Differential inhibitor sensitivity of anaplastic lymphoma kinase variants found in neuroblastoma

Scott C Bresler, Andrew C Wood, Elizabeth A Haglund, Joshua Courtright, Lili T Belcastro, Jefferson S Plegaria, Kristina Cole, Yana Toporovskaya, Huaqing Zhao, Erica L Carpenter, James G Christensen, John M Maris, Mark A Lemmon, Yaël P Mossé, Scott C Bresler, Andrew C Wood, Elizabeth A Haglund, Joshua Courtright, Lili T Belcastro, Jefferson S Plegaria, Kristina Cole, Yana Toporovskaya, Huaqing Zhao, Erica L Carpenter, James G Christensen, John M Maris, Mark A Lemmon, Yaël P Mossé

Abstract

Activating mutations in the anaplastic lymphoma kinase (ALK) gene were recently discovered in neuroblastoma, a cancer of the developing autonomic nervous system that is the most commonly diagnosed malignancy in the first year of life. The most frequent ALK mutations in neuroblastoma cause amino acid substitutions (F1174L and R1275Q) in the intracellular tyrosine kinase domain of the intact ALK receptor. Identification of ALK as an oncogenic driver in neuroblastoma suggests that crizotinib (PF-02341066), a dual-specific inhibitor of the ALK and Met tyrosine kinases, will be useful in treating this malignancy. Here, we assessed the ability of crizotinib to inhibit proliferation of neuroblastoma cell lines and xenografts expressing mutated or wild-type ALK. Crizotinib inhibited proliferation of cell lines expressing either R1275Q-mutated ALK or amplified wild-type ALK. In contrast, cell lines harboring F1174L-mutated ALK were relatively resistant to crizotinib. Biochemical analyses revealed that this reduced susceptibility of F1174L-mutated ALK to crizotinib inhibition resulted from an increased adenosine triphosphate-binding affinity (as also seen in acquired resistance to epidermal growth factor receptor inhibitors). Thus, this effect should be surmountable with higher doses of crizotinib and/or with higher-affinity inhibitors.

