scholarly journals Methotrexate and cytarabine for adult patients with newly diagnosed Langerhans cell histiocytosis: A single arm, single center, prospective phase 2 study

2020 ◽  
Vol 95 (9) ◽  
Author(s):  
Xin‐Xin Cao ◽  
Jian Li ◽  
Ai‐Lin Zhao ◽  
Tian‐Hua He ◽  
Xue‐Min Gao ◽  
...  
Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 635-635 ◽  
Author(s):  
David M. Hyman ◽  
Eli Diamond ◽  
Vivek Subbiah ◽  
Jean-Yves Blay ◽  
A. Craig Lockhart ◽  
...  

Abstract Background: The systemic histiocytic disorders Erdheim-Chester disease (ECD) and Langerhans cell histiocytosis (LCH) are rare hematologic malignancies with heterogenous clinical courses and prognoses that share a common primary event: the pathologic accumulation and infiltration of cells thought to be of the monocyte/macrophage lineage in affected tissue. The recent discovery of BRAFV600E mutations in a high proportion (50-60%) of patients with LCH/ECD has provided pivotal insight into the cause and potential management of these disorders. The VE-BASKET study (ClinicalTrials.gov identifier, NCT01524978) was designed to explore the efficacy and safety of vemurafenib, a selective oral inhibitor of mutated BRAFV600 kinase, in non–melanoma, non–papillary thyroid cancers harboring BRAFV600 mutations. The preliminary efficacy data in adult patients with ECD/LCH are presented herein. Methods: Simon 2-stage adaptive-design, open-label, multicenter, multinational, phase 2 study of vemurafenib in patients with cancer harboring BRAFV600 mutations. The primary objective was to evaluate the efficacy of vemurafenib in patients with BRAFV600 mutation–positive cancers at week 8 by investigator-assessed response rate using RECIST version 1.1. Tumor assessment was performed by computed tomography, magnetic resonance imaging, or physical examination every 8 weeks. Patients without RECIST v1.1 measurable disease were followed up by use of positron emission tomography; these response data will be presented separately. Data cutoff was March 18, 2014. Results: Fifteen patients with ECD/LCH (6 male, 9 female) have been enrolled. Median age is 67 years (range, 35-83). Eleven of 15 patients (73%) received at least 1 prior therapy. Median duration of vemurafenib treatment was 94 days (range, 6-478). Maximal percentage change from baseline in target lesion diameter sum and best overall response are shown in Figure 1A. To date, in 11 patients assessed by RECIST v1.1, overall response rate was 36.4% (95% CI, 10.9-69.2) and clinical benefit rate was 90.9% (95% CI, 58.7-99.8). No patient experienced progression while taking vemurafenib. Best overall responses by RECIST 1.1 are as follows: 1 (9.1%) complete, 3 (27.3%) partial, 6 (54.6%) stable, 0 progressive, and 1 (9.1%) not evaluable. Three patients (20%) discontinued treatment because of adverse events (AEs; grade 3 gastric infection, grade 3 arthralgia, and grade 4 alanine transaminase level elevation); 12 patients are still receiving treatment. Thirteen patients (87%) had at least 1 grade 3 or 4 AE—most commonly skin squamous cell carcinoma (40%)—and actinic keratosis, keratoacanthoma, maculopapular rash, arthralgia, and dehydration (each 13%). No patient died during the study. Two representative patient vignettes are presented herein. Patient 1 is a 68-year-old woman with extensive ECD involving the brain, bones, and retroperitoneum. She previously received high-dose methotrexate and underwent stenting for disease-related renal artery stenosis that resulted in an emergency hypertensive event and renal failure, transiently requiring hemodialysis. At the time of enrollment, this patient had severe gait instability and dysarthria and was unable to perform activities of daily living (ADLs). After 8 weeks of vemurafenib therapy, she achieved partial response (Figure 1B), is ambulating with minimal assistance, and is independent with ADLs. She continues therapy. Patient 2 is a 77-year-old woman with LCH previously treated with vinblastine, methotrexate, an AKT inhibitor, and cladribine. At the time of enrollment she had debilitating cutaneous involvement that required long-term administration of narcotics and resulted in recurrent infection. After 4 weeks of vemurafenib therapy, the patient achieved complete response, including total resolution of skin lesions (Figure 1C). Subsequent biopsy also confirmed complete pathologic response and complete absence of the BRAFV600E mutant protein by immunohistochemistry using the VE1 antibody (Figure 1D). The patient continues therapy. Conclusion: There are no approved therapies for adult patients with multisystem histiocytic disorders. The magnitude and durability of response to vemurafenib in patients with multisystemic BRAFV600-mutated ECD and LCH are encouraging. Updated results will be presented at the American Society of Hematology annual meeting. Figure 1 Figure 1. Disclosures Hyman: Chugai Pharma: Consultancy; Atara Biotherapeutics: Consultancy. Off Label Use: Vemurafenib is a BRAF inhibitor currently approved for treatment of patients with unresectable or metastatic melanoma with BRAF V600E mutation as detected by an FDA-approved test. The data included in this abstract is a subgroup analysis of VE-BASKET, a phase 2 study of vemurafenib in non-melanoma cancer patients harboring BRAFV600 mutations in order to explore the efficacy of vemurafenib in other cancer populations. Subbiah:MD Anderson Cancer Center: Employment. Blay:University Lyon I: Employment; Centre Leon Berard: Employment. Sirzen:F. Hoffmann-La Roche: Employment, Equity Ownership. Veronese:F. Hoffmann-La Roche: Employment. Laserre:F. Hoffmann-La Roche: Employment. Baselga:Roche: Consultancy.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 294-294
Author(s):  
Xinxin Cao ◽  
Jian Li ◽  
Ai-lin Zhao ◽  
Xue-min Gao ◽  
Hua-cong Cai ◽  
...  

