Cystic acne due to imatinib therapy for chronic myelocytic leukemia

2018 ◽  
Vol 25 (4) ◽  
pp. 972-974 ◽  
Author(s):  
Andrew Hwang ◽  
Andrew Iskandar ◽  
Michael del Rosario ◽  
Constantin A Dasanu

Imatinib mesylate is a tyrosine kinase inhibitor used in the treatment of several malignancies. Its use, however, is associated with a number of toxic effects including adverse cutaneous reactions. Herein, we present a case of facial cystic acne in a patient receiving imatinib therapy for chronic myelocytic leukemia. This side effect resolved with cessation of therapy. To the best of our knowledge, this clinical entity has never been previously reported in the medical literature.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4475-4475
Author(s):  
Yingjia Wang ◽  
Yanmin Zhao ◽  
Lizhen Liu ◽  
Weijie Cao ◽  
Kangni Wu ◽  
...  

Abstract Abstract 4475 Imatinib mesylate is a selective tyrosine kinase inhibitor targeting BCR-ABL fusion protein. In the therapy of either interferon refractory or newly diagnosed chronic myeloid leukemia (CML), imatinib significantly improved the prognosis of the disease, albeit with typically mild to moderate adverse effects. In recent years, increasing attention has been focused on the skin hypopigmentation developed after the initiation of imatinib therapy, especially in Asian countries such as India, with the incidence between 33% and 85%, though sparsely reported in western countries. The inhibition of c-kit by imatinib may be a possible explanation, but the exact mechanism for this hypopigmentation caused by imatinib and its potential risk in patients is poorly understood. Here we report our clinical observation of this side effect of imatinib therapy among Chinese CML patients in our institution, as well as a brief in vitro study aiming to investigate the mechanism of the hypopigmentory effect and evaluate its potential risk. Among 116 patients who took imatinib for CML therapy, 90 of them (77.6%) had reported to develop generalized hypopigmentation or whitening in the skin. This side effect usually developed within 3 months after initiation of imatinib, persistent during the therapy, and reversible upon imatinib discontinuation, according to our interviews and observation. More interestingly, in three of these CML patients who originally had vitiligo, the cosmetic appearance seemed to be improved during imatinib therapy due to the whitening effect on normal pigmented skin areas. To further confirm and understand this side effect, we examined the impact of imatinib on cultured normal human melanocytes derived from the foreskins of healthy Chinese adults. Melanocytes were treat with various concentrations of imatinib (1~16 μ M) for different periods of time (1~7 days). Cell proliferation and apoptosis were detected by cell counting kit-8 and flow cytometric assay, respectively. The melanogenic activity of the melanocytes was detected by melanin content assay and tyrosinase activity assay. Moreover, the role of stem cell factor (SCF) and its receptor c-kit in imatinib induced hypopigmentation were investigated. We found that imatinib not only concentration-dependently inhibited melanocyte proliferation in vitro (IC50=6.23 μ M), but also resulted in massive melanocyte apoptosis at concentrations higher than 8 μ M (P<0.01). Even at low concentrations (1-8 μ M), imatinib was able to cause decreases in total melanin content (P<0.05) and tyrosinase activity (P<0.05) which were responsible for skin pigmentation. In addition, imatinib (1-8 μ M) completely abrogated SCF (5 ng/ml) induced melanocyte proliferation (P<0.01) and melanogenesis (P<0.01), through the inhibition of the phosphorylation of c-kit receptor (Tyr 703 and Tyr 719) and the downstream signal transduction, which is pivotal for melanocyte proliferation and melanin synthesis after ultraviolet exposure. These results reveal that imatinib may cause destruction or dysfunction of melanocytes in patients through the inactivation of SCF/c-kit signaling, leading to inability of the skin to produce melanin and perform protection against ultraviolet radiation. Therefore, CML patients who take imatinib are necessarily to be warned for this side effect, and diminish the intensive exposure to sunlight. Although an ethnic or genetic basis for the prevalence difference can not be excluded yet, we tend to believe that the hypopigmentation effect of imatinib was probably underestimated in caucasian patients due to their light skin color. Since the melanocytes used in this study were all isolated from the skin samples of Chinese volunteers, further study may be done to determine whether the similar results can be achieved in primary melanocytes from other ethnic group. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3352-3352
Author(s):  
Klaus Podar ◽  
Melissa Simoncini ◽  
Yu-Tzu Tai ◽  
Martin Sattler ◽  
Kenji Ishitsuka ◽  
...  

