Targeting SAMHD1 with hydroxyurea in first-line cytarabine-based therapy of newly diagnosed acute myeloid leukaemia: Results from the HEAT-AML trial

Martin Jädersten, Ingrid Lilienthal, Nikolaos Tsesmetzis, Magda Lourda, Sofia Bengtzén, Anna Bohlin, Cornelia Arnroth, Tom Erkers, Brinton Seashore-Ludlow, Géraldine Giraud, Giti S Barkhordar, Sijia Tao, Linda Fogelstrand, Leonie Saft, Päivi Östling, Raymond F Schinazi, Baek Kim, Torsten Schaller, Gunnar Juliusson, Stefan Deneberg, Sören Lehmann, Georgios Z Rassidakis, Martin Höglund, Jan-Inge Henter, Nikolas Herold, Martin Jädersten, Ingrid Lilienthal, Nikolaos Tsesmetzis, Magda Lourda, Sofia Bengtzén, Anna Bohlin, Cornelia Arnroth, Tom Erkers, Brinton Seashore-Ludlow, Géraldine Giraud, Giti S Barkhordar, Sijia Tao, Linda Fogelstrand, Leonie Saft, Päivi Östling, Raymond F Schinazi, Baek Kim, Torsten Schaller, Gunnar Juliusson, Stefan Deneberg, Sören Lehmann, Georgios Z Rassidakis, Martin Höglund, Jan-Inge Henter, Nikolas Herold

Abstract

Background: Treatment of newly diagnosed acute myeloid leukaemia (AML) is based on combination chemotherapy with cytarabine (ara-C) and anthracyclines. Five-year overall survival is below 30%, which has partly been attributed to cytarabine resistance. Preclinical data suggest that the addition of hydroxyurea potentiates cytarabine efficacy by increasing ara-C triphosphate (ara-CTP) levels through targeted inhibition of SAMHD1.

Objectives: In this phase 1 trial, we evaluated the feasibility, safety and efficacy of the addition of hydroxyurea to standard chemotherapy with cytarabine/daunorubicin in newly diagnosed AML patients.

Methods: Nine patients were enrolled and received at least two courses of ara-C (1 g/m2 /2 h b.i.d. d1-5, i.e., a total of 10 g/m2 per course), hydroxyurea (1-2 g d1-5) and daunorubicin (60 mg/m2 d1-3). The primary endpoint was safety; secondary endpoints were complete remission rate and measurable residual disease (MRD). Additionally, pharmacokinetic studies of ara-CTP and ex vivo drug sensitivity assays were performed.

Results: The most common grade 3-4 toxicity was febrile neutropenia (100%). No unexpected toxicities were observed. Pharmacokinetic analyses showed a significant increase in median ara-CTP levels (1.5-fold; p = 0.04) in patients receiving doses of 1 g hydroxyurea. Ex vivo, diagnostic leukaemic bone marrow blasts from study patients were significantly sensitised to ara-C by a median factor of 2.1 (p = 0.0047). All nine patients (100%) achieved complete remission, and all eight (100%) with validated MRD measurements (flow cytometry or real-time quantitative polymerase chain reaction [RT-qPCR]) had an MRD level <0.1% after two cycles of chemotherapy. Treatment was well-tolerated, and median time to neutrophil recovery >1.0 × 109 /L and to platelet recovery >50 × 109 /L after the start of cycle 1 was 19 days and 22 days, respectively. Six of nine patients underwent allogeneic haematopoietic stem-cell transplantation (allo-HSCT). With a median follow-up of 18.0 (range 14.9-20.5) months, one patient with adverse risk not fit for HSCT experienced a relapse after 11.9 months but is now in second complete remission.

Conclusion: Targeted inhibition of SAMHD1 by the addition of hydroxyurea to conventional AML therapy is safe and appears efficacious within the limitations of the small phase 1 patient cohort. These results need to be corroborated in a larger study.

Keywords: SAMHD1; acute myeloid leukaemia; cytarabine; hydroxyurea; precision medicine; targeted therapy.

Conflict of interest statement

T. S. is employed by Heidelberg ImmunoTherapeutics, not relevant to this work. J. I. H. is a consultant for SOBI, not relevant to this work. The other authors declare no conflicts of interest.

© 2022 The Authors. Journal of Internal Medicine published by John Wiley & Sons Ltd on behalf of Association for Publication of The Journal of Internal Medicine.

