The Effect of Initial Molecular Profile on Response to Recombinant Interferon Alpha (rIFNα) Treatment in Early Myelofibrosis

Introduction Successful treatment of early myelofibrosis (eMF), defined as patients (pts) with low or intermediate-1 (int-1) risk and ≥15% hematopoietic foci in the marrow biopsy, with rIFNα has been reported, but effects of driver and epigenetic mutations on outcomes were not considered. We now report results of our single center phase II study of 30 pts with primary (PM) or secondary (SMF) eMF, correlating the baseline molecular profile with response to rIFNα treatment. Methods We used WHO 2008 and IWG-MRT criteria for the diagnosis of PM and post-polycythemia vera MF (pPV MF) or post-essential thrombocythemia MF (pET MF), respectively. Prognosis was assessed using the Dynamic International Prognostic Scoring System (DIPSS). Response classifications were complete response (CR), partial response (PR), clinical improvement (CI), stable disease (SD), progressive disease (PD). Evaluation included history, examination, spleen size (cm below left costal margin), CBC and differential counts, routine chemistries, liver, renal, and thyroid function tests, marrow biopsy, cytogenetic and molecular genetic evaluations. Marrow specimens were stained routinely for morphology, reticulin, and collagen; fibrosis was graded using the European consensus system. JAK2, CALR and MPL mutations were detected and quantified by amplicon based next generation sequencing (NGS). NGS of 45 genes recurrently mutated in myeloid malignancies was performed on 25 of 30 baseline specimens, including ASXL1, IDH1/2, EZH2 and SRSF2, previously defined as high molecular risk (HMR) genes. Descriptive statistics were calculated and Fisher's exact test used to explore the relationship between mutations and various results. Pts received either rIFNα-2b or peg-rIFNα-2a starting at a 5x105 to 1x106 units SC 3X wk or 45 to 90 mcg weekly, respectively; dose increase dependent upon tolerance and/or response. Results Of 96 pts with PM or SMF, 30 met study criteria, including 16 women and 14 men (med. dx age 58 yrs). Twenty-one had PM, 7 pPV MF, and 2 pET MF. Twenty-one pts were low risk, 9 int-1. Twenty-two were JAK2V617F(+), 6 CALR(+), and 2 MPL(+). All 7 pPV MF pts were JAK2(+). One pET MF was JAK2(+) and 1 CALR(+). Of PM pts, 14 were JAK2(+), 5 CALR(+), and 2 MPL(+). Med. rx duration has been 5.6 yrs and is ongoing. Clinical toxicity, grade 1 or 2, included mild depression, dry skin, cough, and myalgia. Hem. toxicity included anemia (grade 1-2: 17 pts), leukopenia (grade 1-2: 9 pts), and thrombocytopenia (grade 1-2: 10; grade 3: 2 pts). In these cases, rIFNα dose was temporarily reduced/interrupted until cytopenias resolved. One pt developed hyperthyroidism; treatment was discontinued. Of 30 pts, 2 (7%) had CR, 9 (30%) PR, 4 (13%) CI, 7 (23%) SD. Four (13%) had PD, and 4 (13%) died. Nine of 21 low risk pts had CR or PR, compared with 2 of 9 int-1. A small spleen at baseline was associated with better response in the absence of HMR abnormalities. Of 18 pts with slight or no splenomegaly, 11 had CR or PR, and of the non-responders, 2 had a HMR. Of 12 pts with significant or moderate splenomegaly, none had CR or PR, including 2 with a HMR. Follow up biopsies in 25 pts (83%) a reduction in reticulin fibrosis in 5, increase in 9, and stable in 10. Decrease in cellularity occurred in 12. There was no correlation between decrease in JAK2 allele burden, response, fibrosis, or spleen size. There was no correlation between driver mutations and response (Table). Pts with a HMR had worse responses; of 4 such pts, 0 responded, 1 remained stable, 1 progressed and 2 died. Specifically, pts with ASXL1 mutations seemed to do worse than those without it, although sample size precludes a definitive statement (p=0.19). Conclusion This is the first study considering the effect of baseline mutation status on response to rIFNα treatment in eMF pts. 73% of pts improved or remained stable regardless of initial low or int-1 score or driver mutation, but spleen size and HMR adversely affected treatment response. Although baseline driver mutation status was not prognostically useful, a molecular profile should be obtained initially to identify HMR mutations, which negatively affect response to treatment including rIFNα. In 73% of selected cases, low dose rIFNα in eMF resulted in marrow reversion, regression of splenomegaly or disease stability with tolerable toxicity. Early treatment of MF with rIFNα in pts without HMR may prevent the development of marked splenomegaly, anemia, and florid myelofibrosis. ![Figure][1] Disclosures Silver: Incyte: Membership on an entity's Board of Directors or advisory committees. Ritchie: Pfizer: Honoraria; Incyte: Speakers Bureau; Celgene: Speakers Bureau; Novartis: Honoraria, Research Funding; Arian: Speakers Bureau. Roboz: Agois: Consultancy; Amgen: Consultancy; Amphivena: Consultancy; Astex: Consultancy; AstraZeneca: Consultancy; Boehringer Ingelheim: Consultancy; Celator: Consultancy; Celgene: Consultancy; CTI Biopharma: Consultancy; Genoptix: Consultancy; Janssen Pharamaceuticals: Consultancy; Junno: Consultancy; MedImmune: Consultancy; MEI Pharma: Consultancy; Novartis: Consultancy; Onconova: Consultancy; Pfizer: Consultancy; • Roche Pharmaceuticals: Consultancy; Sunesis: Consultancy; Teva: Consultancy. Orazi: Novartis: Honoraria. Tam: Millennium Pharmaceuticals, Inc.: Consultancy. Cross: Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau. [1]: pending:yes

[1]  M. Cazzola,et al.  Epidemiology and clinical relevance of mutations in postpolycythemia vera and postessential thrombocythemia myelofibrosis: A study on 359 patients of the AGIMM group , 2016, American journal of hematology.

