当前位置: 首页 > 期刊 > 《美国医学杂志》 > 2005年第1期 > 正文
编号:11119726
Treatment of Cockroach Allergen Asthma Model with Imatinib Attenuates Airway Responses
http://www.100md.com 《美国医学杂志》
     ABSTRACT

    In the present study it was determined whether a pharmacologic approach to blocking receptor tyrosine kinase-mediated activation during allergic airway responses could be beneficial. To examine these responses, allergic mice were given a single oral dose of imatinib at clinically relevant concentrations, ranging from 0.05 to 50 mg/kg, by oral gavages just before allergen challenge. The reduction in the allergen-induced responses was significant and centered on reducing overall inflammation as well as pulmonary cytokine levels. In particular, the treatment of the mice with imatinib significantly attenuated airway hyperreactivity and peribronchial eosinophil accumulation, and significantly reduced Th2 cytokines, interleukin-4 and interleukin-13. In addition, chemokines previously associated with allergen-induced pulmonary disease, CCL2, CCL5, and CCL6, were significantly reduced in the lungs of the imatinib-treated animals. Together these data demonstrate that the pharmacologic inhibitor imatinib may provide a clinically attractive therapy for allergic, asthmatic responses.

    Key Words: asthma ? eosinophils ? imatinib

    Peribronchial leukocyte accumulation is the hallmark of asthma, which affects a significant proportion of the worldwide population (1–6). The major pathophysiologic event that occurs during asthma is airway hyperreactivity (AHR) during the late-phase response. In particular, eosinophils have been reported to be the primary cell associated with induction of bronchial mucosal injury, and are believed to participate in bronchial obstruction and AHR (7–11). Several therapeutic strategies have focused on attenuating airway inflammation, including glucocorticoids and other agents that nonspecifically affect the response. The limited therapeutic options for the treatment of the disease likely reflect the lack of our understanding of the mechanisms that cause airway inflammation and hyperreactivity. A correlation can be drawn between the intensity of the leukocyte infiltration and the severity of the late-phase response. In particular, the activation and degranulation of eosinophils may be one of the causative events related to the exacerbation of airway disease (12–14). The apparent correlation with Th2 type immune responses has closely linked the intensity of the allergic responses with the production of interleukin (IL)-4 and IL-13 within the lungs of individuals with asthma.

    The activation of cytokine and growth factor receptors relies on activation of receptor tyrosine kinases for initiation of critical inflammatory and immune processes. Several drug development programs have begun to focus on inhibiting specific receptor tyrosine kinases. Most inhibitors have some nonspecific activity that makes it difficult to target a specific pathway. However, it appears that some success has been demonstrated for targeting specific classes of tyrosine kinases. The tyrosine kinase inhibitor imatinib mesylate (imatinib), also known as Gleevec, has revolutionized therapy for chronic myeloid leukemia with more than 90% of previously untreated patients in chronic phase achieving a complete hematologic response and more than 70% a complete cytogenetic response after one year of therapy (15–17). Specifically, imatinib was developed to inhibit BCR-ABL kinase activity. However, imatinib has been demonstrated to have significant efficacy in blocking both c-kit ligand, stem cell factor (SCF) and PDGFR- protein tyrosine kinase activity, whereas it spares most other kinase-activated pathways (18). In addition to being an attractive and well tolerated therapy for a number of cancerous diseases, imatinib mesylate has recently demonstrated efficacy in hypereosinophilic syndrome patients where few other options for therapy exist (19–21). Interestingly, a role for SCF overproduction in the airway during allergen-induced responses has been identified that was associated with alterations of eosinophil accumulation and hyperreactivity (22–25). Although SCF appears to be intimately involved in the initial phases of the response and mast cell activation, the down-stream effects on the late-phase response after blocking SCF function were profound and may be linked to the ability of SCF to directly activate eosinophils (23, 24, 26, 27). A most recent study indicated a significant increase in both SCF and c-kit expression in the airways of individuals with asthma related to the severity of disease (28). Thus, at least one of the targets of imatinib mesylate, SCF/c-kit activation pathway, may be a focus for therapy in asthmatic disease, but other receptor tyrosine kinases may also be inhibited by imatinib mesylate.

