scholarly journals Interobserver Variability with the Diagnosis of Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS) ¿Is the Threshold of 20% Bone Marrow Blasts Reproducible?

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2607-2607
Author(s):  
Patricia Font Lopez ◽  
Javier Loscertales ◽  
Montserrat Lopez Rubio ◽  
María Teresa Cedena ◽  
Carlos Soto ◽  
...  

Abstract Introduction. The boundaries between MDS and AML are still a matter of debate. In the 2001 WHO Classification, the myeloblast count distinguishing AML and MDS was lowered from 30% to 20% of the bone marrow (BM) cells or peripheral blood (PB) leukocytes. It was justified on the basis that treating patients with 20-29% BM blasts with intensive chemotherapy showed a similar outcome to those with > 30% BM blasts. However, the better knowledge of the biology of both diseases is showing that in several cases AML and high risk MDS share identical genetic profiles, as it is well known in AML with myelodysplasia- related changes (AML-MRC). Currently there are new therapeutic options, less toxic, and suitable for elderly people.The threshold of 20% BM blast is artificial, but it is still the main criterion used in clinical trials and also in real life to discriminate patients that probably belong to the spectrum of the same biological entity. Treatment of patients with MDS or AML is widely based in this relatively arbitrary condition. Objective: To study if the threshold of 20% bone marrow blasts, distinguishing MDS with excess of blasts type 2 (MDS EB 2) and AML, is reproducible among different observers. Methods. 120 bone marrow samples from patients previously diagnosed with MDS-EB-2, AML or therapy-related myeloid neoplasms (t-MN) according to 2016 WHO classification were included. The diagnosis of MDS required cytogenetics and/or FISH, and the cases with AML should have been classified following the 2017 ELN recommendations, regarding immunophenotyping, cytogenetics and molecular biology. The design of the study was established to include cases with <40% BM blasts, WBC <25x109/L and less than 20% myeloblasts in peripheral blood. The proportion of samples from each category was not predefined. Specimens were collected from 12 hospitals and were evaluated by 12 morphologists. Each observer evaluated 20 samples, and each sample was analyzed independently by two morphologists. The second observer was blinded to the clinical and laboratory data, except for the peripheral blood (PB) counts. The interobserver concordance was evaluated using the Cohen kappa test. Results. Finally 116/120 samples were considered suitable for the study. Regarding 2016 WHO categories, 55 cases showed MDS EB-2, 44 AML-MRC, 8 t-MN, 4 AML- NOS, 2 NPM1-mutated AML, 2 RUNX1-RUNX1T1 AML, 1 BCR-ABL1+ AML. Next generation sequencing was performed in 79 cases. Discordance was observed in 34/116 cases (29.3%). 14 cases with MDS-EB2 (1 NPM1+) were classified as AML-MRC by the second observer, 16 AML cases as MDS EB-2, 3 MDS EB-2 as MDS- EB1 and 1 AML as MDS- EB1. The genetic and /or molecular profile of the discordant cases was heterogeneous. Regarding the threshold of 20% BM blasts, discrepancies were 31/116 (26.7%, I Kappa test = 0.46, moderate agreement). The agreement between MDS-EB-2 and AML-MRC, with discordance in 28/98 cases (28.6%), was moderate-fair (Kappa test= 0.42). Conclusion. The threshold of 20% BM blasts did not accurately separate AML from MDS EB-2. Particularly less concordance was seen for AML-MRC. Incorporation of genetic and molecular characteristics to the morphologic diagnosis is needed to optimize the definition of both entities. Acknowledgment: Angel Cedillo, Secretaría Técnica AMHH. Disclosures Font Lopez: GILEAD: Membership on an entity's Board of Directors or advisory committees; CELGENE-BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees. Loscertales: Janssen, Abbvie, Astra-Zeneca, Beigene, Roche, Gilead: Consultancy; Janssen, Abbvie, Roche, Gilead: Speakers Bureau. Cedena: Janssen, Celgene and Abbvie: Honoraria.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 17-18
Author(s):  
Shaadi Mehr ◽  
Daniel Auclair ◽  
Mark Hamilton ◽  
Leon Rozenblit ◽  
Hearn Jay Cho ◽  
...  

