Adjunction of a fish oil emulsion to cytarabine and daunorubicin induction chemotherapy in high-risk AML

Emmanuel Gyan, Arnaud Pigneux, Mathilde Hunault, Pierre Peterlin, Martin Carré, Jacques-Olivier Bay, Caroline Bonmati, Maria-Pilar Gallego-Hernanz, Bruno Lioure, Philippe Bertrand, Nicolas Vallet, David Ternant, François Darrouzain, Frédéric Picou, Marie-Christine Béné, Christian Récher, Olivier Hérault, Emmanuel Gyan, Arnaud Pigneux, Mathilde Hunault, Pierre Peterlin, Martin Carré, Jacques-Olivier Bay, Caroline Bonmati, Maria-Pilar Gallego-Hernanz, Bruno Lioure, Philippe Bertrand, Nicolas Vallet, David Ternant, François Darrouzain, Frédéric Picou, Marie-Christine Béné, Christian Récher, Olivier Hérault

Abstract

The treatment of acute myeloid leukemia (AML) with unfavorable cytogenetics treatment remains a challenge. We previously established that ex vivo exposure of AML blasts to eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), or fish oil emulsion (FO) induces Nrf2 pathway activation, metabolic switch, and cell death. The FILO group launched a pilot clinical study to evaluate the feasibility, safety, and efficacy of the adjunction of a commercial FO emulsion to 3 + 7 in untreated AML with unfavorable cytogenetics. The primary objective was complete response (CR). Thirty patients were included. FO administration raised the plasma levels of eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids (p < 0.001). The pharmacokinetics of cytarabine and daunorubicin were unaffected. A historical comparison to the LAM2001 trial (Lioure et al. Blood 2012) found a higher frequency of grade 3 serious adverse events, with no drug-related unexpected toxicity. The CR rate was 77%, and the partial response (PR) 10%, not significantly superior to that of the previous study (CR 72%, PR 1%). RT-qPCR analysis of Nrf2 target genes and antioxidant enzymes did not show a significant in vivo response. Overall, FO emulsion adjunction to 3 + 7 is feasible. An improvement in CR was not shown in this cohort of high-risk patients. The present data does not support the use of FO in adjunction with 3 + 7 in high-risk AML patients.ClinicalTrials.gov identifier: NCT01999413.

Conflict of interest statement

EG received honoraria from Fresenius Kabi. All other authors declare no competing interests relevant for this publication.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
FAMYLY study design. Patients eligible for the study were treated with fish oil emulsion, daunorubicin and cytarabine at the indicated doses. If the baseline white blood count (WBC) was below 30 G/L, patients received 48 of fish oil before the start of chemotherapy. If the WBC was above or equal to 30 G/L, all treatments were started on the same day. An evaluation bone marrow (BM) evaluation was performed on D15. If the BM blasts were higher than 5%, a second induction was provided at the indicated doses.
Figure 2
Figure 2
PUFA plasma pharmacokinetics. Plasma samples were drawn at baseline (H0), 24 and 48 h after the start of FO infusion, after the end of infusion (D10), and at disease evaluation (D28–D35). The PUFA composition of plasma was evaluated by gas chromatography. The sum of docosahexaenoic acid (DHA) and eicosapentaenoic (EPA) acid concentration is indicated for each individual patient (colored lines and dots). Means are indicated by a dash for each timepoint.
Figure 3
Figure 3
Historical comparison of survival data from the FAMYLY and LAM2001 trials. Overall survival (OS) was plotted from the inclusion date to the date of death or last follow up. The vertical marks indicate censored observations.