    METHODS

    Sensitization and Induction of the Airway Response

    To induce a TH2 type response, the following procedure was established in normal Balb/c mice, as previously described (29, 30). The mice were immunized with 10 μg of cockroach allergen (Holister-Stier, Spokane, WA) in incomplete Freund's adjuvant on Day 0. On Day 14 the mice were given an intranasal challenge of 10 μg of cockroach allergen in 10 μl of diluent to localize the response to the airway. Mice were then rechallenged 6 days later by intratracheal administration of 6 μg of cockroach allergen in 50 μl of sterile phosphate-buffered saline (PBS). Naive control animals were also given cockroach allergen intratracheally as a control. The magnitude of leukocyte recruitment in both the vehicle control- and cockroach allergen-challenged mice was examined histologically, and AHR was determined at 24 hours postchallenge.

    Enumeration of Peribronchial Eosinophil Accumulation

    Morphometric analysis of peribronchial eosinophil accumulation was performed from lungs of mice immunized and challenged with cockroach allergen on lungs preserved with 1 ml of 4% paraformaldehyde at 24 hours postchallenge. The fixed lungs were embedded in paraffin and multiple 50 μm sections were differentially stained with Wright-giemsa for the identification of eosinophils and viewed at 1,000 x. The individual eosinophils were counted from 50 high-powered fields per lung at each time point using multiple step sections of lung. Only the eosinophils in the peribronchial region were counted, which assured the enumeration of only those eosinophils within or immediately adjacent to an airway. The inflammation observed in this model is completely associated with the airway with little or no alveolitis.

    Measurement of AHR

    AHR was measured as previously described using a direct ventilation mouse plethysmograph (Buxco, Troy, NY), which is specifically designed for low tidal volumes (25, 30). Briefly, the mouse to be tested was anesthetized with sodium pentobarbital and intubated via cannulation of the trachea with an 18-gauge metal tube. The mouse was subsequently ventilated with a Harvard pump ventilator (VT = 0.4 ml, frequency = 120 breaths/minute, positive end-expiratory pressure 2.5–3.0 cm H2O) and the tail vein was cannulated with a 27-gauge needle for injection of the methacholine challenge. After determining a dose response curve (0.001 to 0.5 mg/kg), an optimal dose was chosen, 0.1 mg/kg of methacholine, which induced minimal AHR in naive control mice but a very significant induction of AHR in allergic animals. After the methacholine challenge, the response was monitored and the peak airway resistance recorded as a measure of AHR.

    ELISAs

    Cytokines were quantified from homogenized (phosphate-buffered saline [PBS] with 0.05% Triton X-100 nonionic detergent and antiproteases) lung aqueous extracts using a double-ligand ELISA system. The murine ELISAs were set up using standardized antibodies purchased from R&D Systems (Rochester, MN) that detect protein at concentrations above 10 pg/ml, are specific, and do not cross react with any other cytokines.

    Statistics

    Statistical significance was determined using analysis of variance with p values less than 0.05.

    RESULTS

    Treatment of Mice with Imatinib Attenuates Cockroach Allergen-induced Pulmonary Disease