Figures

Fig. 1
Fig. 1
Treatment outcome. The nine patients have been followed for 14.9–20.5 months. Six patients have undergone allogeneic haematopoietic stem cell transplantation (allo‐HSCT). Measurable residual disease (MRD) was performed in accordance with national guidelines after cycle 2 and cycle 4 or prior to allo‐HSCT. In non‐transplanted patients with NPM1, MRD by real‐time quantitative polymerase chain reaction (RT‐qPCR) was performed every 3 months. After allo‐HSCT, patients were routinely monitored every 3 months either by RT‐qPCR (NPM1) or flow cytometry (all others). Eight of nine patients remain in MRD negative remission (as defined by MRD <0.1% by flow cytometry or RT‐qPCR). *, Patient 1106 had bone marrow blasts 5.5% after cycle 1 and did not fulfil complete remission (CR) criteria until after cycle 2. However, blasts may have been elevated by granulocyte colony‐stimulating factor (G‐CSF) usage prior to sampling; †, patient 1106 relapsed after 11.9 months and was treated with a combination of azacitidine–venetoclax–gilteritinib (AZA‐VEN‐GILT) and is now in a second CR; ‡, in patient 1107, NPM1 RT‐qPCR MRD increased from negative to 0.00024%, was rechecked and was then negative again. Before receiving the result of the confirmatory MRD, the patient was put on azacitidine–venetoclax (AZA‐VEN), and three cycles were given as an additional consolidation due to the inconsistent results; §, patient 1108 received one cycle of AZA as bridging while waiting for allo‐HSCT.
Fig. 2
Fig. 2
Leukemic clearance after cycle 2. Validated real‐time quantitative polymerase chain reaction (RT‐qPCR) markers for NPM1 and DEK::NUP214, respectively, were analysed in four patients, and all showed mutational clearance <0.1% (relative to the diagnostic level) after cycle 2. In the remaining five patients, one or two mutations were monitored for measurable residual disease (MRD) using deep sequencing (reporting the variant allele frequency). Mutation in RUNX1 was seen in four patients and was reduced to 0.74%–10.5% after cycle 2. In two of four patients with RUNX1, a FLT3‐TKD was also present, but was reduced below 0.05% after two cycles of treatment. After cycle 2, MRD levels below the detection limit for the method are indicated with open circles.
Fig. 3
Fig. 3
SAMHD1 protein expression in acute myeloid leukaemia (AML) blasts and in remission bone marrow. (a) Upper images show haematoxylin and eosin routine staining in diagnostic bone marrow biopsies. The lower images show the corresponding SAMHD1‐CD68 double immunohistochemical staining. SAMHD1 (in brown) is mainly expressed in the nucleus of the AML blasts, while CD68+ macrophages (magenta) are strongly positive for SAMHD1 and serve as an internal positive control. Low, intermediate and high expression levels were defined as <25%, 25%–75% and >75% positive cells, respectively, as previously described [21]. (b) Upper images show haematoxylin–eosin routine staining in remission bone marrow biopsies from patients 1105 (left), 1101 (middle) and 1104 (right), respectively, with cellularity within normal range and morphology suggestive of reactive bone marrow. Lower images show SAMHD1/CD34 double immunostaining without double‐positive blasts. Occasionally, SAMHD1+/CD34− normal blasts are present. The SAMHD1+/CD34− cells are morphologically mostly histiocytes and reactive lymphocytes. All images are shown at 400× magnification.
Fig. 4
Fig. 4
Effect of hydroxyurea on peak ara‐C triphosphate (ara‐CTP) levels in circulating mononuclear cells and ex vivo sensitivity to ara‐C. The left panel shows ara‐CTP levels measured in circulating mononuclear cells without hydroxyurea (w/o HU) as compared to ara‐CTP with hydroxyurea 1000 mg taken p.o. 1 h prior to start of ara‐C infusion, normalised to without hydroxyurea (n = 6). The right panel shows the fold reduction in IC50 values of ara‐C in diagnostic bone marrow mononuclear cells treated ex vivo in the presence or absence of 100 μM hydroxyurea, normalised to the absence of hydroxyurea (n = 7). Individual dots correspond to individual patients; colours represent levels of SAMHD1 expression at diagnosis (green, <25%, orange 25%–75% and red >75%).

