Use of Sorafenib in FLT3-Mutant Myeloid Sarcoma: A Case Report
Giulia Zamprogna*, Elisa Zucchetti, Sofia Bozzani, Giovanni Grillo, Roberto Cairoli
Department of Hematology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano, Piazza Ospedale Maggiore 3, Milano, Italy
*Corresponding author: Giulia Zamprogna, Department of Hematology, Niguarda Cancer Center, ASST Grande Ospedale Metropolitano, Piazza Ospedale Maggiore 3, Milano, Italy. Tel: +39 02 64442668; Fax: +39 02 64443263; Email: giulia.zamprogna@gmail.com
Received Date: 13 September, 2019; Accepted Date: 16 October, 2019; Published Date: 21 October, 2019
Citation: Zamprogna G, Zucchetti E, Bozzani S, Grillo G, Cairoli R (2019) Use of Sorafenib in FLT3-Mutant Myeloid Sarcoma: A Case Report. Ann Case Report 12: 259. DOI: 10.29011/2574-7754/100259
Abstract
A 53-years old man with FLT3-ITD-mutant Acute Myeloid Leukemia relapsed ten months after hemopoietic allogeneic stem cell transplant as multiple cutaneous Myeloid Sarcomas. Molecular testing was performed on the core-needle biopsy of the lesion and since positive for the FLT3 mutation, a targeted-therapy with the tyrosine kinase inhibitor sorafenib as single agent was started, obtaining a sustained complete response. After 18 months of therapy patient relapsed on the previously involved sites, because of acquisition of sorafenib-resistant mutation and switched to systemic therapy. Sorafenib proved to be able to distribute in skin and subcutaneous tissues and could be safely used. Molecular testing on Myeloid Sarcoma can be therefore an innovative and effective approach in the management of extra medullary leukemia.
Keywords
FLT3 Mutation; Myeloid Sarcoma; Sorafenib; Target-Therapy
Introduction
FMS-Like Tyrosine Kinase 3 (FLT3) is a receptor tyrosine kinase expressed by hematopoietic stem cells whose activity is essential to normal stem cell function and development. Mutations of this gene are described in about 30% of patients with Acute Myeloid Leukemia (AML) [1,2]. These mutations proved to significantly affect the course, prognosis and outcome of AML, for this reason in recent years a huge effort on the study of potential inhibitors of FLT3 has been performed. FLT3 inhibitors act by blocking the proliferative boost due to the mutation, playing a relevant role in the treatment of FLT3-mutant AML, either as monotherapy or in combination with standard chemotherapy, with promising results [3-5].
Hence FLT3 mutation detection, along with other common genetic alterations, has become a gold standard for diagnosis and management of AML [6]. By contrast it is not usually searched for Myeloid Sarcoma (MS) a rare disease that can present as an isolated or multiple extra medullary leukemic tumor, concurrently with or at relapse of AML [7]. In view of the strict relation of these conditions, we report the case of a patient with FLT3-mutant AML whose disease relapsed after allogeneic stem cell transplant as a MS, on which FLT3 mutation was tested, and since positive, managed by using the FLT3-inhibitor sorafenib, based on the proved synergistic effect of the Tyrosine Kinase Inhibitor (TKI) after Allogeneic Stem Cell Transplantation (allo-SCT) [8].
Case Reports
A 53 years old man diagnosed with FLT3-ITD mutated AML was treated with standard induction and consolidation therapy before receiving, in first complete molecular remission, hematopoietic allogeneic stem cell transplant forms an HLA-identical sibling. Complete hematological, cytogenetic and molecular remission was confirmed at the hundred days’ post-transplant evaluation. Ten months after transplant a subcutaneous nodule of augmented consistence appeared in the pectoral region. Patient was completely asymptomatic, with normal lab tests and bone marrow evaluation showed negative MRD. Ultrasound of the nodule showed an ovoid shaped mass with a 2 cm diameter and mixed structure. After 1 month of observation, new subcutaneous lesions with similar US features started to appear in the right cervical region, in the context of left deltoid muscle, right intercostal and left dorsal regions, on both thighs and on right tibial surface (Figure1).