    Previous studies have demonstrated a role for SCF/c-kit activation pathway in the development of allergen-induced AHR and inflammation (23–25, 31, 32). To further examine the potential for targeting this activation pathway, we treated mice with an inhibitor for c-kit–associated tyrosine kinase, imatinib. Allergen-sensitized mice were treated by oral gavages with 0.2 ml of PBS containing various doses of clinical-grade imatinib covering a 4 log10 dose of inhibitor. Thirty minutes after the oral gavage of PBS or PBS + imatinib, allergic mice were challenge by intratracheal injection with cockroach allergen (6 μg) suspended in 40 μl of PBS. As a control, naive mice were also challenged with cockroach allergen. After 24 hours, the mice were examined for induction of AHR after a methacholine challenge (see METHODS). The results in Figure 1 depict that treatment of mice with imatinib significantly reduced the induction of AHR at higher doses, 50 and 5 mg/kg, but not at the lower doses, 0.5 and 0.05 mg/kg. The level of airway resistance that was measured by direct ventilation plethysmography in the imatinib-treated group was reduced to nearly the levels observed in the naive control-challenged group.

    A significant correlation can often be drawn between the intensity of the pulmonary inflammation and AHR responses in animal models of asthma. Figure 2 depicts that those animals treated with imatinib demonstrated significant reduction in the overall inflammatory response to the cockroach allergen challenge compared with vehicle-treated allergic animals. This was further examined by enumerating the number of eosinophils that accumulated around the airways of the allergen-challenged animals (Figure 3). The imatinib nearly abrogated the presence of eosinophils around the airways and directly correlated with the reduction in airway hyperresponsiveness observed above in the higher doses of imatinib. In contrast, in addition we examined the expression of MUC5AC mucin by quantitative polymerase chain reaction and airway goblet cells by periodic acid-Schiff/alcian blue stain, and neither was altered by the treatment of a single dose of imatinib before allergen challenge (data not shown). Thus, there appeared to be an alteration in the inflammation and airway physiology but no decrease in mucus.

    Alteration of Pulmonary Cytokine and Chemokine Levels with Imatinib

    To further characterize the nature of the response with imatinib treatment of mice, separate studies were designed to examine an oral dose response (0.5 to 50 mg/kg). Whole-lung homogenates were analyzed for cytokine and chemokine levels using specific ELISAs (Figure 4). The data indicate that both IL-4 and IL-13 levels were significantly reduced by treating the mice with all doses of imatinib (Figure 4A). Even at a dose of imatinib that did not alter AHR, 0.5 mg/kg, there was a significant reduction in both IL-4 and IL-13. When a number of allergen response related chemokines were examined, we also found a significant reduction in CCL2, CCL5, and CCL6, especially at the higher doses (Figure 4B). Because we found a significant reduction in several of the cytokines/chemokines measured even at the lower dose used, we performed a separate experiment using an even lower dose of 0.05 mg/kg and found that these mice had no significant reduction in cytokine or chemokine production (data not shown), reflecting the AHR and eosinophil responses observed above. These mediators have all been independently implicated in the progression of allergic responses in several different models of airway diseases and relate to the overall inflammation within the lungs. Thus, the overall reduction of inflammation may be a result of inhibition of a number of interrelated responses.

    DISCUSSION

    The identification of effective therapy for patients with moderate-to-severe asthma has been relegated in recent years to developing more efficient delivery of steroids to the airway (33, 34). These nonspecific compounds decrease the production and release of a wide array of immune/inflammatory mediators and significantly limit the effect of the overall immune response. However, the ability to specifically block certain critical activation pathways utilizing signaling blockades may prove to be beneficial to alleviate long-term chronic responses. In the present studies, on the basis of previously published data from our laboratory and others (23–25, 27, 32), we initiated an analysis of whether we could alleviate responses in our preclinical model of cockroach antigen-induced asthma by blocking receptor tyrosine kinases related to c-kit. Imatinib has been shown to inhibit the c-kit and PDGFR activation pathways as well as arginine kinase pathways, but not other receptor tyrosine kinase pathways examined (18, 35). The data from the present study have been striking, as not only was the development of AHR significantly reduced, but also the inflammatory response was nearly abrogated. In particular, the Th2 cytokines that dominate the allergic airway responses were reduced in the lung postchallenge. Although it is not clear which specific tyrosine kinase pathways were altered with imatinib, these studies demonstrate that this approach and, more importantly, this drug may provide a viable therapeutic option for blocking certain aspects of asthmatic responses. However, further studies also found that although AHR and inflammation were reduced after a single treatment of allergic mice with imatinib, neither serum IgE levels nor airway mucus expression were reduced, indicating that not all aspects of chronic asthma are alleviated. This latter issue will surely need to be addressed in additional studies focused on longer term treatment with imatinib during the development of allergic airway responses.