References

    1. Dores GM, Devesa SS, Curtis RE, Linet MS, Morton LM. Acute leukemia incidence and patient survival among children and adults in the United States, 2001–2007. Blood. 2012;119(1):34–43.
    1. Juliusson G, Antunovic P, Derolf A, Lehmann S, Möllgård L, Stockelberg D, et al. Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood. 2009;113(18):4179–87.
    1. Juliusson G. Older patients with acute myeloid leukemia benefit from intensive chemotherapy: an update from the Swedish Acute Leukemia Registry. Clin Lymphoma Myeloma Leuk. 2011;11(Suppl 1):S54–9.
    1. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–47.
    1. Fernandez HF, Sun Z, Yao X, Litzow MR, Luger SM, Paietta EM, et al. Anthracycline dose intensification in acute myeloid leukemia. N Engl J Med. 2009;361(13):1249–59.
    1. Dinardo CD, Wei AH. How I treat acute myeloid leukemia in the era of new drugs. Blood. 2020;135(2):85–96.
    1. Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454–64.
    1. Borthakur G, Kantarjian H. Core binding factor acute myelogenous leukemia‐2021 treatment algorithm. Blood Cancer J. 2021;11(6):114.
    1. Cornelissen JJ, Van Putten WLJ, Verdonck LF, Theobald M, Jacky E, Daenen SMG, et al. Results of a HOVON/SAKK donor versus no‐donor analysis of myeloablative HLA‐identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle‐aged adults: benefits for whom? Blood. 2007;109(9):3658–66.
    1. Mayer RJ, Davis RB, Schiffer CA, Berg DT, Powell BL, Schulman P, et al. Intensive postremission chemotherapy in adults with acute myeloid leukemia. Cancer and Leukemia Group B. N Engl J Med. 1994;331(14):896–903.
    1. Lowenberg B. Sense and nonsense of high‐dose cytarabine for acute myeloid leukemia. Blood. 2013;121(1):26–8.
    1. Tsesmetzis N, Paulin CBJ, Rudd SG, Herold N. Nucleobase and nucleoside analogues: resistance and re‐sensitisation at the level of pharmacokinetics, pharmacodynamics and metabolism. Cancers. 2018;10(7):240.
    1. Liliemark JO, Plunkett W, Dixon DO. Relationship of 1‐beta‐D‐arabinofuranosylcytosine in plasma to 1‐beta‐D‐arabinofuranosylcytosine 5'‐triphosphate levels in leukemic cells during treatment with high‐dose 1‐beta‐D‐arabinofuranosylcytosine. Cancer Res. 1985;45(11 Pt 2):5952–7.
    1. Yamauchi T, Kawai Y, Kishi S, Goto N, Urasaki Y, Imamura S, et al. Monitoring of intracellular 1‐beta‐D‐arabinofuranosylcytosine 5'‐triphosphate in 1‐beta‐D‐arabinofuranosylcytosine therapy at low and conventional doses. Jpn J Cancer Res. 2001;92(5):546–53.
    1. Estey E, Plunkett W, Dixon D, Keating M, McCredie K, Freireich EJ. Variables predicting response to high dose cytosine arabinoside therapy in patients with refractory acute leukemia. Leukemia. 1987;1(8):580–3.
    1. Plunkett W, Heinemann V, Estey E, Keating M. Pharmacologically directed design of leukemia therapy. Haematol Blood Transfus. 1990;33:610–3.
    1. Plunkett W, Iacoboni S, Estey E, Danhauser L, Liliemark JO, Keating MJ. Pharmacologically directed ara‐C therapy for refractory leukemia. Semin Oncol. 1985;12(2 Suppl 3):20–30.
    1. Riva‐Lavieille C, Ressayre C, Barra Y, Carcassonne Y, Iliadis A. Calculation of individual dosage regimen of cytosine arabinoside (ara‐C) based on metabolite levels in leukemic cells. Ther Drug Monit. 1994;16(4):375–9.
    1. Herold N, Rudd SG, Ljungblad L, Sanjiv K, Myrberg IH, Paulin CBJ, et al. Targeting SAMHD1 with the Vpx protein to improve cytarabine therapy for hematological malignancies. Nature Medicine. 2017;23(2):256–63.
    1. Herold N, Rudd SG, Sanjiv K, Kutzner J, Myrberg IH, Paulin CBJ, et al. With me or against me: tumor suppressor and drug resistance activities of SAMHD 1. Exp Hematol. 2017;52:32–9.