Abstract: Title: Architecture of sample preparation and data governance of Immuno-genomic data collected from bone marrow and peripheral blood samples obtained from multiple myeloma patients In multiple myeloma (MM), the interactions between malignant plasma cells and the bone marrow microenvironment is crucial to fully understand tumor development, disease progression, and response to therapy. The core challenge in understanding those interactions has been the establishment of a standard process and a standard model for handling the data quality workflow and the underlying data models. Here we present the Platform (Figure 1), an integrated data flow architecture designed to create data inventory and process tracking protocols for multi-dimensional and multi-technology immune data files. This system has been designed to inventory and track peripheral blood and bone marrow samples from multiple myeloma subjects submitted for immune analysis under the MMRF Immune Atlas initiative (figure 2), and the processing and storage of Single Cell RNA-seq (scRNA-seq) and Mass Cytometry time-of-flight (CyTOF) data files derived from these immune analyses. While these methods have been previously applied on both tumor and immune populations in MM [2,3], this level of multi-institutional and multi-technology is unique. The Cloud Immune-Precision platform contains standardized protocols and bioinformatics workflows for the identification and categorization of immune cell populations and functional states based upon scRNA-seq gene signatures (ref: Bioinformatics manuscript in submission) and CyTOF protein signatures. Upon further expansion, it will contain high dimensional scRNAseq and CyTOF immune data from both bone marrow and peripheral blood samples from myeloma patients enrolled in the Multiple Myeloma Research Foundation (MMRF) CoMMpass study (NCT01454297) [1] (Figure 3). The architecture covers the automation of data governance protocols, data transformation and ETL model developments that will create an immune proteomic and profiling database and its integration into clinical and genomics databases: e.g. the MMRF CoMMpass clinical trial. This large-scale data integration will establish a cutting-edge Immune-Precision central platform supporting large scale, immune-focused advanced analytics in multiple myeloma patients. This platform will allow researchers to interrogate the relationships between immune transcriptomic and proteomic signatures and tumor genomic features, and their impact on clinical outcomes, to aid in the optimization of therapy and therapeutic sequencing. Furthermore, this platform also promotes the potential to (further) elucidate the mechanisms-of-action of approved and experimental myeloma therapies, drive biomarker discovery, and identify new targets for drug discovery. Figure 1: Cloud Immune-Precision Platform (Integrated data flow architecture designed to create data inventory and process tracking protocols for multi-dimensional and multi-technology immune data files) Figure 2: Sample tracking process architecture Figure 3: Data file creation and repository process tracking References: 1- Settino, Marzia et al. "MMRF-CoMMpass Data Integration and Analysis for Identifying Prognostic Markers." Computational Science - ICCS 2020: 20th International Conference, Amsterdam, The Netherlands, June 3-5, 2020, Proceedings, Part III vol. 12139 564-571. 22 May. 2020, doi:10.1007/978-3-030-50420-5_42 2- Ledergor, Guy et al. "Single cell dissection of plasma cell heterogeneity in symptomatic and asymptomatic myeloma." Nature medicine vol. 24,12 (2018): 1867-1876. doi:10.1038/s41591-018-0269-2 3- Hansmann, Leo et al. "Mass cytometry analysis shows that a novel memory phenotype B cell is expanded in multiple myeloma." Cancer immunology research vol. 3,6 (2015): 650-60. doi:10.1158/2326-6066.CIR-14-0236-T Figure 1 Disclosures Bhasin: Canomiiks Inc: Current equity holder in private company, Other: Co-Founder. Dhodapkar:Amgen: Membership on an entity's Board of Directors or advisory committees, Other; Celgene/BMS: Membership on an entity's Board of Directors or advisory committees, Other; Janssen: Membership on an entity's Board of Directors or advisory committees, Other; Roche/Genentech: Membership on an entity's Board of Directors or advisory committees, Other; Lava Therapeutics: Membership on an entity's Board of Directors or advisory committees, Other; Kite: Membership on an entity's Board of Directors or advisory committees, Other.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2592-2592
Author(s):  
Nicholas J. Short ◽  
Keyur Patel ◽  
Maher Albitar ◽  
Miguel Franquiz ◽  
Rashmi Kanagal-Shamanna ◽  
...  