[2]  R. Silver,et al.  Optimal therapy for polycythemia vera and essential thrombocythemia: Preferred use of interferon therapy based on phase 2 trials , 2016, Hematology.

[3]  R. Silver,et al.  Recombinant interferon-α in myelofibrosis reduces bone marrow fibrosis, improves its morphology and is associated with clinical response , 2015, Modern Pathology.

[4]  H. Kantarjian,et al.  Correlation of mutation profile and response in patients with myelofibrosis treated with ruxolitinib. , 2015, Blood.

[5]  O. Hofmann,et al.  VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research , 2016, Nucleic acids research.

[6]  R. Silver,et al.  Interferon in polycythemia vera and related neoplasms. Can it become the treatment of choice without a randomized trial? , 2015, Expert review of hematology.

[7]  Mingming Jia,et al.  COSMIC: exploring the world's knowledge of somatic mutations in human cancer , 2014, Nucleic Acids Res..

[8]  Ayalew Tefferi,et al.  CME Information , 2014, Stahl's Illustrated Violence.

[9]  M. Cazzola,et al.  Clinical effect of driver mutations of JAK2, CALR, or MPL in primary myelofibrosis. , 2014, Blood.

[10]  P. Guglielmelli,et al.  CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients , 2014, Leukemia.

[11]  M. Cazzola,et al.  Impact of mutational status on outcomes in myelofibrosis patients treated with ruxolitinib in the COMFORT-II study. , 2014, Blood.

[12]  M. Cazzola,et al.  Mutations and prognosis in primary myelofibrosis , 2013, Leukemia.

[13]  Francisco Cervantes,et al.  Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet (ELN) consensus report. , 2013, Blood.

[14]  Heng Li Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM , 2013, 1303.3997.

[15]  T. Barbui,et al.  Experience with pegylated interferon α-2a in advanced myeloproliferative neoplasms in an international cohort of 118 patients , 2012, Haematologica.

[16]  J. Reilly,et al.  Improving survival trends in primary myelofibrosis: an international study. , 2012, Journal of Clinical Oncology.

[17]  Joaquín Dopazo,et al.  SNPeffect 4.0: on-line prediction of molecular and structural effects of protein-coding variants , 2011, Nucleic Acids Res..

[18]  R. Silver,et al.  Recombinant interferon-α may retard progression of early primary myelofibrosis: a preliminary report. , 2011, Blood.

[19]  R. Hoffman,et al.  The renaissance of interferon therapy for the treatment of myeloid malignancies. , 2011, Blood.

[20]  F. Passamonti,et al.  DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  M. Cazzola,et al.  A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). , 2010, Blood.

[22]  H. Kantarjian,et al.  The role of cytogenetic abnormalities as a prognostic marker in primary myelofibrosis: applicability at the time of diagnosis and later during disease course. , 2009, Blood.

[23]  P. Campbell,et al.  Proposed criteria for the diagnosis of post-polycythemia vera and post-essential thrombocythemia myelofibrosis: a consensus statement from the international working group for myelofibrosis research and treatment , 2008, Leukemia.

[24]  C. Bloomfield,et al.  Proposals and rationale for revision of the World Health Organization diagnostic criteria for polycythemia vera, essential thrombocythemia, and primary myelofibrosis: recommendations from an ad hoc international expert panel. , 2007, Blood.

[25]  A. Jones,et al.  Amplification refractory mutation system, a highly sensitive and simple polymerase chain reaction assay, for the detection of JAK2 V617F mutation in chronic myeloproliferative disorders. , 2007, The Journal of molecular diagnostics : JMD.

[26]  D. Oscier,et al.  Minimal molecular response in polycythemia vera patients treated with imatinib or interferon alpha. , 2005, Blood.

[27]  A. Tefferi,et al.  Prognostic diversity among cytogenetic abnormalities in myelofibrosis with myeloid metaplasia , 2005, Cancer.

[28]  R. Mesa,et al.  Cytogenetic findings and their clinical relevance in myelofibrosis with myeloid metaplasia , 2001, British journal of haematology.

[29]  Helen E White,et al.  Evaluation of methods to detect CALR mutations in myeloproliferative neoplasms. , 2015, Leukemia research.

[30]  M. Cazzola,et al.  The number of prognostically detrimental mutations and prognosis in primary myelofibrosis: an international study of 797 patients , 2014, Leukemia.

[31]  J. Rey,et al.  PEG-IFN-alpha-2a therapy in patients with myelofibrosis: a study of the French Groupe d'Etudes des Myelofibroses (GEM) and France Intergroupe des syndromes Myéloprolifératifs (FIM). , 2009, British journal of haematology.

[32]  Ayalew Tefferi,et al.  Primary myelofibrosis. , 2008, Cancer treatment and research.

[33]  J. Sloan,et al.  The burden of fatigue and quality of life in myeloproliferative disorders (MPDs) , 2007, Cancer.

[34]  J. D. van der Walt,et al.  European consensus on grading bone marrow fibrosis and assessment of cellularity. , 2005, Haematologica.