CC BY-NC-ND 4.0 · South Asian J Cancer 2024; 13(02): 132-141
DOI: 10.1055/s-0042-1757911
Original Article
Hematological Malignancie

A Retrospective Analysis of BCR-ABL1 Kinase Domain Mutations in the Frontline Drug Intolerant or Resistant Chronic Myeloid Leukemia Patients: An Indian Experience from a High-End Referral Laboratory

Atreye Majumdar*
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Rahul Katara*
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Avshesh Mishra
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Aastha Gupta
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Deepak K. Sharma
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Aman K. Srivastava
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Shivani Sharma
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Ankita Jaiswal
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Mallika Dixit
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Vipin Kumar
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Sachin Kumar
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Varun Kumar
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
Rahul Sharma
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
,
1   Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, Haryana, India
2   Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, Odisha, India
› Author Affiliations
Funding This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
 

Abstract

Zoom Image
Atreye Majumdar
Zoom Image
Sambit K. Mohanty

Objective This article identifies and evaluates the frequency of mutations in the BCR-ABL1 kinase domain (KD) of chronic myeloid leukemia (CML) patients who showed suboptimal response to their current tyrosine kinase inhibitor (TKI) regime and assesses their clinical value in further treatment decisions.

Materials and Methods Peripheral and/or bone marrow were collected from 791 CML patients. Ribonucleic acid was extracted, reverse transcribed, and Sanger sequencing method was utilized to detect single-nucleotide variants (SNVs) in BCR-ABL1 KD.

Results Thirty-eight different SNVs were identified in 29.8% (n = 236/791) patients. T315I, E255K, and M244V were among the most frequent mutations detected. In addition, one patient harbored a novel L298P mutation. A subset of patients from the abovementioned harbored compound mutations (13.3%, n = 33/236). Follow-up data was available in 28 patients that demonstrated the efficacy of TKIs in correlation with mutation analysis and BCR-ABL1 quantitation. Molecular response was attained in 50% patients following an appropriate TKI shift. A dismal survival rate of 40% was observed in T315I-harboring patients on follow-up.

Conclusion This study shows the incidence and pattern of mutations in one of the largest sets of Indian CML patients. In addition, our findings strengthen the prognostic value of KD mutation analysis among strategies to overcome TKI resistance.


#

Introduction

Chronic myeloid leukemia (CML) accounts for 15 to 20% of all leukemia in adults and is characterized by the Philadelphia chromosome (Ph), formed by the translocation of genetic material between the long arms of chromosomes 9 and 22 [t(9; 22)(q34;q11)].[1] At a molecular level this leads to the fusion of two genes, breakpoint cluster region (BCR) and Abelson murine leukemia gene (ABL), a proto-oncogene that encodes a tyrosine kinase involved in cell division, differentiation, apoptosis, and deoxyribonucleic acid (DNA) damage repair. The BCR-ABL1 gene fusion leads to a constitutively active ABL1 kinase that impairs terminal differentiation in the myeloid series cells leading to accumulation of cells of granulocytic lineage at variable stages of maturation ranging from blasts to mature neutrophils depending on the clinical stage of CML.[2]

CML is considered as a “poster child” for precision medicine since 2001 when the first tyrosine kinase inhibitor (TKI) imatinib was introduced as the frontline therapy to target Ph+ leukemic cells with minimal damage to the normal human cells.[3] Its mechanism of action is through a competitive binding at adenosine triphosphate-binding sites of the tyrosine kinase,[4] and was the first Food and Drug Administration-approved drug that has emerged as a therapeutic “gold standard.” Further, second-generation TKIs (2GTKIs) such as nilotinib, dasatinib, and bosutinib were developed to alleviate imatinib resistant or intolerant CML and were eventually approved for first-line therapy. Given the relative predictability of CML management with TKIs, a precise treatment guideline rendered by the National Comprehensive Cancer Network (NCCN) and the European LeukemiaNet (ELN) for the CML patients has been widely accepted.[5]

Despite the promising efficacy of imatinib, 33 to 40% of the patients acquire resistance or intolerance to the drug and are unable to reach the desired milestones.[6] [7] Most importantly, up to 50% patients even acquire resistance to the second or third line drug. Based on the criteria established by the NCCN guidelines and ELN, resistance is defined as: less than a complete hematologic response (CHR) and/or no cytogenetic response (CyR: Ph+ bone marrow metaphases > 95%) at 3 months, BCR-ABL1 transcript levels > 10%, and/or less than a partial CyR (≤ 35% Ph+ metaphases) at 6 months, BCR-ABL1 transcript levels above 1% and/or less than a complete CyR (CCyR; no Ph+ metaphases) at 12 months, and BCR-ABL1 transcript levels > 0.1%, or loss of a CHR or CCyR or confirmed loss of major molecular response (MMR; BCR-ABL1 transcript levels < 0.1%), mutations, or clonal chromosome abnormalities in Ph-cells at any subsequent time during therapy.[5] [8]

Acquisition of mutations in the tyrosine kinase domain (TKD) is the best characterized among the BCR-ABL1-dependent mechanism.[9] [10] Mutations in the TKD interfere with the binding of TKI molecule to their targeted site in the BCR-ABL1 protein.[5] [11] Detection of mutations in BCR-ABL1 gene can predict the outcome and risk of relapse of a CML patient. The presence of a TKD mutation or compound mutations can aid in prognostication and appropriate TKI selection. More than 100 mutations have been identified in the kinase domain (KD) region and the current 2GTKIs and third-generation (3G)-TKIs have shown variable sensitivity toward specific mutation profiles.[5] [12] T315I, the “gatekeeper mutation” is one of the most frequently detected single-nucleotide variants that evades all TKIs except the 3GTKI, ponatinib.[13] The spectrum of KD mutations is expanding, that makes the assessment of resistant CML critical and warrants tailored treatment for individual patients keeping in mind the goal of therapy, that is, overall survival or treatment-free remission.

