Glasdegib in combination with cytarabine and daunorubicin in patients with AML or high-risk MDS: Phase 2 study results

Jorge E Cortes, B Douglas Smith, Eunice S Wang, Akil Merchant, Vivian G Oehler, Martha Arellano, Daniel J DeAngelo, Daniel A Pollyea, Mikkael A Sekeres, Tadeusz Robak, Weidong Wendy Ma, Mirjana Zeremski, M Naveed Shaik, A Douglas Laird, Ashleigh O'Connell, Geoffrey Chan, Mark A Schroeder, Jorge E Cortes, B Douglas Smith, Eunice S Wang, Akil Merchant, Vivian G Oehler, Martha Arellano, Daniel J DeAngelo, Daniel A Pollyea, Mikkael A Sekeres, Tadeusz Robak, Weidong Wendy Ma, Mirjana Zeremski, M Naveed Shaik, A Douglas Laird, Ashleigh O'Connell, Geoffrey Chan, Mark A Schroeder

Abstract

Glasdegib is a Hedgehog pathway inhibitor. This ongoing, open-label, phase 2 study (NCT01546038) evaluated glasdegib plus cytarabine/daunorubicin in patients with untreated acute myeloid leukemia (AML) or high-risk myelodysplastic syndromes (MDS). Patients received glasdegib 100 mg orally, once daily in continuous 28-day cycles from day -3, with intravenous cytarabine 100 mg/m2 on days 1-7 and daunorubicin 60 mg/m2 on days 1-3. Patients in remission then received consolidation therapy (2-4 cycles of cytarabine 1 g/m2 twice daily on days 1, 3, 5 of each cycle), followed by maintenance glasdegib (maximum 6 cycles). Primary endpoint was complete remission (CR) in patients aged ≥55 years. Secondary endpoints included overall survival (OS), safety and outcome by mutational status. Patients had a median (range) age of 64.0 (27-75) years, 60.0% were male, and 84.5% were white. In 69 evaluable patients, 46.4% (80% confidence interval [CI]: 38.7-54.1) achieved investigator-reported CR. Among patients ≥55 years old (n = 60), 40.0% (80% CI 31.9-48.1) achieved CR. Among all 69 patients, median OS was 14.9 (80% CI 13.4-19.3) months, with 12-month survival probability 66.6% (80% CI 58.5-73.4). The most common treatment-related adverse events (≥50% patients) were diarrhea and nausea. There were no significant associations between mutational status (12 genes) and clinical response, suggesting potential benefit across diverse molecular profiles. Glasdegib plus cytarabine/daunorubicin was well tolerated and associated with clinical activity in patients with untreated AML or high-risk MDS. A randomized phase 3 trial of glasdegib in combination with chemotherapy (7 + 3 schedule) is ongoing.

© 2018 The Authors. American Journal of Hematology published by Wiley Periodicals, Inc.

Figures

Figure 1
Figure 1
A, Kaplan‐Meier plot of OS in all patients; B, patients with AML by cytogenetic risk and C, patients aged ≥55 years with and without censoring for patients receiving a transplant. Adv, adverse; AML, acute myeloid leukemia; CI, confidence interval; NE, not evaluable; Fav, favorable; Int, intermediate; OS, overall survival [Color figure can be viewed at http://wileyonlinelibrary.com]