Figures

Fig. 1. Constitutive activity and inhibitor sensitivity…
Fig. 1. Constitutive activity and inhibitor sensitivity of F1174L and R1275 ALK mutants
(A) Immunoblots of total ALK and pALK in hTERT-RPE1 cells infected with retroviruses directing expression of wild-type or mutated ALK. Lower panel is actin loading control. (B) Proliferation of neuroblastoma cell lines over 72 hours of incubation with 333nM crizotinib. Growth inhibition ± S.D. is reported for at least three independent experiments. p values were calculated for marked comparisons using two-sided exact Wilcoxon-Mann-Whitney tests. Cell lines were (left to right): wild-type ALK amplified (NB1); R1275Q (NB1643, LAN5); F1174L (SH-SY5Y, KELLY, NBSD, SMS-SAN); wild-type ALK, normal copy number (NB1691, NB-EBc1, IMR5, NB16, NLF, IMR32, NBLS, SKNBE2C, NGP, SKNAS, SKNFI).
Fig. 2. Crizotinib activity in vivo for…
Fig. 2. Crizotinib activity in vivo for wild-type and mutated ALK
Subcutaneously implanted neuroblastoma tumors were monitored in CB17 scid mice treated with crizotinib (solid red lines), or vehicle (dashed blue lines). Tumor volume (left panels) is displayed as mean ± S.E.M. Study end points for survival analysis (right panels) were tumor volume ≥1.5cm3 or treatment-related death. (A) NB1643 (R1275Q) xenografts: inset shows immunoblot of ALK and pALK; (B) SH-SY5Y (F1174L); (C) NBSD (F1174L); (D) NB1 (wild-type amplified with strong phospho-ALK staining); (E) NB-EBc1 (wild-type, with weak phospho-ALK staining); (F) SKNAS (wild-type, undetectable pALK). Statistical treatment is described in Materials and Methods.
Fig. 3. Inhibition of constitutive ALK autophosphorylation…
Fig. 3. Inhibition of constitutive ALK autophosphorylation by crizotinib
Representative immunoblots of ALK autophosphorylation in neuroblastoma cell-lines after treatment with different crizotinib concentrations (0 to 10µM). Whole cell lysates were immunoblotted for phospho-ALK (using pY1604 antibody), total ALK, and actin (loading control) for (A) NB1643 cells (R1275Q ALK), (B) SH-SY5Y cells (F1174L ALK). Downstream signaling molecules are analyzed in fig. S2. (C) Quantitation of phospho-ALK levels (220kDa species) as a function of crizotinib concentration.
Fig. 4. Analysis of ALK-TKD activation in…
Fig. 4. Analysis of ALK-TKD activation in vitro
(A) Separation of differently autophosphorylated ALK-TKD species by native gel electrophoresis to monitor autophosphorylation at 25°C in the absence (top) and presence (bottom) of vesicles containing 10% DOGS-NTA-Ni (100µM total lipid), 10mM MgCl2, and 2mM ATP. (B) Autophosphorylation of wild-type and mutated ALK-TKD (10µM) with saturating ATP (2mM) and 10mM MgCl2 at 37°C. Results are quantitated in fig. S4. (C) Rate of 32P incorporation at 25°C into substrate peptide (see Materials and Methods) for autophosphorylated ALK-TKD (10nM) and unphosphorylated ALK-TKD (500nM) as peptide substrate concentration is increased. ATP concentration was 2mM. (D) Km, ATP determination for autophosphorylated (10nM) and unphosphorylated (500nM) ALK-TKD at fixed peptide substrate concentration (1mM). (E) Enhancement of EGFR-TKD kinase (1µM) by vesicles containing increasing mole percentages of DOGS-NTA-Ni (10µM DOGS-NTA-Ni, 100–200µM total lipid). (F) Effect on autophosphorylated ALK-TKD (1µM) activity of adding DOGS-NTA-Ni vesicles. Data are shown as means ± SEM from at least three independent experiments. All experiments except those in (B) were performed at 25°C.
Fig. 5. Comparison of wild-type ALK-TKD with…
Fig. 5. Comparison of wild-type ALK-TKD with F1174L and R1275Q variants in vitro
(A,B) Rates of 32P incorporation into ‘YYY’ peptide at saturating ATP (2mM) for: (A) unphosphorylated wild-type (500nM) or mutated (50nM) ALK-TKD; and (B) phosphorylated wild-type and mutated ALK-TKD (all at 10nM). (C,D) Km, ATP determination (with YYY peptide fixed at 1mM) for: (C) unphosphorylated wild-type (500nM) or mutated (50nM) ALK-TKD; and (D) phosphorylated ALK-TKD variants (all at 10nM). (E) Comparison of catalytic efficiencies (kcat/Km, peptide) for unphosphorylated and phosphorylated ALK-TKD variants. (F) Comparison of Km, ATP values. (G,H) Inhibition of unphosphorylated F1174L and R1275Q ALK-TKD (50nM) by crizotinib in peptide phosphorylation assays ([peptide] is 0.5mM) at 0.2mM ATP (G) and 2mM ATP (H). All data are shown as means ± SEM from at least three independent experiments. Experiments were performed at 25°C.
Fig. 6. Structural basis for ALK-TKD activation…
Fig. 6. Structural basis for ALK-TKD activation by F1174L and R1275Q mutations
(A) Cartoon representations of inactive ALK-TKD (26) from PDB entry 3LCS (cyan), inactive EGFR-TKD (23) from PDB entry 2GS7 (grey), and active EGFR from PDB entry 1M14 (green). The activation loop is colored magenta in each structure. Positions of F1174 (red) and R1275 (blue) are marked in ALK-TKD, as are their structural equivalents in EGFR (V745 and L837). (B) Detail of interactions between the short activation loop helix (magenta) and helix αC in inactive ALK-TKD (left) and inactive EGFR-TKD (right). (C) Close-up of relationship between F1174 side chain, the ‘DFG motif’ and ATP/drug binding site. In the upper panel, taken from PDB entry 3LCT (26), which contains bound ADP, an Mg2+ ion (not reported in this structure) was placed based on its location in the active insulin receptor TKD. Lower panel (with bound crizotinib) taken from PDB entry 2XP2.

