CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma

Blood ◽  
2005 ◽  
Vol 105 (7) ◽  
pp. 2941-2948 ◽  
Author(s):  
Suzanne Trudel ◽  
Zhi Hua Li ◽  
Ellen Wei ◽  
Marion Wiesmann ◽  
Hong Chang ◽  
...  

Abstract The t(4;14) translocation that occurs uniquely in a subset (15%) of patients with multiple myeloma (MM) results in the ectopic expression of the receptor tyrosine kinase (RTK), fibroblast growth factor receptor 3 (FGFR3). Inhibition of activated FGFR3 in MM cells induces apoptosis, validating FGFR3 as a therapeutic target in t(4;14) MM and encouraging the clinical development of FGFR3 inhibitors for the treatment of these patients, who have a poor prognosis. We describe here the characterization of a novel, small-molecule inhibitor of class III, IV, and V RTKs, CHIR-258, as an inhibitor of FGFR3. CHIR-258 potently inhibits FGFR3 with an inhibitory concentration of 50% (IC50) of 5 nM in in vitro kinase assays and selectively inhibited the growth of B9 cells and human myeloma cell lines expressing wild-type (WT) or activated mutant FGFR3. In responsive cell lines, CHIR-258 induced cytostatic and cytotoxic effects. Importantly, addition of interleukin 6 (IL-6) or insulin growth factor 1 (IGF-1) or coculture on stroma did not confer resistance to CHIR-258. In primary myeloma cells from t(4;14) patients, CHIR-258 inhibited downstream extracellular signal-regulated kinase (ERK) 1/2 phosphorylation with an associated cytotoxic response. Finally, therapeutic efficacy of CHIR-258 was demonstrated in a xenograft mouse model of FGFR3 MM. These studies support the clinical evaluation of CHIR-258 in MM.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 641-641 ◽  
Author(s):  
Suzanne Trudel ◽  
Zhi Hua Li ◽  
Ellen Wei ◽  
Marion Wiesmann ◽  
Katherine Rendahl ◽  
...  

Abstract The t(4;14) translocation that occurs uniquely in a subset (15%) of multiple myeloma (MM) patients results in the ectopic expression of the receptor tyrosine kinase, Fibroblast Growth Factor Receptor3 (FGFR3). Wild-type FGFR3 induces proliferative signals in myeloma cells and appears to be weakly transforming in a hematopoeitic mouse model. The subsequent acquisition of FGFR3 activating mutations in some MM is associated with disease progression and is strongly transforming in several experimental models. The clinical impact of t(4;14) translocations has been demonstrated in several retrospective studies each reporting a marked reduction in overall survival. We have previously shown that inhibition of activated FGFR3 causes morphologic differentiation followed by apoptosis of FGFR3 expressing MM cell lines, validating activated FGFR3 as a therapeutic target in t(4;14) MM and encouraging the clinical development of FGFR3 inhibitors for the treatment of these poor-prognosis patients. CHIR258 is a small molecule kinase inhibitor that targets Class III–V RTKs and inhibits FGFR3 with an IC50 of 5 nM in an in vitro kinase assay. Potent anti-tumor and anti-angiogenic activity has been demonstrated in vitro and in vivo. We employed the IL-6 dependent cell line, B9 that has been engineered to express wild-type FGFR3 or active mutants of FGFR3 (Y373C, K650E, G384D and 807C), to screen CHIR258 for activity against FGFR3. CHIR258 differentially inhibited FGF-mediated growth of B9 expressing wild-type and mutant receptors found in MM, with an IC50 of 25 nM and 80 nM respectively as determined by MTT proliferation assay. Growth of these cells could be rescued by IL-6 demonstrating selectivity of CHIR258 for FGFR3. We then confirmed the activity of CHIR258 against FGFR3 expressing myeloma cells. CHIR258 inhibited the viability of FGFR3 expressing KMS11 (Y373C), KMS18 (G384D) and OPM-2 (K650E) cell lines with an IC50 of 100 nM, 250 nM and 80 nM, respectively. Importantly, inhibition with CHIR258 was still observed in the presence of IL-6, a potent growth factors for MM cells. U266 cells, which lack FGFR3 expression, displayed minimal growth inhibition demonstrating that at effective concentrations, CHIR258 exhibits minimal nonspecific cytotoxicity on MM cells. Further characterization of this finding demonstrated that inhibition of cell growth corresponded to G0/G1 cell cycle arrest and dose-dependent inhibition of downstream ERK phosphorylation. In responsive cell lines, CHIR258 induced apoptosis via caspase 3. In vitro combination analysis of CHIR258 and dexamethasone applied simultaneously to KMS11 cells indicated a synergistic interaction. In vivo studies demonstrated that CHIR258 induced tumor regression and inhibited growth of FGFR3 tumors in a plasmacytoma xenograft mouse model. Finally, CHIR258 produced cytotoxic responses in 4/5 primary myeloma samples derived from patients harboring a t(4;14) translocation. These data indicate that the small molecule inhibitor, CHIR258 potently inhibits FGFR3 and has activity against human MM cells setting the stage for a Phase I clinical trial of this compound in t(4;14) myeloma.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-14
Author(s):  
Ida Steiro ◽  
Pegah Abdollahi ◽  
Magne Børset ◽  
Tobias S. Slørdahl