All sites presented an increased uptake at PET scan with a SUVmax of 5.22. A percutaneous biopsy of the pectoral nodule was done. Histological examination described the presence of myeloid blasts consistent with the diagnosis of MS, which was confirmed at the immunophenotype that showed: CD68+; CD45+, MPO+, CD117+, CD30+, MIB 1/Ki67 80%, TdT negative, CD20-, CD3-, CD34-, TRKA-. As FLT3-ITD mutation with an allelic ratio of 10,22 was detected in the analyzed specimen, a targeted therapy with sorafenib was started. Sorafenib was administrated at escalating doses till target 800 mg die (400 mg bid), reached in 45 days. Therapy was well tolerated, showing a rapid effect on subcutaneous nodules, which not only stopped to grow out but fully disappear in two weeks. A PET scan was performed showing a Complete Response which was consolidated by DLI infusion (6,8 x 10^7/kg CD3+) at week 17 after sorafenib beginning, with no complications and no GVHD manifestations. Six weeks from first DLI, a second infusion (1x10^8/Kg CD3+) was performed, again no significant complication occurred.
Because of the appearance of diarrhea and palmar-plantar erythrodysesthesia, side effects related to sorafenib, dose was adjusted to alternated 600-800 mg die at the time of first DLI infusion. Due to persistence of diarrhea, sorafenib was reduced to a 600 mg daily 2 weeks from first adjustment with complete resolution of the symptom. Thirty-three weeks from sorafenib start and 8 weeks from the second DLI infusion a firm lesion appeared in the context of left deltoid muscle at level of the insertion in the humerus, turning out to be a single MS relapse site at PET scan, with bone marrow negativity. Radiotherapy (RT) of the lesion (36 Gy) was started obtaining a rapid and complete response. Molecular biology done on the deltoid lesion specimen demonstrated again a positivity for FLT3 mutation, but this time either in the Internal Tandem Duplication (ITD) region and in the Tyrosine Kinase 2 Domain (TKD) at Asp835His. Eighteen months after the start of sorafenib and 5 months after RT a new rapidly growing lesion appeared in left axilla positive to PET scan. Sorafenib was then discontinued and patient switched to Azacitidine plus DLI obtaining the fourth complete response.
Discussion
Myeloid Sarcoma (MS) is a rare extra medullary tumor composed of immature myeloid cells expressing CD34, CD117, CD68, MPO and TdT negative, characterized by the occurrence of one or more tumor masses that can involve any site of the body. MS may precede myeloid neoplasms, being concomitant to, represent the relapse, or occur de novo. An increased report of MS presentation after allogeneic stem cell transplantation suggested to possibly represent a reduced graft-versus-leukemia effect at extramedullary sites [9,10]. The outcome of MS is poor independently from the setting of onset and median survival is less than 24 months in most cases [11,12]. A variety of chromosomal abnormalities have been described, among which translocation t (8;21) (q22; q22) is regarded as a recurrent one [9]. Although the study of genetic lesions is critical for AML, it is not routinely performed in MS; thus our approach represented an innovation in this setting where only few studies on genetic information are available. Notably Nucleophosmin (NPM) 1 mutation was identified in a set of MS patients, while FLT3-ITD mutation was detected in a 33% of patients presenting with MS and concurrent AML [13,14].
Activating mutations in the FLT3 gene, involving Internal Tandem Duplications (ITD) in the juxtamembrane domain, and missense point mutations in the Tyrosine Kinase Domain (TKD) involving Aspartic Acid Amino Position 835 (D835), are frequently observed in patients with AML, with ITD mutations being much more frequent than TKDs. They are detected in up to 30% of adult AML cases, with an increased incidence in patients with normal karyotype [3,4]. These mutations lead to overexpression or constitutive activation of the tyrosine kinase sustaining tumor growth and are associated with poor prognosis mainly related to increased relapse risk, there is no study describing the impact of FLT3 mutation in MS [4,15]. Several agents targeting FLT3 have been studied among which the oral multi-kinase inhibitor sorafenib, which is able to inhibit the activities of FLT3, VEGFR-2, VEGFR-3, and members of the platelet-derived growth factor receptor family such as PDGFR-beta and Kit both in vitro and in vivo. It is known to potently inhibit FLT3 enzymatic and cellular activities and to induce significant tumor growth inhibition [16,17].