References

    1. D'Angio GJ, Evans AE, Koop CE. Special pattern of widespread neuroblastoma with a favourable prognosis. Lancet. 1971;1:1046–1049.
    1. Hobbie WL, Moshang T, Carlson CA, Goldmuntz E, Sacks N, Goldfarb SB, Grupp SA, Ginsberg JP. Late effects in survivors of tandem peripheral blood stem cell transplant for high-risk neuroblastoma. Pediatr. Blood Cancer. 2008;51:679–683.
    1. Iwahara T, Fujimoto J, Wen D, Cupples R, Bucay N, Arakawa T, Mori S, Ratzkin B, Yamamoto T. Molecular characterization of ALK, a receptor tyrosine kinase expressed specifically in the nervous system. Oncogene. 1997;14:439–449.
    1. Morris SW, Kirstein MN, Valentine MB, Dittmer KG, Shapiro DN, Saltman DL, Look AT. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. Science. 1994;263:1281–1284.
    1. Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, Bando M, Ohno S, Ishikawa Y, Aburatani H, Niki T, Sohara Y, Sugiyama Y, Mano H. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561–566.
    1. Palmer RH, Vernersson E, Grabbe C, Hallberg B. Anaplastic lymphoma kinase: signalling in development and disease. Biochem. J. 2009;420:345–361.
    1. Mossé YP, Laudenslager M, Longo L, Cole KA, Wood A, Attiyeh EF, Laquaglia MJ, Sennett R, Lynch JE, Perri P, Laureys G, Speleman F, Kim C, Hou C, Hakonarson H, Torkamani A, Schork NJ, Brodeur GM, Tonini GP, Rappaport E, Devoto M, Maris JM. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature. 2008;455:930–935.
    1. Carén H, Abel F, Kogner P, Martinsson T. High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumours. Biochem. J. 2008;416:153–159.
    1. Chen Y, Takita J, Choi YL, Kato M, Ohira M, Sanada M, Wang L, Soda M, Kikuchi A, Igarashi T, Nakagawara A, Hayashi Y, Mano H, Ogawa S. Oncogenic mutations of ALK kinase in neuroblastoma. Nature. 2008;455:971–974.
    1. George RE, Sanda T, Hanna M, Frohling S, Luther W, 2nd, Zhang J, Ahn Y, Zhou W, London WB, McGrady P, Xue L, Zozulya S, Gregor VE, Webb TR, Gray NS, Gilliland DG, Diller L, Greulich H, Morris SW, Meyerson M, Look AT. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature. 2008;455:975–978.
    1. Janoueix-Lerosey I, Lequin D, Brugieres L, Ribeiro A, de Pontual L, Combaret V, Raynal V, Puisieux A, Schleiermacher G, Pierron G, Valteau-Couanet D, Frebourg T, Michon J, Lyonnet S, Amiel J, Delattre O. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature. 2008;455:967–970.
    1. De Brouwer S, De Preter K, Kumps C, Zabrocki P, Porcu M, Westerhout EM, Lakeman A, Vandesompele J, Hoebeeck J, Van Maerken T, De Paepe A, Laureys G, Schulte JH, Schramm A, Van Den Broecke C, Vermeulen J, Van Roy N, Beiske K, Renard M, Noguera R, Delattre O, Janoueix-Lerosey I, Kogner P, Martinsson T, Nakagawara A, Ohira M, Caron H, Eggert A, Cools J, Versteeg R, Speleman F. Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clin. Cancer Res. 2010;16:4354–4363.
    1. Murugan AK, Xing M. Novel oncogenic mutations of the ALK gene in anaplastic thyroid cancer. Cancer Res. 2011;71:4403–4411.