Although there have been a few studies describing the BCR-ABL1 KD mutation patterns from India; however, these studies are restricted to specific geographic locations.[7] [14] [15] [16] [17] [18] [19] Herein, we sought to identify and characterize the patterns of KD mutation in one of the largest cohorts of Indian CML patients across the country, especially those who presented with resistance or intolerance to frontline therapy.


#

Materials and Methods

Patient Characteristics and Data Collection

This retrospective study included a total of 791 patients of CML referred to our laboratory during the period of August 2018 to August 2019 for BCR-ABL1 KD mutation test upon approval from the institutional review board. The diagnoses of CML were based on clinical, hematologic, and cytogenetic profile of the patient. All patients included in the cohort met the two following criteria: (1) confirmed CML diagnosis undergoing TKI therapy, (2) BCR-ABL1 mutation analysis performed in our laboratory during the fixed time period of the data analysis, that is, August 2018 to August 2019. Data collection for each patient included age, gender, contact information, geographical location, clinical history, ongoing therapy, BCR-ABL1 quantitative test results, if any, and BCR-ABL1 KD mutation test results. The clinical, treatment-related, and follow-up parameters wherever available were charted ([Table 2]).


#

BCR-ABL1 KD Mutational Analysis

The tests were prescribed under the suspicion of resistance to ongoing therapy. Indications of mutation analysis were based on the clinical practice guidelines.[1] [8] [20] Loss of hematologic response (HR), CyR, or molecular response or delay in attaining defined landmarks was indications for recommending TKD mutation analysis. Poor response was defined as not achieving HR, CyR, and MMR at the defined time points by ELN.[21]


#

Sample Procurement

For each patient, 3 to 5 mL blood or bone marrow sample was collected in an ethylenediaminetetraacetic acid vial. Each sample was checked for cell count using an automated cell counter (Sysmex, USA). Blood/bone marrow sample acceptance criteria includes observed stability of specimen, ≥ 3 mL of sample in the vial, and total leucocyte count ≥ 1,000/µL.[22] Samples not meeting the criteria were duly rejected.


#

Ribonucleic acid Extraction and Complementary DNA Preparation

Total ribonucleic acid (RNA) was extracted using QiaAmp RNA blood mini kit (Qiagen, United States) as per the manufacturer's protocol from the 3 to 5 mL whole blood or bone marrow. The purified total RNA was evaluated for concentration (OD260) and purity (OD260/280) by spectrophotometric method (Nanodrop, Thermoscientific, United States). Since optimal assay sensitivity is dependent on the RNA input, it was ensured to use an RNA isolation method yielding greater than 200 ng/μL and to target greater than 1,000 ng total RNA input per test. Purified total RNA was analyzed immediately after extraction or stored below –20°C until ready for testing.


#

Polymerase Chain Reaction and Sanger Sequencing

As the first step toward polymerase chain reaction (PCR) analysis, complementary DNA was prepared from a minimum of 100 ng quality-checked RNA using reverse transcriptase (3B Black Biotech, India) according to the manufacturer's protocol. A two-step PCR assay is conducted for our in-house routine KD mutation analysis. The first step included the amplification of the BCR-ABL fusion transcript followed by a second PCR reaction which amplified 579 base pair of the ABL1 KD region for any mutations present. Finally, the amplicon was subjected to Sanger sequencing using the SeqStudio genetic analyzer (Applied Biosystems, United States) to identify point mutations. The generated sequencing data was analyzed using SeqStudio analysis software (Applied Biosystems).


#

Statistical Analysis

All statistical analysis and calculations were done using GraphPad prism8 software and Microsoft Excel. Mutation rates were calculated and frequency patterns were compared in each mutation group. Mann–Whitney test and two-tailed Student's t-test were used to assess association of variables wherever applicable. A p-value of < 0.05 was considered to indicate statistically significant difference.


#

Ethical Approval

The procedures followed were in accordance with the ethical standards of the responsible committee on human experimentation and with the Helsinki Declaration of 1964, as revised in 2013.


#
#

Results

A total of 791 CML patient samples reported in this study were screened for BCR-ABL1 KD mutations following TKI resistance of which 236 (29.8%) presented with single or multiple mutations in the KD. In the overall cohort, the male:female ratio was 1.8:1 and median age was 42 years. A majority (n = 106, 45%) of the patients presenting with resistance was between 30 and 50 years. In mutation positive patients (n = 236), the male:female ratio was 2.4:1 and median age was 41 years (data not shown). Mutation positive cases were subjected to further analysis ([Fig. 1]).

Zoom Image
Fig. 1 Flowchart describing study design: Distribution and segregation of patients according to kinase domain (KD) mutation status and number of patients selected for each analysis step.

Distribution of Kinase Domain Mutation

In this cohort, 38 types of mutations were detected ([Table 1]). Among the most prevalent mutations, single-occurring T315I was the most frequent as observed in 15.67% (n = 37/236) cases followed by E255K (10.16%, n = 24/236) and M244V (9.32%, n = 22/236) ([Fig 2]). Each specific mutation, categorized based on their functional position in the KD, were identified and listed along with the number of cases that harbored them ([Table 1]).

Table 1

Distribution and frequency of mutations based on their structural positions in the kinase domain with most frequent (in bold) and novel mutation (in bold italics)

Structure

Hotspot mutation

No. of cases[a] (N = 236)

Frequency

M244V

30

12.7118644

P-loop

L248V

5

2.11864407

G250E

19

8.05084746

Q252H

6

2.54237288

Y253F

1

0.42372881

Y253H

15

6.3559322

E255K

32

13.559322

E255V

6

2.54237288

C-helix

E275K

2

0.84745763

D276G

4

1.69491525

E279K

6

2.54237288

I293V

1

0.42372881

L298P

1

0.42372881

V299L

2

0.84745763

ATP-binding site

F311L

3

1.27118644

F311I

11

4.66101695

T315I

48

20.3389831

F317L

17

7.20338983

SH2

M351T

10

4.23728814

E355G

4

1.69491525

Substrate binding site

F359V

22

9.3220339

F359I

3

1.27118644

A-loop

E373K

1

0.42372881

L387M

3

1.27118644

M388L

2

0.84745763

H396P

3

1.27118644

H396R

9

3.81355932

H396Y

1

0.42372881

A397P

1

0.42372881

a Total number of cases (with frequency) in which mutation was detected either single or with a comutation.