    A striking aspect of these studies is the reduction in eosinophils within and around the airway. Recent studies in patients with hypereosinophilic syndrome have established a role for imatinib in the reduction of eosinophil numbers and the associated pathophysiology of this often devastating disease (19–21, 36–42). Although it is not completely clear by what mechanism imatinib is operating during hypereosinophilic syndrome, it has had an extremely beneficial effect in a significant number of patients related to a mutation in PDRFR- (37, 38). The data from the present studies suggest that imatinib may have an overall effect on the immune/inflammatory response during antigen-specific reactions. Others and we have established that inhibition of SCF in the airways of allergic mice can significantly attenuate the inflammatory/immune responses (23–25, 27, 32). Previous data suggest that the role of SCF would be multifactorial by inhibiting not only the local mast cell populations involved early on during the response but also the recruitment and activation of eosinophils (22, 23, 43–45). Furthermore, the recent identification of increased expression of both SCF and c-kit in airways of individuals with asthma gives additional support for targeting this activation pathway (28). However, because imatinib may also have effects on other receptor tyrosine kinase family members, such as PDGFR, the effects observed in this study likely were an outcome of blocking other pathways as well. The responses are also likely to encompass the alteration of bone marrow-derived cells, especially if imatinib is given long term. We found no alteration of circulating leukocyte numbers (data not shown), but have not performed extensive studies to examine bone marrow or peripheral leukocyte counts in the present studies. However, in these studies imatinib was given a single time just before allergen challenge, and the effects may be centered on the alleviation of the inflammatory responses directly activated within the airways. The reduction of chemokines may have resulted from decreased Th2 cytokines as well as the direct effects of blocking specific signaling processes related to specific receptor tyrosine kinase pathways.

    Previous observations in patients treated with imatinib for chronic myeloid leukemia have identified increases in IFN after a 3-month treatment protocol, potentially suggesting an alteration of the overall balance of Th1 and Th2 type responses (46). In contrast, another study that examined T cell responses in patients with chronic myeloid leukemia before and after imatinib treatment found no difference in the Th1/Th2 cytokine levels on polyclonal activation (47). Interestingly, data suggest that imatinib may produce long-term, event-free survival in patients with T cell lymphoid blastic phase (48). Another recent publication has indicated that imatinib treatment affects the development of CD34+ progenitor cells into dendritic cells (49), further supporting a role for imatinib in altering the development of detrimental immune responses. Related to the current study are previous observations where SCF has been specifically blocked in the airway, either by antibody or antisense therapy, and an alteration in Th2 cytokines observed (31, 32). Thus, by blocking the initiation of this pathway a significant effect can be observed in the expression of a number of allergen-induced cytokines.

    These studies have identified a potential avenue of treatment that centers on blocking certain activation pathways that have previously not been considered for asthma therapy. These results, although striking, deserve additional investigation within preclinical models of allergic airway disease and possibly, subsequent investigation in populations of patients with asthma.

    FOOTNOTES

    Supported in part by National Institutes of Health grants HL58178 and AI36302.

    Conflict of Interest Statement: A.A.B. does not have a financial relationship with a commercial entity that has an interest in the subject of this manuscript; N.W.L. does not have a financial relationship with a commercial entity that has an interest in the subject of this manuscript.

    Received in original form March 22, 2004; accepted in final form September 15, 2004

    REFERENCES

    Busse WW, Banks-Schlegel S, Wenzel SE. Pathophysiology of severe asthma. J Allergy Clin Immunol 2000;106:1033–1042.