Both in newly diagnosed multiple myeloma (MM) and during progression of the disease, malignant plasma cells are found circulating in peripheral blood as well as in the bone marrow (BM). The disseminated nature of MM is strongly dependent on the interplay between the cancer cells and the BM microenvironment, promoting myeloma cell migration in the BM. Matriptase (ST14), a type-II transmembrane serine protease primarily found in epithelial tissues, is overexpressed in a variety of human malignancies and is sufficient to induce tumour formation in mice. Frequently, a concomitant reduction in the levels of its cognate inhibitor hepatocyte growth factor activator inhibitor (HAI)-1 (SPINT1) is observed in carcinomas, while expression and function of the related inhibitor HAI-2 (SPINT2) is yet to be clarified. Dysregulated expression causing increased matriptase proteolytic activity has been associated with cancer growth, survival and metastasis. Here, we show for the first time a role of matriptase as a possible tumour suppressor in myeloma pathogenesis. Gene expression analysis of primary cells from MM patients (n=24) and human myeloma cell lines (n=8) revealed highly variable levels of matriptase, HAI-1 and HAI-2. This observation prompted us to investigate the functional role of matriptase in vitro. We showed that stable overexpression of matriptase in INA-6, a MM cell line with no endogenous ST14 expression, reduced migration by more than 50% in response to the combination of the pro-migratory cytokines stromal cell-derived factor-1 alpha (SDF-1α) and hepatocyte growth factor (HGF, Fig. 1A). Conversely, stable knockdown of matriptase in two MM cell lines with high endogenous matriptase expression (RPMI-8226 and JJN-3) significantly enhanced migration in vitro. Mechanistically, matriptase overexpression blocked activation of Src kinase (Fig. 1B), well-known as a critical player in metastasis formation promoting cancer cell motility, invasiveness and angiogenesis. In agreement with our result, previous studies have demonstrated the activation of Src family kinases (SFK) downstream SDF-1/CXCR4-signaling. Finally, we performed survival analyses in the public available MMRF CoMMpass trial database (release version IA14). Low ST14 expression was associated with significant worse overall survival (P=0.05, Fig. 1C) and progression-free survival (P=0.02, Fig. 1D). Altogether, our data are in marked contrast to the role ascribed to matriptase in epithelial and certain non-epithelial tumours as an oncogenic protein and an unfavourable prognostic marker. In conclusion, these findings suggest a novel role of matriptase as a tumour suppressor in MM pathogenesis. Disclosures Slørdahl: Celgene: Consultancy; Janssen and Celgene: Honoraria.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 247-247 ◽  
Author(s):  
Joseph Negri ◽  
Nicholas Mitsiades ◽  
Qingwei Deng ◽  
Zhaoqin Wen ◽  
David C. Geer ◽  
...  