Zoom Image
Fig. 2 Mutations detected in BCR-ABL1 kinase domain (KD). Pie chart representation of KD mutations detected across the cohort in order of their frequency and type.

A subset of cases (13.3%, n = 33/236) harbored compound mutations, that is, two or more mutations ([Supplementary Table S1, available online only]). A novel mutation L298P was detected in one case with T3151 as comutant. Three cases harbored triple mutations, E255K/T315I/F317L, E279K/G250E/M244V, and G250E/L248V/H396R. In patients with complex mutations, T3151 was the most frequent mutation partner observed (n = 11/33) followed by M244V (n = 8/33) and G250 (n = 6/33) ([Supplementary Table S1]).


#

Treatment Follow-Up

Therapy timeline and molecular response data since diagnosis was available for 28 patients with detected mutations detailed in [Table 2]. The average time taken to assess for a TKD mutation was 3 years after start of first-line TKI. Imatinib 400 mg was the choice of first-line therapy for most of these cases (89.2%). Postdetection of resistant mutation, patients with T315I mutations were administered the 3G ponatinib 45 mg (n = 4). All other TKD mutation carrier patients were administered a 2G TKI dasatinib with dosage range of 50 to 100 mg once daily (n = 10) or nilotinib 400 mg twice daily (n = 9) and 300 mg twice daily (n = 1). A few patients continued on imatinib at the same or higher dose of 600 to 800 mg (n = 4). A third TKI was required by eight patients after their previous two TKIs produced suboptimal response. One patient (E255K) chose to discontinue TKI therapy and has not been included in [Table 2]. There was no significant association of mutation with age or gender. Among patients with TKD mutation, 50% (n = 12/24) patients reached MMR after a TKI shift within the median time of 7 months and median time of 40 months (∼3.5 years) since diagnosis. Death occurred in 60% (n = 3/5) T315I carriers. Patients with F317L, F359V showed 100% survival and E255K/V showed 66% survival post-TKI shift. The survival was not statistically significant between mutant groups F359V/F317L versus T315I (p-value = 0.07) or E255K versus T315I (p-value =0.3) ([Table 3]).

Table 2

Details of tyrosine kinase inhibitor shifts and associated response in 28 patients on follow-up

Age

Gender

Disease phase

1st line TKI

Time of first mutation analysis since diagnosis (mo)

Mutation detected

2nd line TKI

Time of follow-up since TKI shift (mo)

BCR-ABL1 transcript level % (other symptom)

TKD analysis repeated (months since diagnosis)

Mutation detected

3rd line TKI

Time of follow-up since TKI shift (mo)

BCR-ABL1 transcript level % (other symptom)

Other

Status (March 2020)

51

M

CML-CP

Imatinib

16

No mutation

Dasatinib

14

0.3

MR

Yes (36)

E255K

Nilotinib

5

0.1

MMR

Alive

59

M

CML-CP

Imatinib

24

E255K

Dasatinib

3

19.1 (cytopenia)

Not tested

Not tested

Nilotinib

5

Not tested

Alive

36

M

CML-CP

Imatinib

24

F359V, G250E

Dasatinib

24

33.4

Yes (48)

E255K

Bosutinib

1

No response

Dead

28

M

CML-CP

Imatinib

96

E255K

Imatinib 600 mg

13

Not tested

Alive

80

M

CML-CP

Imatinib

84

E255V

Dasatinib

12

Not tested

Alive

33

M

CML-CP

Imatinib

24

E255V

Nilotinib

2

No response

Dead

16

M

CML-CP

Imatinib

19

E279K

Nilotinib

7

0.1

MR

Alive

46

M

CML-CP

Imatinib

132

E355G

Dasatinib

7

0.003 MR 4.5

Alive

40

M

CML-CP

Nilotinib

56

F311I

Nilotinib resumed

12

0.12

MR

Alive

45

M

CML-CP

Imatinib

204

F311L

Imatinib 800 mg

4

No response

Dead

37

M

CML-CP

Imatinib

36

F317L

Nilotinib

4

0.6

∼CCyR

Alive

48

F

CML-CP

Imatinib

60

F317L

Nilotinib

4

Not tested

Alive

42

M

CML-CP

Imatinib

27

F317L

Dasatinib

7

23.9

No

No

Nilotinib

6

5.21

∼PCyR

Alive

25

F

CML-CP

Imatinib

144

F359V

Nilotinib

48

Not tested (leukocytosis)

Dasatinib

12

Not tested

Alive

47

F

CML-CP

Dasatinib

168

F359V

Nilotinib

3

1.96

> PCyR

Alive

39

M

CML-CP

Nilotinib

15

F359V

Dasatinib

3

0

DMR

Alive

29

M

CML-CP

Imatinib

48

G250E

Nilotinib

8

0

DMR

Alive

45

M

CML-CP

Imatinib

Not tested

Nilotinib

Not tested

Not tested (intolerance)

Yes (9)

G250E

Dasatinib

15

2.9

∼CCyR

Alive

51

M

CML-CP

Imatinib

144

H396P

Dasatinib

12

0.29

∼MR

Alive

39

F

CML-CP

Imatinib

72

H396R

Nilotinib

6

No response

Dead

30

F

CML-CP

Imatinib

Not tested

Dasatinib

60

28.82

Yes (192)

M244V

Nilotinib

7

1.91

∼CCyR

Alive

29

M

CML-CP

Imatinib

24

Y235H

Imatinib 800 mg

6

Not tested (leukocytosis)

Not tested

Not tested

Dasatinib

3

No data

Alive

36

M

CML-CP

Imatinib

24

M388I

Imatinib 400 mg continued

6

0.07

MMR

Yes(6)