Background: Circulating cell-free DNA (ccfDNA) is highly fragmented DNA in plasma that is released by normal or tumor cells when they undergo apoptosis or necrosis. ccfDNA allows for non-invasive sampling of somatic genomic alterations and is informative in various solid tumors, including as a marker of measurable residual disease (MRD). We sought to assess the utility of baseline assessment and tracking of leukemia-associated mutations through peripheral blood sampling of ccfDNA in patients (pts) with acute leukemias. Methods: Plasma ccfDNA was isolated and analyzed using a next-generation sequencing (NGS) assay of 275 genes. This NGS analysis is based on Single Primer Extension library preparation with unique molecular identifier (Qiagen, Germantown, MD); a sequence coverage ≥ 100X (after removing duplicates) was required. Amplicon-based NGS was also performed on DNA extracted from the bone marrow (BM) in a CLIA-certified molecular diagnostics laboratory. This BM panel detects mutations in the coding sequence of 28 leukemia-associated genes, with an analytic sensitivity of 5-10%. The ccfDNA panel included all 28 genes evaluated on the BM NGS panel (ABL1, ASXL1, BRAF, DNMT3A, EGFR, EZH2, FLT3, GATA1, GATA2, HRAS, IDH1, IDH2, IKZF1, JAK2, KIT, KRAS, MDM2, MLL, MPL, MYD88, NOTCH1, NPM1, NRAS, PTPN11, RUNX1, TET2, TP53, WT1). Established bioinformatics pipelines were used to identify somatic variants. Results: Twenty-four pts (AML, n=22; ALL, n=2) underwent paired ccfDNA and BM sequencing at diagnosis prior to receiving frontline intensive chemotherapy. For baseline samples, ccfDNA was collected a median of 6 days after BM collection (range, 0-27 days) and a median of 0.5 days after start of induction chemotherapy (range, -7 to 7 days). Eleven pts (46%) also had ccfDNA collected at ≥1 time point during remission. Among the 28 genes of interest, the median number of mutations per pt detected in BM and in ccfDNA was 1 (range, 0-4) for both assays (P=0.39). A total of 40 mutations were detected: 18 mutations (45%) were detected by both methods, 7 (18%) were detected only in ccfDNA, and 15 (38%) were detected only in BM. Time from start of chemotherapy until ccfDNA collection did not appear to impact the concordance of ccfDNA and BM mutation analysis (P=0.87). Among mutations detected by ccfDNA in baseline samples, the median variant allelic frequency (VAF) was 33.7% (range, 2.7-90.8%). Among the 18 overlapping mutations, the concordance of VAF assessment by both methods was high (R2 = 0.849). Mutations detected by only one of the two methods were generally of lower VAF than those detected by both methods, suggesting that either method may miss small subclonal populations. The median VAF of mutations (as measured in ccfDNA) that were detected by both methods was higher than those detected only in ccfDNA (39.8% vs 25.2%, respectively; P=0.04); similarly, the median VAF of mutations (as measured in BM) that were detected by both methods was higher than those detected only in BM (40.2% vs 6.6%; P=0.001). Among the 7 mutations detected only by ccfDNA, ASXL1 was detected in 2 pts, WT1 in 1 pt, IDH1 in 1 pt, and BRAF and two EGFR mutations in 1 pt. Among the 5 pts in whom mutations were detected in ccfDNA but not BM, 2 eventually relapsed. In both pts, the discordant mutation (IDH1 and ASXL1) was detected in the relapse BM, suggesting that these were true mutations that were missed by NGS of the baseline BM. ccfDNA detected leukemia-associated mutations during remission that appeared to herald overt relapse (Figure 1). Two pts with t(8;21) AML developed new RUNX1 mutations detected by ccfDNA while in remission and subsequently relapsed 3 months and 14 months later. In both of these pts, the new RUNX1 mutation was confirmed in the BM at the time of morphological relapse. Another pt with AML had persistent TP53 and TET2 mutations detected by ccfDNA 1 month after allogeneic stem cell transplant and subsequently relapsed 1 month later. Conclusions: This study demonstrates that sequencing of ccfDNA can identify prognostic or targetable mutations not detected by BM NGS. However, true mutations were missed by both ccfDNA and BM analysis, suggesting that these methodologies may be complementary in the assessment and monitoring of pts with leukemia. The use of ccfDNA as a non-invasive method to detect mutations and track MRD in AML and other leukemias should be evaluated in larger, prospective cohorts. Disclosures Short: Takeda Oncology: Consultancy, Research Funding; AstraZeneca: Consultancy; Amgen: Honoraria. Jabbour:Amgen: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding; Cyclacel LTD: Research Funding; AbbVie: Consultancy, Research Funding; Adaptive: Consultancy, Research Funding; Takeda: Consultancy, Research Funding. Garcia-Manero:Amphivena: Consultancy, Research Funding; Helsinn: Research Funding; Novartis: Research Funding; AbbVie: Research Funding; Celgene: Consultancy, Research Funding; Astex: Consultancy, Research Funding; Onconova: Research Funding; H3 Biomedicine: Research Funding; Merck: Research Funding. Kantarjian:BMS: Research Funding; Amgen: Honoraria, Research Funding; Agios: Honoraria, Research Funding; Immunogen: Research Funding; Takeda: Honoraria; Novartis: Research Funding; Ariad: Research Funding; Astex: Research Funding; Pfizer: Honoraria, Research Funding; Actinium: Honoraria, Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Research Funding; Jazz Pharma: Research Funding; Cyclacel: Research Funding; AbbVie: Honoraria, Research Funding. Ravandi:Macrogenix: Consultancy, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Xencor: Consultancy, Research Funding; Menarini Ricerche: Research Funding; Cyclacel LTD: Research Funding; Selvita: Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2353-2353
Author(s):  
Berengere Gruson ◽  
Sarah Ivanoff ◽  
Céline Berthon ◽  
Veronique Harrivel ◽  
Olivier Nibourel ◽  
...  