    1. Christensen JG, Zou HY, Arango ME, Li Q, Lee JH, McDonnell SR, Yamazaki S, Alton GR, Mroczkowski B, Los G. Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-Met, in experimental models of anaplastic large-cell lymphoma. Mol. Cancer Ther. 2007;6:3314–3322.
    1. Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SH, Dezube BJ, Janne PA, Costa DB, Varella-Garcia M, Kim WH, Lynch TJ, Fidias P, Stubbs H, Engelman JA, Sequist LV, Tan W, Gandhi L, Mino-Kenudson M, Wei GC, Shreeve SM, Ratain MJ, Settleman J, Christensen JG, Haber DA, Wilner K, Salgia R, Shapiro GI, Clark JW, Iafrate AJ. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N. Engl. J. Med. 2010;363:1693–1703.
    1. Choi YL, Soda M, Yamashita Y, Ueno T, Takashima J, Nakajima T, Yatabe Y, Takeuchi K, Hamada T, Haruta H, Ishikawa Y, Kimura H, Mitsudomi T, Tanio Y, Mano H ALK Lung Cancer Study Group. EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N. Engl. J. Med. 2010;363:1734–1739.
    1. Sasaki T, Okuda K, Zheng W, Butrynski J, Capelletti M, Wang L, Gray NS, Wilner K, Christensen JG, Demetri G, Shapiro GI, Rodig SJ, Eck MJ, Janne PA. The neuroblastoma associated F1174L ALK mutation causes resistance to an ALK kinase inhibitor in ALK translocated cancers. Cancer Res. 2010;70:10038–10043.
    1. Katayama R, Khan TM, Benes C, Lifshits E, Ebi H, Rivera VM, Shakespeare WC, Iafrate AJ, Engelman JA, Shaw AT. Therapeutic strategies to overcome crizotinib resistance in non-small cell lung cancers harboring the fusion oncogene EML4-ALK. Proc. Natl. Acad. Sci. U.S.A. 2011;108:7535–7540.
    1. Mazot P, Cazes A, Boutterin MC, Figueiredo A, Raynal V, Combaret V, Hallberg B, Palmer RH, Delattre O, Janoueix-Lerosey I, Vigny M. The constitutive activity of the ALK mutated at positions F1174 or R1275 impairs receptor trafficking. Oncogene. 2011;30:2017–2025.
    1. Martinsson T, Eriksson T, Abrahamsson J, Caren H, Hansson M, Kogner P, Kamaraj S, Schönherr C, Weinmar J, Ruuth K, Palmer RH, Hallberg B. Appearance of the novel activating F1174S ALK mutation in neuroblastoma correlates with aggressive tumor progression and unresponsiveness to therapy. Cancer Res. 2011;71:98–105.
    1. Moog-Lutz C, Degoutin J, Gouzi JY, Frobert Y, Brunet-de Carvalho N, Bureau J, Créminon C, Vigny M. Activation and inhibition of anaplastic lymphoma kinase receptor tyrosine kinase by monoclonal antibodies and absence of agonist activity of pleiotrophin. J. Biol. Chem. 2005;280:26039–26048.
    1. Shrout AL, Montefusco DJ, Weis RM. Template-directed assembly of receptor signaling complexes. Biochemistry. 2003;42:13379–13385.
    1. Zhang X, Gureasko J, Shen K, Cole PA, Kuriyan J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell. 2006;125:1137–1149.
    1. Bossi RT, Saccardo MB, Ardini E, Menichincheri M, Rusconi L, Magnaghi P, Orsini P, Avanzi N, Borgia AL, Nesi M, Bandiera T, Fogliatto G, Bertrand JA. Crystal structures of anaplastic lymphoma kinase in complex with ATP competitive inhibitors. Biochemistry. 2010;49:6813–6825.