No mutation detected

Alive

26

M

CML-CP

Imatinib

24

No mutation detected

Nilotinib

7

No response

Yes (72)

T315I

Ponatinib

7

No response

Dead

37

M

CML-CP

Imatinib

Not tested

Nilotinib

3

Not tested (T-lymphoid blast crisis)

Yes (84)

T315I

Ponatinib

12

No response

ASCT

Dead

53

F

CML-CP

Imatinib

No data

T315I

Dasatinib

3

15.24

No

No

Ponatinib

8

0.04

MMR

Dead

32

M

CML-CP

Imatinib

Not tested

Nilotinib

60

Not tested

Yes (84)

T315I

Ponatinib

15

0.01

MMR4

Alive

52

M

CML-CP

Imatinib

6

T315I

Imatinib 800 mg

5

4.18

∼PCyR

Alive

Abbreviations: CCyR, complete cytogenetic response; CyR, cytogenetic response; CML, chronic myeloid leukemia; CP, chronic phase; DMR, deep molecular response; MMR, major molecular response; MR, molecular response; PCyR, partial cytogenetic response.


Table 3

Survival analysis of the patients harboring mutations in selected groups with criteria set as: three or greater than three patients carrying a mutation

Mutations

Rate of survival (%)

Median time to reach MMR since last TKI shift (mo)

Median time to reach CyR/MMR response since diagnosis (mo)

Survival vs. T315I group p-value (significant)

E255K

66.6

5

51.5

0.3 (No)

F317L

100

5

40

0.07 (No)

F359V

100

3

94

0.07 (No)

T315I

40%

11

85

Abbreviations: CyR, cytogenetic response; MMR, major molecular response; TKI, tyrosine kinase inhibitor.



#
#

Discussion

CML treatment has undergone a sea change since the 20th century, beginning with radiotherapy, busulfan, and hydroxycarbamide, followed by interferon-alpha. During the 1980s, allogeneic stem cell transplant became the treatment of choice for eligible patients but not without risk of mortality.[23] In 1998, introduction of the first TKI, imatinib, revolutionized treatment for newly diagnosed CML by bringing up the survival of treated CML patients comparable to a healthy individual.[24] This TKI has shown promising results and still remains a first-line therapy for CML,[5] as in our patient group where we observe 90% patients being treated with imatinib as first-line TKI. Despite the achievements, there has been a major hurdle in the complete success of imatinib, in the form of acquired resistance and intolerance to the drug.

Over the past two decades, studies have already identified the mechanisms of drug resistance, that include drug metabolism, bioavailability, leukemic cell evolution, and the most frequently responsible KD point mutations or overexpression of the BCR-ABL1 gene.[9] In one of the earliest comprehensive analyses, 144 Australian patients were evaluated for BCR-ABL1 kinase mutations and a highly significant number of patients (89%) with an underlying mutation developed resistance to imatinib therapy.[25] Ever since, multiple studies have proven the point that close molecular monitoring and prompt mutation analysis is critical for disease management and long-term remission.

There are limited studies available on Indian patients[15] [17] [19] [26]; with the most recent next-generation sequencing (NGS)-based study by Chandrasekhar et al that identifies TKD mutations in 50% (n = 21/41) patients, with 13 patients harboring 6 novel missense mutations,[2] a relatively high incidence possibly owing to deep sequencing methodology ([Table 4]). In this report, we present data of one of the largest subsets of CML patients (n = 791), who were assessed after failing to achieve response milestones post-first-line TKI and observed the presence of TKD mutations in 29.8% patients.

Table 4

Comparison of our study with the previous published literature from India

Characteristics

Present study

Chandrasekhar et al 2019

Chaitanya et al 2017

Tripathi et al 2017

Patkar et al 2016

Rajappa et al 2013

Markose et al 2009

Cohort size (n)

791

41

269

46

385

90

76

Age range (median age)

10–85 (42)

29–68 (53.73)

18–66 (36)

15–72 (36.8)

7–73

18–65 (36)

(44)

Male-to-female ratio

1.8:1

1.1:1

1.7:1

1.3:1

3:1

2.4:1

No data

Phases of CML included

29 CP (follow-up)

25 CP,11 AP, and 5 BC

CP

45 CP and 1 AP

331 CP, 29 AP, and 25 BC

CP

54 CP, 14 AP, 8 BC

Frequency of mutations in KD

29.8%

51.2%

32.7%

40%

51.9%

32.2%

33%

Most common mutations

T315I (20%), E255K (14%), M244V (13%)

T315I (33.3%)

F317L (33.3%), novel mutations (61.9%)

T315I (31.8%), E255V/K (5.7%)

M351T (43.75%), Y253H (18.75%),

H396R (18.75%)

T315I (20.7%), Y253H (10.1%), M244V (7.2%), F317L (7.2%)

T315I (31%), G250E (27.6%),

F359V (13.8%)

M244V (16%), T315I (16%), E255K (12%)

Abbreviations: AP, accelerated phase; BC, blast crisis; CML, chronic myeloid leukemia; CP, chronic phase; KD, kinase domain.


This is in corroboration with previous Indian patient reports by Chaitanya et al[16] and Rajappa et al[15] describing TKD mutations in 33 and 29% CML patients, respectively. Overall, the incidence of TKD mutation has been reported in 30 to 50% imatinib-intolerant patients worldwide.[27] [28] [29] [30] Gender distribution of the present cohort is comparable to Chandrasekhar et al,[2] Tripathi et al,[7] and Chaitanya et al[16] but is significantly different from Patkar et al[19] and Rajappa et al.[15] We also observe a slightly higher median age in comparison to other studies ([Table 2] and [4]). In addition, treatment resistant cases that harbored a mutation (n = 236) in the BCR-ABL1 KD show that the female gender has a markedly lower presentation of resistant mutations suggesting alternative mechanisms that may possibly be contributing to their suboptimal response to therapy.[9] [31]

In the spectrum of mutations found, we found one novel missense mutation L298P whose clinical significance warrants further investigation. It was observed with T315I as a comutant which may possibly add to the complexity while deciding the therapy options. In consistency with worldwide data,[32] the “gatekeeper mutation” T315I in the substrate binding site occurs most frequently in our patients (20%) as it is considered the primary mechanism of the BCR-ABL1 positive cells to escape TKI pressure with enhanced leukemogenesis.[33] T315I+ cells renders all TKIs ineffective, except ponatinib due to a different binding mechanism of the drug to the tyrosine kinase protein.[34] The T315I evidently confers poor prognosis in CML patients as seen in our patients with majority (60%) of deaths as compared to other mutations. However, this observation is based on a small subset of patients (n = 5) and warrants a clinical follow-up of a bigger cohort. It is important to mention that one of the factors behind this dismal outcome was the socioeconomic status of the patients who could not afford or procure ponatinib, a situation that needs to be addressed for CML management in India.