References

    1. Song E, Kim H. Docosahexaenoic acid induces oxidative DNA damage and apoptosis, and enhances the chemosensitivity of cancer cells. Int. J. Mol. Sci. 2016;17:1257. doi: 10.3390/ijms17081257.
    1. Ceccarelli V, Racanicchi S, Martelli MP, et al. Eicosapentaenoic acid demethylates a single cpg that mediates expression of tumor suppressor CCAAT/enhancer-binding protein δ in U937 leukemia cells. J. Biol. Chem. 2011;286:27092–27102. doi: 10.1074/jbc.M111.253609.
    1. Yamagami T, Porada CD, Pardini RS, et al. Docosahexaenoic acid induces dose dependent cell death in an early undifferentiated subtype of acute myeloid leukemia cell line. Cancer Biol. Ther. 2009;8:331–337. doi: 10.4161/cbt.8.4.7334.
    1. Arita K, Kobuchi H, Utsumi T, et al. Mechanism of apoptosis in HL-60 cells induced by n-3 and n-6 polyunsaturated fatty acids. Biochem. Pharmacol. 2001;62:821–828. doi: 10.1016/S0006-2952(01)00723-7.
    1. Aires V, Hichami A, Filomenko R, et al. Docosahexaenoic acid induces increases in [Ca2+]i via inositol 1,4,5-triphosphate production and activates protein kinase Cγ and -δ via phosphatidylserine binding site: Implication in apoptosis in U937 cells. Mol. Pharmacol. 2007;72:1545–1556. doi: 10.1124/mol.107.039792.
    1. Picou F, Debeissat C, Bourgeais J, et al. n-3 Polyunsaturated fatty acids induce acute myeloid leukemia cell death associated with mitochondrial glycolytic switch and Nrf2 pathway activation. Pharmacol. Res. 2018;136:45–55. doi: 10.1016/j.phrs.2018.08.015.
    1. Bougnoux P, Hajjaji N, Ferrasson MN, et al. Improving outcome of chemotherapy of metastatic breast cancer by docosahexaenoic acid: A phase II trial. Br. J. Cancer. 2009;101:1978–1985. doi: 10.1038/sj.bjc.6605441.
    1. Hajjaji N, Bougnoux P. Selective sensitization of tumors to chemotherapy by marine-derived lipids: A review. Cancer Treat. Rev. 2013;39:473–488. doi: 10.1016/j.ctrv.2012.07.001.
    1. Werner K, Küllenberg de Gaudry D, Taylor LA, et al. Dietary supplementation with n-3-fatty acids in patients with pancreatic cancer and cachexia: marine phospholipids versus fish oil—A randomized controlled double-blind trial. Lipids Health Dis. 2017;16:104. doi: 10.1186/s12944-017-0495-5.
    1. Shirai Y, Okugawa Y, Hishida A, et al. Fish oil-enriched nutrition combined with systemic chemotherapy for gastrointestinal cancer patients with cancer cachexia. Sci. Rep. 2017;7:4826. doi: 10.1038/s41598-017-05278-0.
    1. Chagas TR, Borges DS, de Oliveira PF, et al. Oral fish oil positively influences nutritional-inflammatory risk in patients with haematological malignancies during chemotherapy with an impact on long-term survival: A randomised clinical trial. J. Hum. Nutr. Diet. 2017;30:681–692. doi: 10.1111/jhn.12471.
    1. Elbarbary NS, Ismail EAR, Farahat RK, El-Hamamsy M. ω-3 fatty acids as an adjuvant therapy ameliorates methotrexate-induced hepatotoxicity in children and adolescents with acute lymphoblastic leukemia: A randomized placebo-controlled study. Nutrition. 2016;32:41–47. doi: 10.1016/j.nut.2015.06.010.
    1. Li NN, Zhou Y, Qin XP, et al. Does intravenous fish oil benefit patients post-surgery? A meta-analysis of randomised controlled trials. Clin. Nutr. 2014;33:226–239. doi: 10.1016/j.clnu.2013.08.013.
    1. Barbosa VM, Miles EA, Calhau C, et al. Effects of a fish oil containing lipid emulsion on plasma phospholipid fatty acids, inflammatory markers, and clinical outcomes in septic patients: A randomized, controlled clinical trial. Crit. Care. 2010;14:1–11. doi: 10.1186/cc8844.
    1. Lioure B, Béné MC, Pigneux A, et al. Early matched sibling hematopoietic cell transplantation for adult AML in first remission using an age-adapted strategy: Long-term results of a prospective GOELAMS study. Blood. 2012;119:2943–2948. doi: 10.1182/blood-2011-05-352989.