Abstract Multiple myeloma (MM) remains an incurable neoplasia and exhibits high propensity for de novo/acquired refractoriness even to novel agents, e.g. thalidomide (Thal) or proteasome inhibitors. This may be due to complex and evolving patterns of molecular lesions potentially conferring hyperactive antiapoptotic signaling with high degree of redundancy upon inhibition of isolated targets within those pathways. We thus hypothesized that, for genetically complex neoplasias such as MM, patient outcome might improved by addition, in the therapeutic armamentarium, of agents that simultaneously inhibit multiple proliferative/antiapoptotic targets. Towards this effort of multi-targeted therapies, we studied the tyrosine kinase inhibitor PKC412 (midausporin, Novartis, Basel, Switzerland). Low-nM levels of PKC412 selectively inhibit tyrosine kinase receptors, such as FLT3. But higher PKC412 concentrations (in μM range), which presumably inhibit (at least partly) other kinases, are achieved in clinical trials without catastrophic toxicities. This suggests that PKC412 can safely suppress in vivo the activity of, not just FLT3, but a broader spectrum of kinases, some of which (individually or cooperatively) might be critical for survival/proliferation of MM cells. Indeed, in vitro kinase activity assays showed that clinically achievable (high nM or low μM) PKC412 concentrations inhibit by >20% important kinases, including p70S6K, IKK-a and Akt,. Consistent with such multi-targeted effect, in vitro MTT colorimetric survival assays showed activity of PKC412 (at sub-μM levels) against panel of 40 MM cell lines and 10 primary tumor cells from MM patients (including cells resistant to Dex, alkylating agents, anthracyclines, Thal or its immunomodulatory derivatives, bortezomib, and/or Apo2L/TRAIL), and cell lines from hematologic neoplasias, e.g. B- and T-ALL, CML, various non-Hodgkin’s lymphoma subtypes, and solid tumors (e.g. breast, prostate, lung, thyroid, ovarian, renal Ca, retinoblastoma and sarcomas)). Mechanistic studies confirmed that PKC412 blocks key growth/survival pathways (e.g. PI-3K/Akt, IKK-α/NF-κB), coupled with by downstream effects on suppression of diverse inhibitors of apoptosis (e.g. FLIP, XIAP, cIAP-2, survivin). These molecular sequelae explain, at least partly, the ability of PKC412 to sensitize MM cells to other anti-MM agents (such as Dex, cytotoxic chemotherapy or proteasome inhibitor bortezomib) and overcome protective effects of cytokines (e.g. IL-6) or bone marrow stromal cells. Importantly, PKC412 significantly prolonged the overall survival (p<0.03, Kaplan-Meier analysis) of SCID/NOD mice in a clinically relevant model of diffuse MM bones lesions. These studies provide basis for clinical studies of PKC412 in MM and indicate that kinase inhibitors selectively blocking specific targets at low drug levels, may also have potent anti-tumor activities related to inhibition of multiple other, less specific, nonetheless important targets, thus allowing for anti-tumor activity in a much broader spectrum of tumor types than previously appreciated.


2004 ◽  
Vol 18 (6) ◽  
pp. 1471-1485 ◽  
Author(s):  
Yao Huang ◽  
Sung-Oh Kim ◽  
Ning Yang ◽  
Jing Jiang ◽  
Stuart J. Frank