Apart from imatinib, resistant mutations have been observed postadministration of 2G TKIs as well. The F317L, V299L, and T315A mutations are reported to impart a high degree of resistance to dasatinib.[5] In our patients with F317L on follow-up, all three patients with the mutation ([Table 2]) showed a significant response with nilotinib, with one of them unresponsive to dasatinib before shifting to nilotinib, while one completely failing on continued imatinib leading to death. We observed no incidence of T315A mutation in this cohort. V299L was detected in two patients and this mutation rarely emerges after imatinib therapy and is reported to emerge post-dasatinib as second-line therapy. We lost these two patients to follow-up and could not assess their clinical course. The more frequent mutations within the P-loop (Y253H/F, E255V/K) and F359 mutations were associated with a high degree of resistance to nicotinic in this study. The three patients who harbored F359V mutation ([Table 2]) showed a trend of shifting to dasatinib to reach CyR or MMR. E255K/V was the second most frequent mutation (16%; n = 38) observed in our patient group, similar to incidence reported in previous patient studies (8–17%) and is associated with poor prognosis.[13] [35] We observe a variable trend in TKI shift in the six patients with E255K/V mutations with varying response to both dasatinib and nilotinib, and one continuing on a higher imatinib dose without 2GTKI shift. One patient with Y253H who is currently on dasatinib as second-line therapy after 30 months of resistance to imatinib is stable and awaiting BCR-ABL1 transcript levels. However, a larger patient set would be needed to assess the statistical significance of a specific mutation and their TKI course.

Tripathi et al in 2017, in contrast to previous studies, reported M351T as the most frequent mutation (43.5%) in their patient group and surprisingly no T315I, touting geographical variation as a possible reason. In our patient group M351T is found in a much lower frequency (4.3%) from patients Pan-India. A comparative analysis by Kim et al between Asians and non-Asian population cohorts, has stated the distinct pattern of mutations in Asians with highest prevalence of T3151 and P-loop mutations in Asians that has been reinstated this study.[36] It has been observed that patients who achieve molecular response are less likely to relapse to a high leukemia burden, hence achieving this milestone has become a primary objective during TKI therapy.[37] Among our patients who were followed up, each subject presented with BCR-ABL1 level > 10% (HR) before TKD mutation analysis. We observed MMR achievement after TKI shift in half of the patients in follow-up. Regular molecular quantitative monitoring of BCR-ABL1 plays a crucial role in identifying lack of response and must be adhered to for maximum benefits. Lack of testing, irregularities in follow-up, and economic burden are some of the factors behind treatment gaps in majority of patients. Evidently, the relatively small follow-up data available for this patient cohort is a limitation of this study and must be addressed in future studies.

It has been shown that sequential exposure to TKIs lead to accumulation of compound (≥ 2 mutations in the same BCR-ABL1 clone) or polyclonal mutations (≥ 2 mutations in different BCR-ABL1 clones).[38] Double/triple mutations were observed in 14% patients (n = 33) in our cohort. Compound mutations are known to arise under 2G- or 3GTKI pressure, partnering with preexisting mutation against first-line or second-line TKI and pose a clinical challenge. The combination of mutations is associated with particular TKI administered as dasatinib and nilotinib are known to give rise to distinct compound mutation profiles. Double mutations are the most common form detected as reiterated by our analysis along with T315I observed as the most frequent mutation partner.[39] Triple mutations found in this patient group characterized as E255K/T315I/F317L, E279K/G250E/M244V, and G250E/L248V/H396R have not been reported previously. Presence of compound mutations gives us insight into the complex interplay of TKIs and CML clonal behavior.

Sanger sequencing is currently the routine technique for mutation analysis with a marked specificity and relative affordability. Nevertheless, this technique is limited in sensitivity to detect mutations at a frequency of 20 to 30%.[13] This must be considered when patients are tested and detected with “no mutations” despite showing clinical signs of intolerance or resistance to therapy. The possibility of missing out on mutant clones at very low variant frequency (< 10%) can be addressed with advanced technologies such as the NGS. Therefore, NCCN recommends the use of NGS, if available.[5] Identifying the low burden mutant clones can be advantageous as a preemptive effort to reduce leukemia burden in the overall course of disease.


#

Conclusion

Knowledge on CML continues to evolve in context to molecular diagnostics and clinical management. It is advisable to perform BCR-ABL1 KD mutation analysis before administering a new TKI at any time point to attain desirable results. Novel mutations such as L298P as identified in this study can have distinct response to TKIs and thus, are recommended to be monitored for their clinical significance. Regular quantitation of BCR-ABL1 transcript according to the treatment guidelines is critical for evaluating appropriate response to TKIs as each TKI seems to show a distinct clinical profile. In addition, NGS-based mutation analysis could benefit patients at baseline to aid in the decision of the correct frontline TKI for the individual by identifying preexisting KD mutations at low frequencies independent of TKI selection pressure.


#
#

Conflict of Interest

None declared.

Acknowledgment

We are thankful to all our esteemed clinicians for their support and Ms. Ayushi Singh for her assistance. A part of this research has been presented as an abstract [poster] at the Association of Molecular Pathologists' Annual Meeting at San Antonio, TX (November 1st to 3rd, 2018).

