Therefore, the cost of their treatments is excessive, particularly for patients with T315I mutation. Compared with allogeneic SCT, neither ponatinib nor asciminib is a good treatment value. In a retrospective analysis of patients with T315I mutation, treatment with ponatinib and/or asciminib or allogeneic SCT was independently associated with improved OS.55 Therefore, in patients with T315I mutation, ponatinib can be used for a short period as a bridge to allogeneic SCT followed by (if indicated based on BCR::ABL1 transcripts) maintenance therapy with either ponatinib or an alternative second-generation TKI. Compared with a cost of $270,000 to >$1,400,000/year for third-generation TKIs, a one-time allogeneic SCT procedure costs approximately $20,000 to $40,000 in some countries such as India, Egypt, and Mexico.61,62 In the United States, a one-time allogeneic SCT costs $300,000 to $500,000.

Conclusions

With the current availability of highly affordable imatinib generics, and hopefully second-generation TKI generics soon, all patients with CML-CP should have access to affordable, highly effective, and safe treatments. Using generic second-generation TKIs at reduced doses (dasatinib 50 mg daily) may be as effective, less toxic, and less costly than at standard doses, possibly making them preferable to imatinib as frontline therapy. Physicians should carefully select the most affordable TKI based on the goals of treatment and the patient’s comorbidities. In later lines of therapies and following resistance to second-generation TKIs, we favor dose-adjusted ponatinib due to longer follow-up and lower cost compared with asciminib. Allogeneic SCT should be considered in patients in whom treatment with second-generation TKIs fails, those with no access to newer TKIs, those with T315I-mutated CML, and selected patients with poor compliance.