Abstract GH and IGF-I are critical regulators of growth and metabolism. GH interacts with the GH receptor (GHR), a cytokine superfamily receptor, to activate the cytoplasmic tyrosine kinase, Janus kinase 2 (JAK2), and initiate intracellular signaling cascades. IGF-I, produced in part in response to GH, binds to the heterotetrameric IGF-I receptor (IGF-IR), which is an intrinsic tyrosine kinase growth factor receptor that triggers proliferation, antiapoptosis, and other biological actions. Previous in vitro and overexpression studies have suggested that JAKs may interact with IGF-IR and that IGF-I stimulation may activate JAKs. In this study, we explore interactions between GHR-JAK2 and IGF-IR signaling pathway elements utilizing the GH and IGF-I-responsive 3T3-F442A and 3T3-L1 preadipocyte cell lines, which endogenously express both the GHR and IGF-IR. We find that GH induces formation of a complex that includes GHR, JAK2, and IGF-IR in these preadipocytes. The assembly of this complex in intact cells is rapid, GH concentration dependent, and can be prevented by a GH antagonist, G120K. However, it is not inhibited by the kinase inhibitor, staurosporine, which markedly inhibits GHR tyrosine phosphorylation. Moreover, complex formation does not appear dependent on GH-induced activation of the ERK or phosphatidylinositol 3-kinase signaling pathways or on the tyrosine phosphorylation of GHR, JAK2, or IGF-IR. These results suggest that GH-induced formation of the GHR-JAK2-IGF-IR complex is governed instead by GH-dependent conformational change(s) in the GHR and/or JAK2. We further demonstrate that GH and IGF-I can synergize in acute aspects of signaling and that IGF-I enhances GH-induced assembly of conformationally active GHRs. These findings suggest the existence of previously unappreciated relationships between these two hormones.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3438-3438
Author(s):  
Håkon Hov ◽  
Unn-Merethe Fagerli ◽  
Erming Tian ◽  
Anders Waage ◽  
Magne Borset ◽  
...  

Abstract HGF is known to be a prognostic factor in multiple myeloma, however the proliferative effect of HGF alone has been moderate. We investigated the effects of the myeloma growth factors IL-6, IGF-1 and IL-21 in combination with HGF in the myeloma cell lines IH-1, INA-6 and OH-2. In proliferation experiments the cells were grown in serum free media with 3 different HGF-concentrations together with various concentrations of IL-6, IGF-1 or IL-21. HGF alone had low effect. However, 200 ng/ml HGF in combination with IL-6 doubled the effect of IL-6 and tripled the effect of IGF-1 in INA-6. Similar results were obtained in OH-2 combining IL-6, IGF-1 and IL-21 with HGF and in IH-1 combining IL-6 with HGF. A combination of IL-6 and IGF-1 in INA-6 gave no synergistic effects, so the synergism seemed to be a specific effect for HGF in combination with the other cytokines.The myeloma cell line ANBL-6 harbours an autocrine growth promoting HGF-loop. We inhibited this autocrine HGF loop with a specific c-Met receptor tyrosine kinase inhibitor PHA-665752 in the presence of IL-6 and IGF-1. IL-6 and IGF-1 potentiated the decrease in proliferation caused by PHA-665752. These results demonstrate synergistic effects between the three cytokines (IL6, IGF-1, IL-21) and HGF. We also combined PHA-665752 and IL-6 in INA-6 cells, and 200 nM of the c-Met inhibitor halved the effect of IL-6. We have not seen any unspecific effects of PHA-665752 at 200 nM. This result raises the very interesting possibility that some of the effects of IL-6 can be due to c-Met signalling. At the protein level the cell lines responding to IL-6, IGF-1 or IL-21 also increased the level of c-Met expression in response to the individual cytokines. In OH-2 there was also an increase in the HGF transcript after IL-6, IGF-1 and IL-21 stimulation, as investigated by RT-PCR, indicating another mechanism for synergy between the HGF-c-Met pathway and the other cytokines in this cell line. In conclusion our results demonstrates the induction of c-Met by IL-6, IGF-1 and IL-21, and proliferative synergy between these cytokines and HGF in myeloma cell lines. Inhibition of c-Met with a specific tyrosine kinase inhibitor decreased the proliferative effect of IL-6. Together these results indicate that inhibition of c-Met signalling would target several pathways and could therefore be a promising treatment strategy in multiple myeloma.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 413-413 ◽  
Author(s):  
Bertrand Arnulf ◽  
David Ghez ◽  
Veronique Leblond ◽  
Sylvain Choquet ◽  
Karim Belhadj ◽  
...  