    1. Donella-Deana A, Marin O, Cesaro L, Gunby RH, Ferrarese A, Coluccia AM, Tartari CJ, Mologni L, Scapozza L, Gambacorti-Passerini C, Pinna LA. Unique substrate specificity of anaplastic lymphoma kinase (ALK): development of phosphoacceptor peptides for the assay of ALK activity. Biochemistry. 2005;44:8533–8542.
    1. Lee CC, Jia Y, Li N, Sun X, Ng K, Ambing E, Gao MY, Hua S, Chen C, Kim S, Michellys PY, Lesley SA, Harris JL, Spraggon G. Crystal structure of the ALK (anaplastic lymphoma kinase) catalytic domain. Biochem. J. 2010;430:425–437.
    1. Cobb MH, Sang BC, Gonzalez R, Goldsmith E, Ellis L. Autophosphorylation activates the soluble cytoplasmic domain of the insulin receptor in an intermolecular reaction. J. Biol. Chem. 1989;264:18701–18706.
    1. Favelyukis S, Till JH, Hubbard SR, Miller WT. Structure and autoregulation of the insulin-like growth factor 1 receptor kinase. Nat. Struct. Mol. Biol. 2001;8:1058–1063.
    1. Furdui CM, Lew ED, Schlessinger J, Anderson KS. Autophosphorylation of FGFR1 kinase is mediated by a sequential and precisely ordered reaction. Mol. Cell. 2006;21:711–717.
    1. Grace MR, Walsh CT, Cole PA. Divalent ion effects and insights into the catalytic mechanism of protein tyrosine kinase Csk. Biochemistry. 1997;36:1874–1881.
    1. Wente SR, Villalba M, Schramm VL, Rosen OM. Mn2(+)-binding properties of a recombinant protein-tyrosine kinase derived from the human insulin receptor. Proc. Natl. Acad. Sci. U.S.A. 1990;87:2805–2809.
    1. Till JH, Becerra M, Watty A, Lu Y, Ma Y, Neubert TA, Burden SJ, Hubbard SR. Crystal structure of the MuSK tyrosine kinase: insights into receptor autoregulation. Structure. 2002;10:1187–1196.
    1. Eck MJ, Yun CH. Structural and mechanistic underpinnings of the differential drug sensitivity of EGFR mutations in non-small cell lung cancer. Biochim. Biophys. Acta. 2010;1804:559–566.
    1. Sabbatini P, Korenchuk S, Rowand JL, Groy A, Liu Q, Leperi D, Atkins C, Dumble M, Yang J, Anderson K, Kruger RG, Gontarek RR, Maksimchuk KR, Suravajjala S, Lapierre RR, Shotwell JB, Wilson JW, Chamberlain SD, Rabindran SK, Kumar R. GSK1838705A inhibits the insulin-like growth factor-1 receptor and anaplastic lymphoma kinase and shows antitumor activity in experimental models of human cancers. Mol. Cancer Ther. 2009;8:2811–2820.
    1. Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat. Rev. Cancer. 2007;7:169–181.
    1. Carey KD, Garton AJ, Romero MS, Kahler J, Thomson S, Ross S, Park F, Haley JD, Gibson N, Sliwkowski MX. Kinetic analysis of epidermal growth factor receptor somatic mutant proteins shows increased sensitivity to the epidermal growth factor receptor tyrosine kinase inhibitor, erlotinib. Cancer Res. 2006;66:8163–8171.
    1. Yun CH, Mengwasser KE, Toms AV, Woo MS, Greulich H, Wong KK, Meyerson M, Eck MJ. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc. Natl. Acad. Sci. U.S.A. 2008;105:2070–2075.
    1. Zhou W, Ercan D, Chen L, Yun CH, Li D, Capelletti M, Cortot AB, Chirieac L, Iacob RE, Padera R, Engen JR, Wong KK, Eck MJ, Gray NS, Jänne PA. Novel mutant-selective EGFR kinase inhibitors against EGFR T790M. Nature. 2009;462:1070–1074.

Source: PubMed

3
订阅