Backgroud: Langerhans cell histiocytosis (LCH) is a rare, heterogeneous histiocytic disorder occurring in patients of all ages from neonates to the elderly. The features of LCH are well described in children, however, they remain poorly defined in adults. There is no standard first-line treatment for adult LCH. The current standard treatment protocol for children is vinblastine plus prednisone, which has never been proven effective for adults in a prospective study. Considering the relatively high frequency of pituitary involvement and late onset of neurodegenerative symptoms, patients may benefit from the combination of cytarabine and methotrexate as both these drugs cross the blood-brain barrier. Methods: This phase 2, prospective, single-center study enrolled 83 newly diagnosed adults multisystem (MS)-LCH or LCH with multifocal single system (SS-m) involved between January 2014 and March 2019 (NCT 02389400). The methotrexate (1g/m2by 24-hour infusion on day 1) and cytarabine (100 mg /m2by 24-hour infusion for 5 days) was administered every 35 days for a cycle and 6 cycles totally. The primary endpoint was event-free survival (EFS). Events were defined as a poor response to chemotherapy, reactivation after chemotherapy or death from any cause. Results: The median age was 33 years (range 18-65 years). Forty-nine patients were male (59.0%). Six patients were SS-m LCH (7.2%), 77 patients were MS LCH (92.8%). The most common organ involved in the total cohort was bone (78.3%), followed by lung (67.5%), pituitary (62.7%) and lymph nodes (38.6%). Twenty-three patients had liver involvement (27.7%), 11 patients with spleen involvement (13.3%), no patients had hematologic involvement. All patients received at least one course of chemotherapy, with median 6 (1-6) courses. Overall 69 patients (83.1%) completed protocol treatment, 14 patients (16.9%) went off protocol (13 patients' decision, 1 poor response). The overall response rate was 87.9%. including 43 patients (51.8%) as having non-active disease and 30 patients (36.1%) as active disease (AD)/better. After a median of 23 months (range 7-79 months) follow-up, one patient died of disease progression and 25 patients had reactivation of the disease. The estimated 3-year OS and EFS were 97.7% and 68.0% separately (Figure 1). To evaluate the prognostic factors of EFS using univariate analysis, liver, spleen, lung and skin involvement at baseline had significantly shorter EFS. EFS were also evaluated using multivariate Cox regression model, liver involvement remained predictive of poorer EFS (P = 0.012; HR 0.339, 95% CI 0.146-0.784). The most common toxicity was hematologic adverse events. All patients experienced neutropenia and thrombocytopenia. Thirty-five patients (42.2%) had grade 4 neutropenia, 43 patients (51.8%) had grade 3 neutropenia. Fourteen patients (16.9%) had grade 4 thrombocytopenia, 13 patients (15.7%) had grade 3 thrombocytopenia. No patients received prophylactic antimicrobial treatment during any of the cycles. Forty patients (48.2%) experienced febrile neutropenia, including 38 (45.8%) grade 3 and 2 (2.4%) grade 4. The most common non-hematological toxicities were gastrointestinal complications. Two patients developed grade 3 nausea. Grade 3 alanine aminotransferase increased occurred in in two patients. No treatment related death. One patient had secondary primary malignancy (oral squamous cell carcinoma), 56 months after the last course of MA regimen. Fifty-two of 82 surviving patients experienced sequelae to the disease that were not influenced by therapy. Forty-eight patients had diabetes insipidus and 4 presented with hypothyroidism. Conclusion: Methotrexate and cytarabine is an efficient and safe regimen for newly diagnosed adult LCH. The involvement of liver at baseline indicates a worse prognosis in adult LCH. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (12) ◽  
pp. 1415-1423 ◽  
Author(s):  
Jean Donadieu ◽  
Frederic Bernard ◽  
Max van Noesel ◽  
Mohamed Barkaoui ◽  
Odile Bardet ◽  
...  

Key Points Patients with LCH, risk organs, refractory to standard VBL-steroid regimen have a poor survival, ∼30%. In a phase 2 study, with 5 years’ median follow-up, cladribine and Ara-C was shown to improve the survival up to 85% for this group.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3157-3157
Author(s):  
Adrian C. Newland ◽  
Jean-Francois Viallard ◽  
María Fernanda López Fernández ◽  
Melissa Jeanne Eisen ◽  
Hossam A Saad ◽  
...  