* These authors have contributed equally and share the first authorship.


Supplementary Material

  • References

  • 1 Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol 2018; 93 (03) 442-459
  • 2 Chandrasekhar C, Kumar PS, Sarma PVGK. Novel mutations in the kinase domain of BCR-ABL gene causing imatinib resistance in chronic myeloid leukemia patients. Sci Rep 2019; 9 (01) 2412
  • 3 Iqbal N, Iqbal N. Imatinib: a breakthrough of targeted therapy in cancer. Chemother Res Pract 2014; 2014: 357027
  • 4 Rosti G, Castagnetti F, Gugliotta G, Baccarani M. Tyrosine kinase inhibitors in chronic myeloid leukaemia: which, when, for whom?. Nat Rev Clin Oncol 2017; 14 (03) 141-154
  • 5 Hochhaus A, Baccarani M, Silver RT. et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia 2020; 34 (04) 966-984
  • 6 Bhamidipati PK, Kantarjian H, Cortes J, Cornelison AM, Jabbour E. Management of imatinib-resistant patients with chronic myeloid leukemia. Ther Adv Hematol 2013; 4 (02) 103-117
  • 7 Tripathi AK, Verma SP, Kumar N. Mutation analysis in chronic myeloid leukemia patient in chronic phase on imatinib having delayed achievement of milestones or loss of response. Indian J Hematol Blood Transfus 2017; 33 (03) 316-320
  • 8 Hochhaus A, Saussele S, Rosti G. et al. Chronic myeloid leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2018; 29 (Supplement_4): iv261
  • 9 Milojkovic D, Apperley J. Mechanisms of resistance to imatinib and second-generation tyrosine inhibitors in chronic myeloid leukemia. Clin Cancer Res 2009; 15 (24) 7519-7527
  • 10 Patel AB, O'Hare T, Deininger MW. Mechanisms of resistance to ABL kinase inhibition in CML and the development of next generation ABL kinase inhibitors. Hematol Oncol Clin North Am 2017; 31 (04) 589-612
  • 11 Radich JP, Deininger M, Abboud CN. et al. Chronic myeloid leukemia, Version 1.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2018; 16 (09) 1108-1135
  • 12 Soverini S, Abruzzese E, Bocchia M. et al. Next-generation sequencing for BCR-ABL1 kinase domain mutation testing in patients with chronic myeloid leukemia: a position paper. J Hematol Oncol 2019; 12 (01) 131
  • 13 Soverini S, Branford S, Nicolini FE. et al. Implications of BCR-ABL1 kinase domain-mediated resistance in chronic myeloid leukemia. Leuk Res 2014; 38 (01) 10-20
  • 14 Srivastava S, Dutt S. Imatinib mesylate resistance and mutations: an Indian experience. Indian J Med Paediatr Oncol 2013; 34 (03) 213-220
  • 15 Rajappa S, Mallavarapu KM, Gundeti S, Roshnipaul T, Jacob RT, Digumarti R. Kinase domain mutations and responses to dose escalation in chronic myeloid leukemia resistant to standard dose imatinib mesylate. Leuk Lymphoma 2010; 51 (01) 79-84
  • 16 Chaitanya PK, Kumar KA, Stalin B, Sadashivudu G, Srinivas ML. The role of mutation testing in patients with chronic myeloid leukemia in chronic phase after imatinib failure and their outcomes after treatment modification: single-institutional experience over 13 years. Indian J Med Paediatr Oncol 2017; 38 (03) 328-333
  • 17 Kagita S, Uppalapati S, Jiwatani S. et al. Incidence of Bcr-Abl kinase domain mutations in imatinib refractory chronic myeloid leukemia patients from South India. Tumour Biol 2014; 35 (07) 7187-7193
  • 18 Sharma P, Mohanty S, Kochupillai V, Kumar L. Mutations in ABL kinase domain are associated with inferior progression-free survival. Leuk Lymphoma 2010; 51 (06) 1072-1078
  • 19 Patkar N, Ghodke K, Joshi S. et al. Characteristics of BCR-ABL kinase domain mutations in chronic myeloid leukemia from India: not just missense mutations but insertions and deletions are also associated with TKI resistance. Leuk Lymphoma 2016; 57 (11) 2653-2660
  • 20 Döhner H, Estey E, Grimwade D. et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017; 129 (04) 424-447
  • 21 Baccarani M, Deininger MW, Rosti G. et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood 2013; 122 (06) 872-884
  • 22 maxwell-rsc-simplyrna-blood-kit-protocol.pdf. Accessed November 4, 2020, at: https://www.promega.com/-/media/files/resources/protocols/technical-manuals/101/maxwell-rsc-simplyrna-blood-kit-protocol.pdf?la=en
  • 23 Goldman JM. Chronic myeloid leukemia: a historical perspective. Semin Hematol 2010; 47 (04) 302-311
  • 24 Deininger MW. Chronic myeloid leukemia: an historical perspective. Hematology (Am Soc Hematol Educ Program) 2008; 2008 (01) 418-418
  • 25 Branford S, Rudzki Z, Walsh S. et al. Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood 2003; 102 (01) 276-283
  • 26 Bagadi S, Saikia T, Pany A, Das B. Analysis of ABL kinase domain mutations conferring resistance to tyrosine kinase inhibitors in chronic myeloid leukemia cases from India. Clin Lab 2011; 57 (7-8): 619-623
  • 27 Marcé S, Zamora L, Cabezón M. et al; Grupo ICO de estudio de mutaciones de ABL en pacientes afectos de LMC. Frequency of ABL gene mutations in chronic myeloid leukemia patients resistant to imatinib and results of treatment switch to second-generation tyrosine kinase inhibitors. Med Clin (Barc) 2013; 141 (03) 95-99
  • 28 Elias MH, Baba AA, Azlan H. et al. BCR-ABL kinase domain mutations, including 2 novel mutations in imatinib resistant Malaysian chronic myeloid leukemia patients-Frequency and clinical outcome. Leuk Res 2014; 38 (04) 454-459
  • 29 Lau A, Seiter K. Second-line therapy for patients with chronic myeloid leukemia resistant to first-line imatinib. Clin Lymphoma Myeloma Leuk 2014; 14 (03) 186-196
  • 30 Markose P, Chendamarai E, Balasubramanian P. et al. Spectrum of BCR-ABL kinase domain mutations in patients with chronic myeloid leukemia from India with suspected resistance to imatinib-mutations are rare and have different distributions. Leuk Lymphoma 2009; 50 (12) 2092-2095
  • 31 Bixby D, Talpaz M. Seeking the causes and solutions to imatinib-resistance in chronic myeloid leukemia. Leukemia 2011; 25 (01) 7-22
  • 32 Soverini S, Hochhaus A, Nicolini FE. et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood 2011; 118 (05) 1208-1215
  • 33 Mian AA, Schüll M, Zhao Z. et al. The gatekeeper mutation T315I confers resistance against small molecules by increasing or restoring the ABL-kinase activity accompanied by aberrant transphosphorylation of endogenous BCR, even in loss-of-function mutants of BCR/ABL. Leukemia 2009; 23 (09) 1614-1621
  • 34 Gibbons DL, Pricl S, Kantarjian H, Cortes J, Quintás-Cardama A. The rise and fall of gatekeeper mutations? The BCR-ABL1 T315I paradigm. Cancer 2012; 118 (02) 293-299
  • 35 Naqvi K, Cortes JE, Luthra R. et al. Characteristics and outcome of chronic myeloid leukemia patients with E255K/V BCR-ABL kinase domain mutations. Int J Hematol 2018; 107 (06) 689-695
  • 36 Kim H, Kim S, Kim H-J. et al. Comparison of frequency and sensitivity of BCR-ABL1 kinase domain mutations in Asian and white patients with imatinib-resistant chronic-phase chronic myeloid leukemia. Clin Lymphoma Myeloma Leuk 2018; 18 (10) e391-e399
  • 37 Mahon F-X. Treatment-free remission in CML: who, how, and why?. Hematology (Am Soc Hematol Educ Program) 2017; 2017 (01) 102-109
  • 38 Akram AM, Iqbal Z, Akhtar T. et al. Presence of novel compound BCR-ABL mutations in late chronic and advanced phase imatinib sensitive CML patients indicates their possible role in CML progression. Cancer Biol Ther 2017; 18 (04) 214-221
  • 39 Khorashad JS, Kelley TW, Szankasi P. et al. BCR-ABL1 compound mutations in tyrosine kinase inhibitor-resistant CML: frequency and clonal relationships. Blood 2013; 121 (03) 489-498