    Hartert TV, Peebles RS. Epidemiology of asthma: the year in review. Curr Opin Pulm Med 2000;6:4–9.

    Salvi SS, Babu KS, Holgate ST. Is asthma really due to a polarized T cell response toward a helper T cell type 2 phenotype? Am J Respir Crit Care Med 2001;164:1343–1346.

    Black JL, Johnson PR. What determines asthma phenotype: is it the interaction between allergy and the smooth muscle? Am J Respir Crit Care Med 2000;161:S207–S210.

    Barnes PJ. Endogenous inhibitory mechanisms in asthma. Am J Respir Crit Care Med 2000;161:S176–S181.

    Haley KJ, Drazen JM. Inflammation and airway function in asthma: what you see is not necessarily what you get. Am J Respir Crit Care Med 1998;157:1–3.

    Walsh GM. Eosinophil granule proteins and their role in disease. Curr Opin Hematol 2001;8:28–33.[CrossRef]

    Gleich GJ. Mechanisms of eosinophil-associated inflammation. J Allergy Clin Immunol 2000;105:651–663.

    Lukacs NW, Tekkanat KK. Role of chemokines in asthmatic airway inflammation. Immunol Rev 2000;177:21–30.

    Holtzman MJ, Morton JD, Shornick LP, Tyner JW, O'Sullivan MP, Antao A, Lo M, Castro M, Walter MJ. Immunity, inflammation, and remodeling in the airway epithelial barrier: epithelial-viral-allergic paradigm. Physiol Rev 2002;82:19–46.

    Kraneveld AD, Folkerts G, Van Oosterhout A J, Nijkamp FP. Airway hyperresponsiveness: first eosinophils and then neuropeptides. Int J Immunopharmacol 1997;19:517–527.

    Hogan SP, Foster PS. Cellular and molecular mechanisms involved in the regulation of eosinophil trafficking in vivo. Med Res Rev 1996;16:407–432

    Garlisi CG, Falcone A, Hey JA, Paster TM, Fernandez X, Rizzo CA, Minnicozzi M, Jones H, Billah MM, Egan RW, et al. Airway eosinophils, T cells, Th2-type cytokine mRNA, and hyperreactivity in response to aerosol challenge of allergic mice with previously established pulmonary inflammation. Am J Respir Cell Mol Biol 1997;17:642–651.

    Hogan SP, Koskinen A, Foster PS. Interleukin-5 and eosinophils induce airway damage and bronchial hyperreactivity during allergic airway inflammation in BALB/c mice. Immunol Cell Biol 1997;75:284–288.

    Hughes T, Branford S. Molecular monitoring of chronic myeloid leukemia. Semin Hematol 2003;40:62–68.

    O'Dwyer ME, Mauro MJ, Druker BJ. STI571 as a targeted therapy for CML. Cancer Invest 2003;21:429–438.

    Capdeville R, Silberman S, Dimitrijevic S. Imatinib: the first 3 years. Eur J Cancer 2002;38:S77–S82.

    Manley PW, Cowan-Jacob SW, Buchdunger E, Fabbro D, Fendrich G, Furet P, Meyer T, Zimmermann J. Imatinib: a selective tyrosine kinase inhibitor. Eur J Cancer 2002;38:S19–S27.

    Pardanani A, Reeder T, Porrata LF, Li CY, Tazelaar HD, Baxter EJ, Witzig TE, Cross NC, Tefferi A. Imatinib therapy for hypereosinophilic syndrome and other eosinophilic disorders. Blood 2003;101:3391–3397.

    Coutre S, Gotlib J. Targeted treatment of hypereosinophilic syndromes and chronic eosinophilic leukemias with imatinib mesylate. Semin Cancer Biol 2004;14:23–31.