Abstract Background: The clinical course of immune thrombocytopenia (ITP) can be categorized into three phases: newly diagnosed (<3 months post-diagnosis), persistent (≥3-≤12 months) or chronic (>12 months). Having previously been restricted to use in patients with chronic ITP, romiplostim (a thrombopoietin receptor agonist) was recently approved in the USA and Europe for use across all phases of ITP in adult patients refractory to 1st line treatments. Its efficacy and safety in adult patients with ITP diagnosed for ≤6 months have been reported in a Phase 2 study (Newland, BJH 2016); however, its effect in patients with newly diagnosed ITP is not well characterized. Here we report post hoc analyses from the same Phase 2 romiplostim study stratified by duration of ITP diagnosis (<3 months and ≥3-≤12 months). Methods: Adult patients with primary ITP diagnosed within 6 months prior to study entry and an insufficient response to first-line therapies (corticosteroids, intravenous immunoglobulin, anti-D immunoglobulin or vinca alkaloids) were enrolled in a Phase 2, interventional, single-arm study (NCT01143038). Patients had to have a single platelet count ≤30 × 10 9/L at any time during the 4-week screening period. Splenectomized patients, those who received rituximab, and those who had previously received romiplostim or other platelet-producing agents were excluded. Patients were to receive 12 months of romiplostim therapy at a starting dose of 1 µg/kg, with dose adjusted to target platelet counts ≥50-<200 ×10 9/L. Concomitant and rescue treatments were permitted to maintain platelet count. Following the 12-month treatment period, romiplostim dose was tapered for patients maintaining platelet count ≥50 × 10 9/L. Tapering could also begin earlier during the 12-month treatment period according to platelet counts and dose adjustment rules. Efficacy outcomes included the duration (expressed as a percentage of months) with platelet response (defined as median platelet count ≥50 × 10 9/L during a month without rescue medication use in the previous 8 weeks) during the 12-month treatment period; treatment-free remission (TFR; weekly platelet count ≥50 × 10 9/L for 24 consecutive weeks with no ITP treatments); time to platelet response; durable platelet response (platelet response for ≥6 of the last 8 weeks of treatment during the first 24 weeks); and sustained platelet response (platelet response for 9 weeks in any 12-week period). Safety was assessed throughout the study. Post hoc analyses were performed by subgroups of ITP duration (newly diagnosed ITP: <3 months; persistent ITP ≥3-≤12 months). Results: Of 75 patients enrolled, 45 had newly diagnosed ITP, 30 had persistent ITP. Baseline demographic are shown in the Table. Mean (standard deviation; SD) duration of platelet response was 80.1% (35.4) of months for patients with newly diagnosed ITP and 80.9% (29.9) for patients with persistent ITP (Table). Median (range) weeks to first platelet response was 2.1 (0-11) in the newly diagnosed group and 2.1 (1-25) in the persistent ITP group. In the newly diagnosed ITP group 21 (46.7%) and 31 (68.9%) patients had durable and sustained platelet response, respectively; in the persistent ITP group, 16 (53.3%) and 24 (80.0%) patients had durable and sustained platelet response, respectively. TFR was achieved by 17 (37.8%) newly diagnosed patients and 7 (23.3%) persistent ITP patients. Median (range) weeks to TFR was 21 (6-57) in the newly diagnosed group and 43 (9-53) in the persistent ITP group. The safety profile of romiplostim was broadly similar across the two groups. Conclusion: In this Phase 2 study of patients with primary ITP and disease duration of ≤6 months, romiplostim demonstrated efficacy irrespective of whether patients had newly diagnosed (<3 months duration) or persistent (≥3-≤12 months) disease. Efficacy outcomes were generally similar in both groups of patients, with rapid time to platelet response (~2 weeks) observed and around half of patients achieving a durable platelet response. Respective TFR rates were 37.8% and 23.3% in patients with newly diagnosed and persistent ITP. Safety was consistent across groups and no new safety signals were observed. This subgroup analysis was consistent with the previously reported overall findings of the study and supports the use of romiplostim for adult patients with primary ITP, including those with newly diagnosed and persistent disease. Figure 1 Figure 1. Disclosures Newland: GSK: Consultancy, Research Funding, Speakers Bureau; Novartis: Consultancy, Research Funding, Speakers Bureau; Grifols: Consultancy, Speakers Bureau; BMS: Research Funding; Argenx: Consultancy, Speakers Bureau; Angle: Consultancy; Amgen: Consultancy, Research Funding, Speakers Bureau; UCB Biosciences: Consultancy; Roche: Speakers Bureau; Octapharma: Research Funding. Viallard: LFB: Consultancy; Grifols: Consultancy; Novartis: Consultancy; Amgen: Consultancy. López Fernández: Amgen: Honoraria. Eisen: Amgen: Current Employment, Current equity holder in publicly-traded company. Saad: Amgen: Current Employment, Current equity holder in publicly-traded company. Hippenmeyer: Amgen: Current Employment, Current equity holder in publicly-traded company. Godeau: Novartis: Consultancy; Sobi: Consultancy; Amgen: Consultancy; Grifols: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document