    1. Mayer K, Fegbeutel C, Hattar K, et al. ω-3 vs. ω-6 lipid emulsions exert differential influence on neutrophils in septic shock patients: Impact on plasma fatty acids and lipid mediator generation. Intensive Care Med. 2003;29:1472–1481. doi: 10.1007/s00134-003-1900-2.
    1. Mayer K, Gokorsch S, Fegbeutel C, et al. Parenteral nutrition with fish oil modulates cytokine response in patients with sepsis. Am. J. Respir. Crit. Care Med. 2003;167:1321–1328. doi: 10.1164/rccm.200207-674OC.
    1. Hong RL, Lin CH, Chao TY, et al. A phase-I study evaluating the combination of pegylated liposomal doxorubicin and paclitaxel as salvage chemotherapy in metastatic breast cancer previously treated with anthracycline. Cancer Chemother. Pharmacol. 2008;61:847–853. doi: 10.1007/s00280-007-0542-4.
    1. Acar N, Berdeaux O, Grégoire S, et al. Lipid composition of the human eye: Are red blood cells a good mirror of retinal and optic nerve fatty acids? PLoS ONE. 2012 doi: 10.1371/journal.pone.0035102.
    1. Vandesompele J, de Preter K, Pattyn F, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002 doi: 10.1186/gb-2002-3-7-research0034.
    1. Fleming T. One-sample multiple testing procedure for phase II clinical trials. Biometrics. 1982;1:143–151. doi: 10.2307/2530297.
    1. Fleming RA, Capizzi RL, Rosner GL, et al. Clinical pharmacology of cytarabine in patients with acute myeloid leukemia: A cancer and leukemia group B study. Cancer Chemother. Pharmacol. 1995;36:425–430. doi: 10.1007/BF00686192.
    1. Krogh-Madsen M, Bender B, Jensen MK, et al. Population pharmacokinetics of cytarabine, etoposide, and daunorubicin in the treatment for acute myeloid leukemia. Cancer Chemother. Pharmacol. 2012;69:1155–1163. doi: 10.1007/s00280-011-1800-z.
    1. Kokenberg E, van der Steuijt K, Löwenberg B, et al. Pharmacokinetics of daunorubicin as a determinant of response in acute myeloid leukemia. Haematol. Blood Transfus. 1987;30:283–287. doi: 10.1007/978-3-642-71213-5_42.
    1. Galettis P, Boutagy J, Ma DDF. Daunorubicin pharmacokinetics and the correlation with P-glycoprotein and response in patients with acute leukaemia. Br. J. Cancer. 1994;70:324–329. doi: 10.1038/bjc.1994.301.
    1. Varatharajan S, Panetta JC, Abraham A, et al. Population pharmacokinetics of Daunorubicin in adult patients with acute myeloid leukemia. Cancer Chemother. Pharmacol. 2016;78:1051–1058. doi: 10.1007/s00280-016-3166-8.
    1. Andersson B, Andersson I, Beran M, et al. Liquid chromatographic monitoring of daunorubicin and daunorubicinol in plasma from leukemic patients treated with daunorubicin or the daunorubicin-DNA complex. Cancer Chemother. Pharmacol. 1979;2:15–17. doi: 10.1007/BF00253099.
    1. Ma C-J, Wu J-M, Tsai H-L, et al. Prospective double-blind randomized study on the efficacy and safety of an n-3 fatty acid enriched intravenous fat emulsion in postsurgical gastric and colorectal cancer patients. Nutr. J. 2015 doi: 10.1186/1475-2891-14-9.
    1. Layani-Bazar A, Skornick I, Berrebi A, et al. Redox modulation of adjacent thiols in vla-4 by as101 converts myeloid leukemia cells from a drug-resistant to drug-sensitive state. Cancer Res. 2014;74:3092–3103. doi: 10.1158/0008-5472.CAN-13-2159.
    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. doi: 10.1056/nejmoa1614359.
    1. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N. Engl. J. Med. 2018;378:2386–2398. doi: 10.1056/nejmoa1716984.
    1. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130:722–731. doi: 10.1182/blood-2017-04-779405.
    1. Lancet JE, Uy GL, Cortes JE, et al. Cpx-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J. Clin. Oncol. 2018;36:2684–2692. doi: 10.1200/JCO.2017.77.6112.
    1. DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2019;133:7–17. doi: 10.1182/blood-2018-08-868752.
    1. Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33:379–389. doi: 10.1038/s41375-018-0312-9.

Source: PubMed

3
Sottoscrivi