    Griffin JH, Leung J, Bruner RJ, Caligiuri MA, Briesewitz R. Discovery of a fusion kinase in EOL-1 cells and idiopathic hypereosinophilic syndrome. Proc Natl Acad Sci USA 2003;100:7830–7835.

    Undem BJ, Lichtenstein LM, Hubbard WC, Meeker S, Ellis JL. Recombinant stem cell factor-induced mast cell activation and smooth muscle contraction in human bronchi. Am J Respir Cell Mol Biol 1994;11:646–650.

    Lukacs NW, Strieter RM, Lincoln PM, Brownell E, Pullen DM, Schock HJ, Chensue SW, Taub DD, Kunkel SL. Stem cell factor (c-kit ligand) influences eosinophil recruitment and histamine levels in allergic airway inflammation. J Immunol 1996;156:3945–3951.

    Campbell E, Hogaboam C, Lincoln P, Lukacs NW. Stem cell factor-induced airway hyperreactivity in allergic and normal mice. Am J Pathol 1999;154:1259–1265.

    Oliveira SH, Hogaboam CM, Berlin A, Lukacs NW. SCF-induced airway hyperreactivity is dependent on leukotriene production. Am J Physiol Lung Cell Mol Physiol 2001;280:L1242–L1249.

    Yuan Q, Austen KF, Friend DS, Heidtman M, Boyce JA. Human peripheral blood eosinophils express a functional c-kit receptor for stem cell factor that stimulates very late antigen 4 (VLA-4)-mediated cell adhesion to fibronectin and vascular cell adhesion molecule 1 (VCAM-1). J Exp Med 1997;186:313–323.

    Oliveira SH, Lukacs NW. Stem cell factor: a hemopoietic cytokine with important targets in asthma. Curr Drug Targets Inflamm Allergy 2003;2:313–318.

    Al-Muhsen SZ, Shablovsky G, Olivenstein R, Mazer B, Hamid Q. The expression of stem cell factor and c-kit receptor in human asthmatic airways. Clin Exp Allergy 2004;34:911–916

    Campbell EM, Charo IF, Kunkel SL, Strieter RM, Boring L, Gosling J, Lukacs NW. Monocyte chemoattractant protein-1 mediates cockroach allergen-induced bronchial hyperreactivity in normal but not CCR2–/– mice: the role of mast cells. J Immunol 1999;163:2160–2167.

    Campbell EM, Kunkel SL, Strieter RM, Lukacs NW. Temporal role of chemokines in a murine model of cockroach allergen-induced airway hyperreactivity and eosinophilia. J Immunol 1998;161:7047–7053.

    Berlin AA, Lincoln P, Tomkinson A, Lukacs NW. Inhibition of stem cell factor reduces pulmonary cytokine levels during allergic airway responses. Clin Exp Immunol 2004;136:15–20.

    Finotto S, Buerke M, Lingnau K, Schmitt E, Galle PR, Neurath MF. Local administration of antisense phosphorothioate oligonucleotides to the c-kit ligand, stem cell factor, suppresses airway inflammation and IL-4 production in a murine model of asthma. J Allergy Clin Immunol 2001;107:279–286.

    Keating GM,Faulds D. Airmax: a multi-dose dry powder inhaler. Drugs 2002;62:1887–1895; discussion 1896–1897.

    Janssens HM, De Jongste JC, Hop WC, Tiddens HA. Extra-fine particles improve lung delivery of inhaled steroids in infants: a study in an upper airway model. Chest 2003;123:2083–2088.

    Traxler P, Bold G, Buchdunger E, Caravatti G, Furet P, Manley P, O'Reilly T, Wood J, Zimmermann J. Tyrosine kinase inhibitors: from rational design to clinical trials. Med Res Rev 2001;21:499–512.

    Salem Z, Zalloua PA, Chehal A, Bitar N, Abboud M, Kadri A, Chami B, Bazarbachi A. Effective treatment of hypereosinophilic syndrome with imatinib mesylate. Hematol J 2003;4:410–412.