    1. Rassidakis GZ, Herold N, Myrberg IH, Tsesmetzis N, Rudd SG, Henter J‐I, et al. Low‐level expression of SAMHD1 in acute myeloid leukemia (AML) blasts correlates with improved outcome upon consolidation chemotherapy with high‐dose cytarabine‐based regimens. Blood Cancer J. 2018;8(11):98.
    1. Rudd SG, Schaller T, Herold N. SAMHD1 is a barrier to antimetabolite‐based cancer therapies. Mol Cell Oncol. 2017;4(2):3.
    1. Schneider C, Oellerich T, Baldauf H‐M, Schwarz S‐M, Thomas D, Flick R, et al. SAMHD1 is a biomarker for cytarabine response and a therapeutic target in acute myeloid leukemia. Nat Med. 2017;23(2):250–5.
    1. Rudd SG, Tsesmetzis N, Sanjiv K, Paulin CBJ, Sandhow L, Kutzner J, et al. Ribonucleotide reductase inhibitors suppress SAMHD1 ara‐CTPase activity enhancing cytarabine efficacy. EMBO Mol Med. 2020;12(3):e10419.
    1. Herold N. Pharmacological strategies to overcome treatment resistance in acute myeloid leukemia: increasing leukemic drug exposure by targeting the resistance factor SAMHD1 and the toxicity factor Top2β. Expert Opin Drug Discov. 2021;16(1):7–11.
    1. Musiałek MW, Rybaczek D. Hydroxyurea—the good, the bad and the ugly. Genes (Basel). 2021;12(7):1096.
    1. Yataganas X, Strife A, Perez A, Clarkson B. Cell kill kinetics with hydroxyurea. Med Pediatr Oncol. 1976;2(1):39–54.
    1. de Montalembert M, Bachir D, Hulin A, Gimeno L, Mogenet A, Bresson JL, et al. Pharmacokinetics of hydroxyurea 1,000 mg coated breakable tablets and 500 mg capsules in pediatric and adult patients with sickle cell disease. Haematologica. 2006;91(12):1685–8.
    1. Gandhi V, Estey E, Keating MJ, Plunkett W. Fludarabine potentiates metabolism of cytarabine in patients with acute myelogenous leukemia during therapy. J Clin Oncol. 1993;11(1):116–24.
    1. Ossenkoppele GJ. The value of fludarabine in addition to ARA‐C and G‐CSF in the treatment of patients with high‐risk myelodysplastic syndromes and AML in elderly patients. Blood. 2004;103(8):2908–13.
    1. Fridland A, Verhoef V. Mechanism for ara‐CTP catabolism in human leukemic cells and effect of deaminase inhibitors on this process. Semin Oncol. 1987;14(2 Suppl 1):262–8.
    1. Marsh JH, Kreis W, Barile B, Akerman S, Schulman P, Allen SL, et al. Therapy of refractory/relapsed acute myeloid leukemia and blast crisis of chronic myeloid leukemia with the combination of cytosine arabinoside, tetrahydrouridine, and carboplatin. Cancer Chemother Pharmacol. 1993;31(6):481–4.
    1. Mamez A‐C, Raffoux E, Chevret S, Lemiale V, Boissel N, Canet E, et al. Pre‐treatment with oral hydroxyurea prior to intensive chemotherapy improves early survival of patients with high hyperleukocytosis in acute myeloid leukemia. Leuk Lymphoma. 2016;57(10):2281–8.
    1. Bagger FO, Kinalis S, Rapin N. BloodSpot: a database of healthy and malignant haematopoiesis updated with purified and single cell mRNA sequencing profiles. Nucleic Acids Research. 2018;47(D1):D881–5.
    1. Tobal K, Frost L, Liu Yin JA. Quantification of DEK‐CAN fusion transcript by real‐time reverse transcription polymerase reaction in patients with t(6;9) acute myeloid leukemia. Haematologica. 2004;89(10):1267–9.
    1. Delsing Malmberg E, Rehammar A, Pereira MB, Abrahamsson J, Samuelsson T, Ståhlman S, et al. Accurate and sensitive analysis of minimal residual disease in acute myeloid leukemia using deep sequencing of single nucleotide variations. J Mol Diagn. 2019;21(1):149–62.