Abstract The tyrosine kinase inhibitor adaphostin is a member of the tyrophostin family of small molecules that interfere with peptide binding rather, than targeting the kinase ATP-binding site. Adaphostin has therefore been examined as an alternative to the 2-phenylaminopyrimidine derivate imatinib mesylate, with remarkable efficacy in the treatment of chronic myeloic leukemia (CML). Previous studies show that adaphostin induces apoptosis: (1) in Bcr/Abl+ cells more rapidly than imatinib mesylate; (2) in imatinib mesylate resistant cells; and (3) in Bcr/ Abl - cells. Imatinib mesylate has minimal, if any activity in MM; the efficacy of adaphostin in multiple myeloma (MM) is unknown. Here we compare the effects of adaphostin and imatinib mesylate against human MM cells. Our results show concentration-dependent apoptosis in MM.1S, U266, OPM-2, INA-6, RPMI8226 and RPMI-Dox40 MM cells after treatment with adaphostin, but not with imatinib mesylate. Imatinib mesylate induced more than 50% apoptosis in K562 cells using concentrations as low as 1mM, which served as a positive control. Moreover, adaphostin, but not imatinib mesylate, induced caspase-9, caspase-8, and PARP cleavage, as well as downregulation of Mcl-1, in MM cells. Further results demonstrated that adaphostin induces peroxide production and DNA strand breaks after long-term treatment. Importantly MM cell proliferation induced by MM cell binding to BMSCs was abrogated by adaphostin- treatment. IL-6 and IGF-1 signaling and sequelae triggered by these cytokines are important growth, survival, and drug resistance factors in MM; conversely, adaphostin but not imatinib mesylate, inhibited phosphorylation of Src tyrosine kinase family, Akt-1, and ERK. Taken together, our studies in MM cells show that (1) adaphostin- inhibits IGF-1- and IL-6- triggered signaling pathways as well as (2) induces reactive oxygen species and apoptosis. These studies therefore provide the preclinical framework for its clinical evaluation to improve patient outcome in MM.


2017 ◽  
Vol 2017 ◽  
pp. 1-8 ◽  
Author(s):  
Rebecca M. Platoff ◽  
William F. Morano ◽  
Luiz Marconcini ◽  
Nicholas DeLeo ◽  
Beth L. Mapow ◽  
...  

Introduction. Recurrence of gastrointestinal stromal tumors (GISTs) after surgical resection and imatinib mesylate (IM) adjuvant therapy poses a significant treatment challenge. We present the case of a patient who underwent surgical resection after recurrence and review the current literature regarding treatment. Case Presentation. A 58-year-old man with a large intra-abdominal jejunal GIST was treated with complete surgical resection followed by IM. The patient experienced disease recurrence 3.5 years later and underwent IM dose escalation and reresection. Conclusion. Current strategies to treat recurrent GIST include dose escalation, modifying adjuvant tyrosine kinase inhibitor therapy, and surgery. High-level evidence will be required to better define the combinatory roles of tyrosine kinase inhibitor therapy, guided by molecular profiling, and surgery in the management of recurrent GIST.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2559-2559 ◽  
Author(s):  
Julia L. Close ◽  
Richard Lottenberg