    Pardanani A, Ketterling RP, Brockman SR, Flynn HC, Paternoster SF, Shearer BM, Reeder TL, Li CY, Cross NC, Cools J, et al. CHIC2 deletion, a surrogate for FIP1L1-PDGFRA fusion, occurs in systemic mastocytosis associated with eosinophilia and predicts response to imatinib mesylate therapy. Blood 2003;102:3093–3096.

    Cools J, Stover EH, Boulton CL, Gotlib J, Legare RD, Amaral SM, Curley DP, Duclos N, Rowan R, Kutok JL, et al. PKC412 overcomes resistance to imatinib in a murine model of FIP1L1-PDGFRalpha-induced myeloproliferative disease. Cancer Cell 2003;3:459–469.

    Klion AD, Noel P, Akin C, Law MA, Gilliland DG, Cools J, Metcalfe DD, Nutman TB. Elevated serum tryptase levels identify a subset of patients with a myeloproliferative variant of idiopathic hypereosinophilic syndrome associated with tissue fibrosis, poor prognosis, and imatinib responsiveness. Blood 2003;101:4660–4666.

    Cortes J, Ault P, Koller C, Thomas D, Ferrajoli A, Wierda W, Rios MB, Letvak L, Kaled ES, Kantarjian H. Efficacy of imatinib mesylate in the treatment of idiopathic hypereosinophilic syndrome. Blood 2003;101:4714–4716.

    Ault P, Cortes J, Koller C, Kaled ES, Kantarjian H. Response of idiopathic hypereosinophilic syndrome to treatment with imatinib mesylate. Leuk Res 2002;26:881–884.

    Schaller JL, Burkland GA. Case report: rapid and complete control of idiopathic hypereosinophilia with imatinib mesylate. MedGenMed 2001;3:9.

    Oliveira SH, Taub DD, Nagel J, Smith R, Hogaboam CM, Berlin A, Lukacs NW. Stem cell factor induces eosinophil activation and degranulation: mediator release and gene array analysis. Blood 2002;100:4291–4297

    Oliveira SH, Lukacs NW. Stem cell factor and igE-stimulated murine mast cells produce chemokines (CCL2, CCL17, CCL22) and express chemokine receptors. Inflamm Res 2001;50:168–174.

    Wershil BK, Tsai M, Geissler EN, Zsebo KM, Galli SJ. The rat c-kit ligand, stem cell factor, induces c-kit receptor- dependent mouse mast cell activation in vivo: evidence that signaling through the c-kit receptor can induce expression of cellular function. J Exp Med 1992;175:245–255.

    Aswald JM, Lipton JH, Aswald S, Messner HA. Increased IFN-gamma synthesis by T cells from patients on imatinib therapy for chronic myeloid leukemia. Cytokines Cell Mol Ther 2002;7:143–149.

    Kiani A, Habermann I, Schake K, Neubauer A, Rogge L, Ehninger G. Normal intrinsic Th1/Th2 balance in patients with chronic phase chronic myeloid leukemia not treated with interferon-alpha or imatinib. Haematologica 2003;88:754–761.

    Atallah E, Talpaz M, O'Brien S, Rios MB, Guo JQ, Arlinghaus R, Fernandes-Reese S, Kantarjian H. Chronic myelogenous leukemia in T cell lymphoid blastic phase achieving durable complete cytogenetic and molecular remission with imatinib mesylate (STI571; Gleevec) therapy. Cancer 2002;94:2996–2999.

    Appel S, Boehmler AM, Grunebach F, Muller MR, Rupf A, Weck MM, Hartmann U, Reichardt VL, Kanz L, Brummendorf TH, et al. Imatinib mesylate affects the development and function of dendritic cells generated from CD34+ peripheral blood progenitor cells. Blood 2004;103:538–544.(Aaron A. Berlin and Nicho)