Abstract The t(4;14)(p16;q32), found in 15% of multiple myeloma (MM) cases, is associated with a short time to progression (TTP) despite a good initial response. In this subset of MM, relapses are resistant to conventional and intensive chemotherapy leading to a poor prognosis. Plasma cells with t(4;14) ectopically express the fibroblast growth factor receptor 3 (FGFR3), a tyrosine kinase receptor which has proven transforming activity and may represent a therapeutic target. We have studied the safety and efficacy of AB1010, an FGFR3 tyrosine kinase inhibitor, in patients with relapsing/refractory t(4;14) MM. 24 MM patients (M 33%, F 67%, median age 55 years) with t(4;14) were enrolled. FGFR3 expression was detected in all but one patient. AB1010 (9 mk/kg/d) was given orally and Dexamethasone (Dex) (40 mg/d X4d/month) was added if progression. In case of explosive relapse (defined by deep cytopenia, renal failure, circulating plasma cells), a chemotherapy was given followed by a wash out period of 1 month before start of AB1010. Two patients were enrolled after the first line of treatment at a plateau state with a response < 75% (<75% decrease of the monoclonal component). Among the 24 patients, 19 were evaluable since 5 had a duration of treatment < 1month. The main toxicities were gastrointestinal (nausea 63,5%, grade I/II, diarrhoea 25%, grade I; anorexia 25%, grade II) and oedema (face 50%, grade I/II and legs 24% grade I). Hematological toxicities were limited to a transient grade III neutropenia in one patient. Dex was added in all cases. Six patients (3 in 1st relapse, 3 in >3rd lines) had explosive relapse. Treatment regimen before start of AB1010 were :VTD (4); Thal/Dex (1); MLPHD (1). In these 6 cases, the TTP range from 1,5 to 4 months. 11 patients had non explosive relapse. Of these, 5 were in 1st relapse in whom 1 near CR (figure) and 1 PR were observed, with a median TTP of 10 months. Six patiens were in > 2nd relapse and 2 MR were obtained. Of the 2 remaining patients enrolled at plateau, 1 progressed at 6 and 1 is still on going after 13 months. In conclusion, AB1010, in combination with Dex, is well tolerated and may be useful in the treatment of patients with t(4;14) MM, especially in early phase of the disease. Responses were observed (40%) in patients with first relapse with a TTP superior as compared to the reported one (4,7 months with Thalidomide/Dex, Jaksic et al, 2006). Given the synergistic effect on MM cells proliferation observed in vitro, a study is now on going to test the safety and the efficacy of the combination AB1010 + Dex + Velcade in early relapsing patients with t(4;14) MM. Figure Figure


Blood ◽  
2012 ◽  
Vol 120 (16) ◽  
pp. 3260-3270 ◽  
Author(s):  
Deborah J. Kuhn ◽  
Zuzana Berkova ◽  
Richard J. Jones ◽  
Richard Woessner ◽  
Chad C. Bjorklund ◽  
...  

Abstract Proteasome inhibition with bortezomib is a validated approach to the treatment of multiple myeloma, but drug resistance often emerges and limits its utility in the retreatment setting. To begin to identify some of the mechanisms involved, we developed bortezomib-resistant myeloma cell lines that, unlike previously reported models, showed no β5 subunit mutations. Instead, up-regulation of the insulin-like growth factor (IGF)–1 axis was identified, with increased autocrine and paracrine secretion of IGF-1, leading to increased activation of the IGF-1 receptor (IGF-1R). Exogenous IGF-1 reduced cellular sensitivity to bortezomib, whereas pharmacologic or small hairpin RNA–mediated IGF-1R suppression enhanced bortezomib sensitivity in cell lines and patient samples. In vitro studies with OSI-906, a clinically relevant dual IGF-1R and insulin receptor inhibitor, showed it acted synergistically with bortezomib, and potently resensitized bortezomib-resistant cell lines and patient samples to bor-tezomib. Importantly, OSI-906 in combination with bortezomib also overcame bor-tezomib resistance in an in vivo model of myeloma. Taken together, these data support the hypothesis that signaling through the IGF-1/IGF-1R axis contributes to acquired bortezomib resistance, and provide a rationale for combining bortezomib with IGF-1R inhibitors like OSI-906 to overcome or possibly prevent the emergence of bortezomib-refractory disease in the clinic.


Sign in / Sign up

Export Citation Format

Share Document