    1. Rosso A, Juliusson G, Lorenz F, Lehmann S, Derolf Ã, Deneberg S, et al. Is there an impact of measurable residual disease as assessed by multiparameter flow cytometry on survival of AML patients treated in clinical practice? A population‐based study. Leuk Lymphoma. 2021;62(8):1973–81.
    1. Pemovska T, Kontro M, Yadav B, Edgren H, Eldfors S, Szwajda A, et al. Individualized systems medicine strategy to tailor treatments for patients with chemorefractory acute myeloid leukemia. Cancer Discov. 2013;3(12):1416–29.
    1. Yadav B, Wennerberg K, Aittokallio T, Tang J. Searching for drug synergy in complex dose‐response landscapes using an interaction potency model. Comput Struct Biotechnol J. 2015;13:504–13.
    1. Potdar S, Ianevski A, Mpindi J‐P, Bychkov D, Fiere CM, Ianevski P, et al. Breeze: an integrated quality control and data analysis application for high‐throughput drug screening. Bioinformatics. 2020;36(11):3602–4.
    1. Yamauchi T, Ueda T, Nakamura T. A new sensitive method for determination of intracellular 1‐beta‐D‐arabinofuranosylcytosine 5'‐triphosphate content in human materials in vivo. Cancer Res. 1996;56(8):1800–4.
    1. Yamauchi T, Negoro E, Kishi S, Takagi K, Yoshida A, Urasaki Y, et al. Intracellular cytarabine triphosphate production correlates to deoxycytidine kinase/cytosolic 5′‐nucleotidase II expression ratio in primary acute myeloid leukemia cells. Biochem Pharmacol. 2009;77(12):1780–6.
    1. Gandhi V, Xu YZ, Estey E. Accumulation of arabinosyluracil 5'‐triphosphate during arabinosylcytosine therapy in circulating blasts of patients with acute myelogenous leukemia. Clin Cancer Res. 1998;4(7):1719–26.
    1. Plunkett W, Hug V, Keating MJ, Chubb S. Quantitation of 1‐beta‐D‐arabinofuranosylcytosine 5'‐triphosphate in the leukemic cells from bone marrow and peripheral blood of patients receiving 1‐beta‐D‐arabinofuranosylcytosine therapy. Cancer Res. 1980;40(3):588–91.
    1. Estey E, Plunkett W, Gandhi V, Rios MB, Kantarjian H, Keating MJ. Fludarabine and arabinosylcytosine therapy of refractory and relapsed acute myelogenous leukemia. Leuk Lymphoma. 1993;9(4–5):343–50.
    1. Juliusson G, Jädersten M, Deneberg S, Lehmann S, Möllgård L, Wennström L, et al. The prognostic impact of FLT3‐ITD and NPM1 mutation in adult AML is age‐dependent in the population‐based setting. Blood Adv. 2020;4(6):1094–101.
    1. Marras T, Dettori M, Caocci G, La Nasa G, Sotgiu G, Saderi L, et al. White blood cell count nadir and duration of aplasia do not associate with treatment outcome in adult patients with acute myeloid leukemia undergoing intensive chemotherapy. Chemotherapy. 2020;65(3–4):110–4.
    1. Juliusson G, Hagberg O, Lazarevic VLJ, Ölander E, Antunovic P, Cammenga J, et al. Improved survival of men 50 to 75 years old with acute myeloid leukemia over a 20‐year period. Blood. 2019;134(18):1558–61.
    1. Bazzell BG, Marini BL, Benitez LL, Bixby D, Burke P, Pettit K, et al. Real world use of FLT3 inhibitors for treatment of FLT3+ acute myeloid leukemia (AML): a single center, propensity‐score matched, retrospective cohort study. J Oncol Pharm Pract. 2021. 10.1177/10781552211020815.
    1. Lindsley RC, Mar BG, Mazzola E, Grauman PV, Shareef S, Allen SL, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood. 2015;125(9):1367–76.
    1. Jongen‐Lavrencic M, Grob T, Hanekamp D, Kavelaars FG, Al Hinai A et al. Molecular minimal residual disease in acute myeloid leukemia. N Engl J Med. 2018;378(13):1189–99.
    1. Wang H, He X, Zhang L, Dong H, Huang F, Xian J, et al. Disruption of dNTPs homeostasis by ribonucleotide reductase hyperactivation overcomes AML differentiation blockade. Blood. 2022:3752–70.

Source: PubMed

3
Sottoscrivi