Abstract Abstract 2559 Poster Board II-536 Background: To date, hydroxyurea (HU) is the only FDA-approved agent to treat sickle cell anemia (SCA). There is a need to identify alternative agents to prevent the complications of sickle cell disease. Clinical observations of patients receiving medications for other indications may provide insights into new opportunities. We present the clinical course of a patient with SCA and chronic myelocytic leukemia (CML) treated with imatinib. Case Report: A 24 year old woman with hemoglobin (Hb) SS had a variably elevated white blood cell (WBC) count for 3 years. WBC counts ranged from 11–39 × 103/cu mm; differential typically included 65–75% neutrophils, 15–20% lymphocytes, 1–13% metamyelocytes, and 1–3% myelocytes. Consistent with previous values, Hb ranged from 9–11 g/dL, and platelet counts from 250–400 × 103/cu mm. Real-time PCR for BCR-ABL obtained from the peripheral blood was positive. Bone marrow biopsy showed a markedly hypercellular marrow with granulocytic hyperplasia. Blasts were not increased in number. Cytogenetic studies revealed 46 XX, t(9;22)(q34;q11.2) in 20/20 analyzed cells. Further characterization by fluorescence in situ hybridization (FISH) technique demonstrated the loss of chromosome 9 long arm material. BCR-ABL interphase FISH confirmed the presence of 9/22 chromosomal translocation, found in 279/300 interphase nuclei. The patient was diagnosed with SCA in early childhood. Molecular analysis of genomic DNA revealed homozygosity for Hb S and single alpha-globin gene deletion of the rightward type. There was no evidence for typical deletions associated with HPFH or delta-beta zero thalassemia. The C>T polymorphism at nt −158 in the G-gamma-globin gene promoter region and other known HPFH point mutations in the G- or A-gamma globin genes were not identified. Her clinical course has not been complicated by stroke, pulmonary hypertension or acute chest syndrome. Radiographic studies revealed evidence of multiple bony infarcts. She was hospitalized with painful episodes 1-2 times per year, until the age of 16. At that time, her frequency of hospitalizations for painful episodes increased to 3-5 times per year, and showed a temporal relationship to her menses. She was started on oral contraceptives and hydroxyurea (HU) at the age of 18. Subsequently, hospitalizations for painful episodes decreased to 0-2 per year. At the age of 22, hydroxyurea was held during pregnancy. After delivery, HU was reinitiated and continued until the diagnosis of CML. She was hospitalized for painful episodes two times in the 12 months preceding the diagnosis of CML. Imatinib mesylate monotherapy at 400 mg/day was initiated. Two weeks later, a hematologic response was documented. At one year, bone marrow cytogenetic and molecular complete responses were documented. In the 17 months since initiation of imatinib, the patient has had a significant reduction in the frequency of painful episodes. No hospitalizations have been required and the patient has had a reduction in daily pain allowing discontinuation of a transcutaneous electrical nerve stimulation unit. During imatinib therapy Hb levels ranged from 8.0 to 9.2 g/dL, WBC counts from 3.3–6.1 × 103/cu mm, and platelet counts from 114–205 × 103/cu mm. Hemoglobin electrophoresis results are shown in the table. Clinical Implications: To our knowledge, the clinical course of patients with SCA treated with imatinib has not previously been reported. Imatinib targets the BCR-ABL fusion tyrosine kinase, c-kit, and platelet-derived growth factor receptor. Myelosuppression is frequently noted as a side effect of this therapy. The improvement in this patient's sickle cell symptoms may be attributable to a reduction in the neutrophil count, as observed with HU therapy, but it is possible imatinib therapy had a positive effect on release of cytokines or other factors involved in vaso-occlusion. The Hb F level was not significantly increased with imatinib therapy. Evaluation of small molecule tyrosine kinase inhibitors in experimental models of SCA should be considered. Disclosures: No relevant conflicts of interest to declare.


2008 ◽  
Vol 14 (5) ◽  
pp. 294-296 ◽  
Author(s):  
Kari K. Eklund ◽  
Ken Lindstedt ◽  
Charlotta Sandler ◽  
Petri T. Kovanen ◽  
Leena Laasonen ◽  
...  

PLoS ONE ◽  
2017 ◽  
Vol 12 (10) ◽  
pp. e0185704 ◽  
Author(s):  
Laurent L. Reber ◽  
Philipp Starkl ◽  
Bianca Balbino ◽  
Riccardo Sibilano ◽  
Nicolas Gaudenzio ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document