Food and Drug Administration approved sorafenib for the treatment of hepatocellular, renal cell and differentiated thyroid carcinomas; its use has been successfully tested in many non-hematologic neoplasms, mainly angiosarcoma and soft tissues sarcomas [18,19]. Sorafenib is a reasonably safe drug and can be used with no need of drug reduction in renal and/or hepatic dysfunction, side effects are easily managed by dose titration [18]. In the setting of FLT3-ITD positive AML this agent has been tested in several trials at induction in combination with cytarabine and anthracycline chemotherapy, as in the SORAML trial, despite an excessive toxicity [20]. On the contrary in the setting of relapsed/refractory disease, it proved to be a valuable option either as a single agent or combined with hypomethylating agents [21,22]. Recent data from SORMAIN trial, showed that the risk of relapse or death is significantly reduced by sorafenib maintenance therapy after allo-SCT, possibly related, as postulated by a previous study, to a synergistic anti-leukemic effect of the Tyrosine Kinase Inhibitor (TKI) and the allo-immune effect of the graft [8,23].
Based on these evidences and to patient history, once FLT3-ITD mutation was detected, we choose to attempt a targeted sorafenib-based treatment to our patient. To our knowledge this is the second time a similar approach has been used in clinical practice [24]. Target therapy provides a valuable alternative to traditional chemotherapy, not only in terms of efficacy but also for a significant impact on the quality of life, considering that lower short term toxicity and no need of hospitalization. Interestingly, in our experience sorafenib proved to be able to distribute into skin and soft tissues and, by reaching the sarcoma, to provide a rapid and complete response although the acquisition in the long run of resistances, that could be seen as the proof of its action on MS. A major mechanism of TKI resistance is caused by selection of specific genetic alterations within the target kinase, even though primary resistances to FLT3-TKI monotherapy were proved in approximately 30% of FLT3-mutated AML.
In clinical studies where FLT3-TKD mutations have appeared in addition to ITD ones during disease progression, it remained unclear whether the double mutants were present before or were acquired after treatment with FLT3 inhibitors. Based on this observation a model of clonal evolution explaining the resistance to FLT3 inhibitors proposed that tumor clones with FLT3-ITD may acquire TKD mutation because of treatment genotoxicity, so double mutant clones carrying both FLT3-ITD and TKD mutations, are selected during treatment leading to disease progression and loss of TKI response [25-27]. In the era of precision medicine our experience suggests to detect molecular lesions sensitive to target therapy in MS as it is routinely done in AML, in view of the unquestionable effectiveness we faced. More studies have to be performed for a better understanding of the action of target agents in MS and whether their combination with other agents, such as hypomethylating agents and DLI, could reduce the risk of resistant mutations onset.
Conclusion
We were able to demonstrate that a complete and durable response could be obtained with sorafenib as single agent in MS after allo-HSCT, hence target therapy can be convincingly used in MS driven by molecular testing, thus representing an innovative approach to this disease that may provide rationale for a genetic-oriented classification and management of MS in the future.
Acknowledgement
The authors declare that they have no conflict of interest.
Figure 1: A 53-years old man
with FLT3-mutant Acute Myeloid Leukemia relapsed 10 months after allogeneic
HSCT as multiple cutaneous Myeloid Sarcomas and was successfully treated with
sorafenib monotherapy. A close view of one of the rapidly growing nodules and
its ultrasound appearance.
References
- Gilliland DG, Griffin JD (2002) The roles of FLT3 in hematopoiesis and leukemia. Blood 100: 1532-1542.
- Meshinchi S, Appelbaum FR (2009) Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res 15: 4263-4269.
- Moreno I, Martin G, Bolufer P, Barragan E, Rueda E, et al. (2003) Incidence and prognostic value of FLT3 internal tandem duplication and D835 mutations in acute myeloid leukemia. Haematologica 88: 19-24.
- Thiede C, Steudel C, Mohr B, Schaich M, Schakel U, et al. (2002) Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 99: 4326-4335.
- Tse KF, Allebach J, Levis M, Smith BD, Bohmer FD, et al. (2002) Inhibition of the transforming activity of FLT3 internal tandem duplication mutants from patients by a tyrosine kinase inhibitor. Leukemia 16: 2027-2036.
- Rowe JM, Tallman MS (2010) How I treat acute myeloid leukemia. Blood 116: 3147-3156.
- Bakst RL, Tallman MS, Douer D, Yahalom J (2011) How I treat extramedullary acute myeloid leukemia. Blood 118: 3785-3793.