Address for correspondence

Sambit K. Mohanty, MD, FCAP, FRCPath
Department of Pathology and Laboratory Medicine
CORE Diagnostics, Fortune Tower II, Udyog Vihar Phase 3, Gurgaon, Haryana 122008
India   
Email: sambit04@gmail.com   

Publication History

Article published online:
31 December 2022

© 2022. MedIntel Services Pvt Ltd. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

  • References

  • 1 Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol 2018; 93 (03) 442-459
  • 2 Chandrasekhar C, Kumar PS, Sarma PVGK. Novel mutations in the kinase domain of BCR-ABL gene causing imatinib resistance in chronic myeloid leukemia patients. Sci Rep 2019; 9 (01) 2412
  • 3 Iqbal N, Iqbal N. Imatinib: a breakthrough of targeted therapy in cancer. Chemother Res Pract 2014; 2014: 357027
  • 4 Rosti G, Castagnetti F, Gugliotta G, Baccarani M. Tyrosine kinase inhibitors in chronic myeloid leukaemia: which, when, for whom?. Nat Rev Clin Oncol 2017; 14 (03) 141-154
  • 5 Hochhaus A, Baccarani M, Silver RT. et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia 2020; 34 (04) 966-984
  • 6 Bhamidipati PK, Kantarjian H, Cortes J, Cornelison AM, Jabbour E. Management of imatinib-resistant patients with chronic myeloid leukemia. Ther Adv Hematol 2013; 4 (02) 103-117
  • 7 Tripathi AK, Verma SP, Kumar N. Mutation analysis in chronic myeloid leukemia patient in chronic phase on imatinib having delayed achievement of milestones or loss of response. Indian J Hematol Blood Transfus 2017; 33 (03) 316-320
  • 8 Hochhaus A, Saussele S, Rosti G. et al. Chronic myeloid leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2018; 29 (Supplement_4): iv261
  • 9 Milojkovic D, Apperley J. Mechanisms of resistance to imatinib and second-generation tyrosine inhibitors in chronic myeloid leukemia. Clin Cancer Res 2009; 15 (24) 7519-7527
  • 10 Patel AB, O'Hare T, Deininger MW. Mechanisms of resistance to ABL kinase inhibition in CML and the development of next generation ABL kinase inhibitors. Hematol Oncol Clin North Am 2017; 31 (04) 589-612
  • 11 Radich JP, Deininger M, Abboud CN. et al. Chronic myeloid leukemia, Version 1.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2018; 16 (09) 1108-1135
  • 12 Soverini S, Abruzzese E, Bocchia M. et al. Next-generation sequencing for BCR-ABL1 kinase domain mutation testing in patients with chronic myeloid leukemia: a position paper. J Hematol Oncol 2019; 12 (01) 131
  • 13 Soverini S, Branford S, Nicolini FE. et al. Implications of BCR-ABL1 kinase domain-mediated resistance in chronic myeloid leukemia. Leuk Res 2014; 38 (01) 10-20
  • 14 Srivastava S, Dutt S. Imatinib mesylate resistance and mutations: an Indian experience. Indian J Med Paediatr Oncol 2013; 34 (03) 213-220
  • 15 Rajappa S, Mallavarapu KM, Gundeti S, Roshnipaul T, Jacob RT, Digumarti R. Kinase domain mutations and responses to dose escalation in chronic myeloid leukemia resistant to standard dose imatinib mesylate. Leuk Lymphoma 2010; 51 (01) 79-84
  • 16 Chaitanya PK, Kumar KA, Stalin B, Sadashivudu G, Srinivas ML. The role of mutation testing in patients with chronic myeloid leukemia in chronic phase after imatinib failure and their outcomes after treatment modification: single-institutional experience over 13 years. Indian J Med Paediatr Oncol 2017; 38 (03) 328-333
  • 17 Kagita S, Uppalapati S, Jiwatani S. et al. Incidence of Bcr-Abl kinase domain mutations in imatinib refractory chronic myeloid leukemia patients from South India. Tumour Biol 2014; 35 (07) 7187-7193
  • 18 Sharma P, Mohanty S, Kochupillai V, Kumar L. Mutations in ABL kinase domain are associated with inferior progression-free survival. Leuk Lymphoma 2010; 51 (06) 1072-1078
  • 19 Patkar N, Ghodke K, Joshi S. et al. Characteristics of BCR-ABL kinase domain mutations in chronic myeloid leukemia from India: not just missense mutations but insertions and deletions are also associated with TKI resistance. Leuk Lymphoma 2016; 57 (11) 2653-2660
  • 20 Döhner H, Estey E, Grimwade D. et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017; 129 (04) 424-447