References

    1. Daver N, Kantarjian H, Ravandi F, et al. A phase II study of decitabine and gemtuzumab ozogamicin in newly diagnosed and relapsed acute myeloid leukemia and high‐risk myelodysplastic syndrome. Leukemia. 2016;30:268‐273.
    1. Stone RM, Mazzola E, Neuberg D, et al. Phase III open‐label randomized study of cytarabine in combination with amonafide l‐malate or daunorubicin as induction therapy for patients with secondary acute myeloid leukemia. J Clin Oncol. 2015;33:1252‐1257.
    1. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX‐351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood. 2014;123:3239‐3246.
    1. Löwenberg B, Ossenkoppele GJ, van Putten W, et al. High‐dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med. 2009;361:1235‐1248.
    1. Seymour JF, Fenaux P, Silverman LR, et al. Effects of azacitidine compared with conventional care regimens in elderly (≥75 years) patients with higher‐risk myelodysplastic syndromes. Crit Rev Oncol Hematol. 2010;76:218‐227.
    1. De Kouchkovsky I, Abdul‐Hay M. Acute myeloid leukemia: a comprehensive review and 2016 update. Blood Cancer J. 2016;6:e441.
    1. Dombret H, Gardin C. An update of current treatments for adult acute myeloid leukemia. Blood. 2016;127:53‐61.
    1. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377:454‐464.
    1. Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001;15:3059‐3087.
    1. Campbell V, Copland M. Hedgehog signaling in cancer stem cells: a focus on hematological cancers. Stem Cells Cloning. 2015;8:27‐38.
    1. Aberger F, Hutterer E, Sternberg C, et al. Acute myeloid leukemia: strategies and challenges for targeting oncogenic Hedgehog/GLI signaling. Cell Commun Signal. 2017;15:8.
    1. Singh M, Chaudhry P, Merchant AA. Primary cilia are present on human blood and bone marrow cells and mediate Hedgehog signaling. Exp Hematol. 2016;44:1181‐1187. e1182.
    1. Irvine DA, Copland M. Targeting Hedgehog in hematologic malignancy. Blood. 2012;119:2196‐2204.
    1. Fukushima N, Minami Y, Kakiuchi S, et al. Small‐molecule Hedgehog inhibitor attenuates the leukemia‐initiation potential of acute myeloid leukemia cells. Cancer Sci. 2016;107:1422‐1429.
    1. Queiroz KC, Ruela‐de‐Sousa RR, Fuhler GM, et al. Hedgehog signaling maintains chemoresistance in myeloid leukemic cells. Oncogene. 2010;29:6314‐6322.
    1. Zhou H, Ma H, Wei W, et al. B4GALT family mediates the multidrug resistance of human leukemia cells by regulating the Hedgehog pathway and the expression of p‐glycoprotein and multidrug resistance‐associated protein 1. Cell Death Dis. 2013;4:e654.
    1. Zhao C, Chen A, Jamieson CH, et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature. 2009;458:776‐779.
    1. Dierks C, Beigi R, Guo GR, et al. Expansion of Bcr‐Abl‐positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell. 2008;14:238‐249.
    1. Lim Y, Gondek L, Li L, et al. Integration of Hedgehog and mutant FLT3 signaling in myeloid leukemia. Sci Transl Med. 2015;7:291ra296.
    1. Munchhof MJ, Li Q, Shavnya A, et al. Discovery of PF‐04449913, a potent and orally bioavailable inhibitor of Smoothened. ACS Med Chem Lett. 2012;3:106‐111.
    1. Jackson‐Fisher AJ, McMahon MJ, Lam J, et al. PF‐04449913, a small molecule inhibitor of Hedgehog signaling, is effective in inhibiting tumor growth in preclinical models. Cancer Res. 2011;71:4504.
    1. Schairer A, Shih A, Geron I, et al. Human blast crisis leukemia stem cell inhibition with a novel Smoothened antagonist. Blood. 2010;116:1223‐1223.
    1. Jackson‐Fisher A, Whalen P, Elliott M, et al. Interrogating Hedgehog pathway and Smoothened inhibition by PF‐04449913 in patient‐derived acute myeloid leukemia models. Cancer Res. 2014;74:1958.
    1. Minami Y, Fukushima N, Sadarangani A, Minami H, Jamieson C, Naoe T. Treatment with Hedgehog inhibitor PF‐913 attenuates leukemia‐initiation potential in acute myeloid leukemia cells. Cancer Res. 2014;74:1884‐1884.
    1. Martinelli G, Oehler VG, Papayannidis C, et al. Treatment with PF‐04449913, an oral smoothened antagonist, in patients with myeloid malignancies: a phase 1 safety and pharmacokinetics study. Lancet Haematol. 2015;2:e339‐e346.
    1. Cortes JE, Heidel FH, Heuser M, et al. A phase 2 randomized study of low dose Ara‐C with or without glasdegib (PF‐04449913) in untreated patients with acute myeloid leukemia or high‐risk myelodysplastic syndrome. Blood. 2016;128:99‐99.
    1. Sadarangani A, Pineda G, Lennon KM, et al. GLI2 inhibition abrogates human leukemia stem cell dormancy. J Transl Med. 2015;13:98.
    1. Tibes R, Al‐Kali A, Oliver GR, et al. The Hedgehog pathway as targetable vulnerability with 5‐azacytidine in myelodysplastic syndrome and acute myeloid leukemia. J Hematol Oncol. 2015;8:114.
    1. Savona MR, Pollyea DA, Stock W, et al. Phase IB study of glasdegib, a Hedgehog pathway inhibitor, in combination with standard chemotherapy in patients with AML or high‐risk MDS. Clin Cancer Res. 2018;24:2294‐2303.