- Metzelder SK, Schroeder T, Finck A, Scholl S, Fey M, et al. (2012) High activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained responses. Leukemia 26: 2353-2359.
- Pileri S, Orazi A, Falini B (2017) Myeloid sarcoma in: Swerdlow S, Campo E, Harris NL, et al. (eds.) WHO Classification of tumors of hematopoietic and lymphoid tissues. V ed. IARC, Lyon; 140-141.
- Pileri SA, Ascani S, Cox MC, Campidelli C, Bacci F, et al. (2007) Myeloid sarcoma: clinico-pathologic, phenotypic and cytogenetic analysis of 92 adult patients. Leukemia 21: 340-350.
- Lan TY, Lin DT, Tien HF, Yang RS, Chen CY, et al. (2009) Prognostic factors of treatment outcomes in patients with granulocytic sarcoma. Acta Haematol 122: 238-246.
- Kaur V, Swami A, Alapat D, Abdallah AO, Motwani P, et al. (2018) Clinical characteristics, molecular profile and outcomes of myeloid sarcoma: a single institution experience over 13 years. Hematology 23: 17-24.
- Falini B, Lenze D, Hasserjian R, Coupland S, Jaehne D, et al. (2007) Cytoplasmic mutated nucleophosmin (NPM) defines the molecular status of a significant fraction of myeloid sarcomas. Leukemia 21: 1566-1570.
- Ansari-Lari MA, Yang CF, Tinawi-Aljundi R, Cooper L, Long P, et al. (2004) FLT3 mutations in myeloid sarcoma. Br J Haematol 126: 785-791.
- Yanada M, Matsuo K, Suzuki T, Kiyoi H, Naoe T (2005) Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis. Leukemia 19: 1345-1349.
- Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, et al. (2006) Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nature Reviews Drug Discovery 5: 835-844.
- Auclair D, Miller D, Yatsula V, Pickett W, Carter C, et al. (2007) Antitumor activity of sorafenib in FLT3-driven leukemic cells. Leukemia 21: 439-445.
- European Medicines Agency. Sorafenib (Nexavar): product information [online].
- Maki RG, D'Adamo DR, Keohan ML, Saulle M, Schuetze SM, et al. (2009) Phase II Study of Sorafenib in Patients with Metastatic or Recurrent Sarcomas. J Clin Oncol 27: 3133-3140.
- Röllig C, Serve H, Hüttmann A, Noppeney R, Müller-Tidow C, et al. (2015) Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. The Lancet Oncology 16: 1691-1699.
- Muppidi MR, Portwood S, Griffiths EA, Thompson JE, Ford LA, et al. (2015) Decitabine and Sorafenib Therapy in FLT-3 ITD-Mutant Acute Myeloid Leukemia. Clin Lymphoma Myeloma Leuk S73-S79.
- Ravandi F, Alattar ML, Grunwald MR, Rudek MA, Rajkhowa T, et al. (2013) Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood 121: 4655-4662.
- Burchert A, Bug G, Finke J, Stelljes M, Rollig C, et al. (2018) Sorafenib as Maintenance Therapy Post Allogeneic Stem Cell Transplantation for FLT3-ITD Positive AML: Results from the Randomized, Double-Blind, Placebo-Controlled Multicentre Sormain Trial. Dec 3; Oral Abstract #661: 60th ASH Annual Meeting and Exposition, San Diego, CA.
- Choschzick M, Bacher U, Ayuk F, Lebeau A (2010) Immunohistochemistry and molecular analyses in myeloid sarcoma of the breast in a patient with relapse of NPM1-mutated and FLT3-mutated AML after allogeneic stem cell transplantation. J Clin Pathol 63: 558-561.
- Kindler T, Lipka DB, Fischer T (2010) FLT3 as a therapeutic target in AML: still challenging after all these years. Blood 116: 5089-5102.
- Piloto O, Wright M, Brown P, Kim KT, Levis M, et al. (2007) Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. Blood 109: 1643-1652.
- Man CH, Fung TK, Ho C, Han HHC, Chow HCH, et al. (2012) Sorafenib treatment of FLT3-ITD+ acute myeloid leukemia: favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation. Blood 119: 5133-5143.
© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. Read More About Open Access Policy.