  • 21 Baccarani M, Deininger MW, Rosti G. et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood 2013; 122 (06) 872-884
  • 22 maxwell-rsc-simplyrna-blood-kit-protocol.pdf. Accessed November 4, 2020, at: https://www.promega.com/-/media/files/resources/protocols/technical-manuals/101/maxwell-rsc-simplyrna-blood-kit-protocol.pdf?la=en
  • 23 Goldman JM. Chronic myeloid leukemia: a historical perspective. Semin Hematol 2010; 47 (04) 302-311
  • 24 Deininger MW. Chronic myeloid leukemia: an historical perspective. Hematology (Am Soc Hematol Educ Program) 2008; 2008 (01) 418-418
  • 25 Branford S, Rudzki Z, Walsh S. et al. Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood 2003; 102 (01) 276-283
  • 26 Bagadi S, Saikia T, Pany A, Das B. Analysis of ABL kinase domain mutations conferring resistance to tyrosine kinase inhibitors in chronic myeloid leukemia cases from India. Clin Lab 2011; 57 (7-8): 619-623
  • 27 Marcé S, Zamora L, Cabezón M. et al; Grupo ICO de estudio de mutaciones de ABL en pacientes afectos de LMC. Frequency of ABL gene mutations in chronic myeloid leukemia patients resistant to imatinib and results of treatment switch to second-generation tyrosine kinase inhibitors. Med Clin (Barc) 2013; 141 (03) 95-99
  • 28 Elias MH, Baba AA, Azlan H. et al. BCR-ABL kinase domain mutations, including 2 novel mutations in imatinib resistant Malaysian chronic myeloid leukemia patients-Frequency and clinical outcome. Leuk Res 2014; 38 (04) 454-459
  • 29 Lau A, Seiter K. Second-line therapy for patients with chronic myeloid leukemia resistant to first-line imatinib. Clin Lymphoma Myeloma Leuk 2014; 14 (03) 186-196
  • 30 Markose P, Chendamarai E, Balasubramanian P. et al. Spectrum of BCR-ABL kinase domain mutations in patients with chronic myeloid leukemia from India with suspected resistance to imatinib-mutations are rare and have different distributions. Leuk Lymphoma 2009; 50 (12) 2092-2095
  • 31 Bixby D, Talpaz M. Seeking the causes and solutions to imatinib-resistance in chronic myeloid leukemia. Leukemia 2011; 25 (01) 7-22
  • 32 Soverini S, Hochhaus A, Nicolini FE. et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood 2011; 118 (05) 1208-1215
  • 33 Mian AA, Schüll M, Zhao Z. et al. The gatekeeper mutation T315I confers resistance against small molecules by increasing or restoring the ABL-kinase activity accompanied by aberrant transphosphorylation of endogenous BCR, even in loss-of-function mutants of BCR/ABL. Leukemia 2009; 23 (09) 1614-1621
  • 34 Gibbons DL, Pricl S, Kantarjian H, Cortes J, Quintás-Cardama A. The rise and fall of gatekeeper mutations? The BCR-ABL1 T315I paradigm. Cancer 2012; 118 (02) 293-299
  • 35 Naqvi K, Cortes JE, Luthra R. et al. Characteristics and outcome of chronic myeloid leukemia patients with E255K/V BCR-ABL kinase domain mutations. Int J Hematol 2018; 107 (06) 689-695
  • 36 Kim H, Kim S, Kim H-J. et al. Comparison of frequency and sensitivity of BCR-ABL1 kinase domain mutations in Asian and white patients with imatinib-resistant chronic-phase chronic myeloid leukemia. Clin Lymphoma Myeloma Leuk 2018; 18 (10) e391-e399
  • 37 Mahon F-X. Treatment-free remission in CML: who, how, and why?. Hematology (Am Soc Hematol Educ Program) 2017; 2017 (01) 102-109
  • 38 Akram AM, Iqbal Z, Akhtar T. et al. Presence of novel compound BCR-ABL mutations in late chronic and advanced phase imatinib sensitive CML patients indicates their possible role in CML progression. Cancer Biol Ther 2017; 18 (04) 214-221
  • 39 Khorashad JS, Kelley TW, Szankasi P. et al. BCR-ABL1 compound mutations in tyrosine kinase inhibitor-resistant CML: frequency and clonal relationships. Blood 2013; 121 (03) 489-498

Zoom Image
Atreye Majumdar
Zoom Image
Sambit K. Mohanty
Zoom Image
Fig. 1 Flowchart describing study design: Distribution and segregation of patients according to kinase domain (KD) mutation status and number of patients selected for each analysis step.
Zoom Image
Fig. 2 Mutations detected in BCR-ABL1 kinase domain (KD). Pie chart representation of KD mutations detected across the cohort in order of their frequency and type.