    1. Cheson BD, Greenberg PL, Bennett JM, et al. Clinical application and proposal for modification of the international working group (IWG) response criteria in myelodysplasia. Blood. 2006;108:419‐425.
    1. Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations of the international working Group for Diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol. 2003;21:4642‐4649.
    1. Rollig C, Bornhauser M, Thiede C, et al. Long‐term prognosis of acute myeloid leukemia according to the new genetic risk classification of the European LeukemiaNet recommendations: evaluation of the proposed reporting system. J Clin Oncol. 2011;29:2758‐2765.
    1. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079‐2088.
    1. Bishop JF, Matthews JP, Young GA, et al. A randomized study of high‐dose cytarabine in induction in acute myeloid leukemia. Blood. 1996;87:1710‐1717.
    1. Buchner T, Schlenk RF, Schaich M, et al. Acute myeloid leukemia (AML): different treatment strategies versus a common standard arm—combined prospective analysis by the German AML intergroup. J Clin Oncol. 2012;30:3604‐3610.
    1. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer. 2006;106:1794‐1803.
    1. Kantarjian H, Oki Y, Garcia‐Manero G, et al. Results of a randomized study of 3 schedules of low‐dose decitabine in higher‐risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood. 2007;109:52‐57.
    1. Kantarjian HM, O'Brien S, Huang X, et al. Survival advantage with decitabine versus intensive chemotherapy in patients with higher risk myelodysplastic syndrome: comparison with historical experience. Cancer. 2007;109:1133‐1137.
    1. Kantarjian HM, O'Brien S, Shan J, et al. Update of the decitabine experience in higher risk myelodysplastic syndrome and analysis of prognostic factors associated with outcome. Cancer. 2007;109:265‐273.
    1. Thépot S, Itzykson R, Seegers V, et al. Azacitidine in untreated acute myeloid leukemia: a report on 149 patients. Am J Hematol. 2014;89:410‐416.
    1. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126:291‐299.
    1. Fernandez HF, Sun Z, Yao X, et al. Anthracycline dose intensification in acute myeloid leukemia. N Engl J Med. 2009;361:1249‐1259.
    1. Fenaux P, Mufti GJ, Hellstrom‐Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol. 2010;28:562‐569.
    1. Burnett AK, Milligan D, Goldstone A, et al. The impact of dose escalation and resistance modulation in older patients with acute myeloid leukaemia and high risk myelodysplastic syndrome: the results of the LRF AML14 trial. Br J Haematol. 2009;145:318‐332.
    1. Boyd AL, Salci KR, Shapovalova Z, McIntyre BAS, Bhatia M. Nonhematopoietic cells represent a more rational target of in vivo Hedgehog signaling affecting normal or acute myeloid leukemia progenitors. Exp Hematol. 2013;41:858‐869. e854.
    1. Chaudhry P, Singh M, Triche TJ, Guzman M, Merchant AA. GLI3 repressor determines Hedgehog pathway activation and is required for response to SMO antagonist glasdegib in AML. Blood. 2017;129:3465‐3475.
    1. Minami Y, Minami H, Miyamoto T, et al. Phase I study of glasdegib (PF‐04449913), an oral smoothened inhibitor, in Japanese patients with select hematologic malignancies. Cancer Sci. 2017;108:1628‐1633.
    1. Merchant A, Matsui W. Targeting Hedgehog—a cancer stem cell pathway. Clin Cancer Res. 2010;16:3130‐3140.
    1. Alonso S, Hernandez D, Chang YT, et al. Hedgehog and retinoid signaling alters multiple myeloma microenvironment and generates bortezomib resistance. J Clin Invest. 2016;126:4460‐4468.
    1. Alonso S, Jones RJ, Ghiaur G. Retinoic acid, CYP26, and drug resistance in the stem cell niche. Exp Hematol. 2017;54:17‐25.
    1. Krug U, Koschmieder A, Schwammbach D, et al. Feasibility of azacitidine added to standard chemotherapy in older patients with acute myeloid leukemia: a randomised SAL pilot study. PLoS One. 2012;7:e52695.
    1. Löwenberg B, Pabst T, Vellenga E, et al. Cytarabine dose for acute myeloid leukemia. N Engl J Med. 2011;364:1027‐1036.
    1. Jain S, Song R, Xie J. Sonidegib: mechanism of action, pharmacology, and clinical utility for advanced basal cell carcinomas. Onco Targets Ther. 2017;10:1645‐1653.
    1. Jacobsen AA, Aldahan AS, Hughes OB, Shah VV, Strasswimmer J. Hedgehog pathway inhibitor therapy for locally advanced and metastatic basal cell carcinoma: a systematic review and pooled analysis of interventional studies. JAMA Dermatol. 2016;152:816‐824.
    1. Ruiz‐Salas V, Alegre M, Lopez‐Ferrer A, et al. Vismodegib: a review. Acta Dermosifiliogr. 2014;105:744‐751.
    1. Sekulic A, Migden MR, Basset‐Seguin N, et al. Long‐term safety and efficacy of vismodegib in patients with advanced basal cell carcinoma: final update of the pivotal ERIVANCE BCC study. BMC Cancer. 2017;17:332.
    1. Lear JT, Migden MR, Lewis KD, et al. Long‐term efficacy and safety of sonidegib in patients with locally advanced and metastatic basal cell carcinoma: 30‐month analysis of the randomized phase 2 BOLT study. J Eur Acad Dermatol Venereol. 2018;32:372‐381.

Source: PubMed

3
Suscribir