Abstract Background:There are few established risk factors for AML. Benzene, radiation, prior chemotherapy treatments for cancer, and cigarette smoking have been causally linked with AML. Few studies have shown that incidence rates are highest in the agricultural region and it has been hypothesized that farming and its related exposures (e.g., pesticides) may account for this increased risk. Nonetheless, there are no do data regarding clinical, cytogenetic and molecular differences between rural and urban area. The primary objective of this study is to look for cytogenetic features of rural patients. Secondary objectives are to search for clinical and molecular characteristics. Patients and methods: We conducted a retrospective analysis of 521 AML patients treated in 2 french institutions (Amiens University Hospital and Lille University Hospital) between 2008 and 2013. Patients with therapy related AML have been excluded. Patient demographics as well as clinical, hematological, cytogenetics and molecular data were collected from the Nord-pas de Calais and Picardie observatory. European Leukemia Net (ELN) classification was used for cytogenetics prognosis. We geocoded addresses using the geographic information system of the french National institute of statistics and economics (INSEE, http://www.insee.fr). According to the INSEE criteria, a location of residence was defined as rural for a city of population under 2,000 inhabitants without a contiguous built area of more than 200 m. Moreover, the population was divided into quartiles according to the percentage of farm in local companies. Results: In our population 120/521 patients (23%) lived in a rural area and 401/521 (77%) in an urban area. Sex ratio (1.6 vs 1.2, p=0.2) and median age at diagnosis (64 vs 61 years, p=0.05) were similar. There was no difference in complete blood count (hemoglobin, platelets, leukocytes and circulating blasts levels) as well as bone marrow blasts percentage, type of AML (de novo vs post MDS or MPN) and WHO classification at diagnosis between the two groups. Concerning cytogenetic anomalies, proportion of favorable and intermediate AML were similar between the two groups. However, there are more patients with unfavorable cytogenetics in rural area (41% vs 28%, p=0.04). Moreover, we observed more 11q23 anomalies in the rural patients group (22% vs 9%, p=0.04). Concerning molecular anomalies, rural patients seem to have less FLT3-ITD positivity (11% vs 19%, p=0.2), and CEBPAmutation (2% vs 7% p= 0.07) without significance. Overall survival was similar between the two groups. Same results have been obtained when we compared patients depending on the proportion of farm in their area (table 1.) with more MLL anomalies (p=0.03) and unfavorable cytogenetics (p=0.005). Conclusions:Our results showed that rural AML might present specific features. These AML would present more like therapy-related AML with more 11q23 anomalies and more unfavorable cytogenetics feature. Abstract 2353. Table 1. Characteristics of AML according to the proportion of farm in the area Proportion of farm p %≤ 0.8 n= 144 0.8<% ≤ 3.9 n=122 3.9<% ≤ 11.3 n=128 %> 11.3 n=130 Age, median (range) 63 (61-65) 62 (60-66) 61 (56-64) 63 (61-66) 0.34 Sex ratio (M/F) 1.25 1.28 1.32 1.43 0.9 Type of AML, secondary (%) 28/136 (20) 18/109 (17) 15/116 (13) 24/124 (19) 0.4 White blood cells, 109/L 6.8 (4.5-10.8) 7.8 (4.6-14.2) 7.9 (5.6-14.4) 4.5 (3.4-7) 0.08 Bone marrow blasts, % 53 (49-60) 59 (46-66) 54 (43-62) 47 (38-56) 0.2 WHO classification, (%) AML with reccurent genetic abnomalities 32/122 (30) 26/103 (25) 26/104 (27) 21/107 (20) 0.7 AML with myelodysplasia related changes 49/122 (40) 33/103 (32) 33/104 (33) 46/107 (43) 0.8 AML NOS 42/122 (34) 43/103 (42) 39/104 (37) 39/107 (36) 0.6 Cytogenetics, (%) Favorable 35/100 (35) 27/93 (29) 29/95 (31) 22/92 (24) 0.4 Intermediate 40/100 (40) 44/92 (48) 32/95 (34) 33/92 (36) 0.2 Unfavorable 25/100 (25) 21/92 (23) 33/94 (35) 37/92 (40) 0.03 MLL rearrangement, (%) 3/78 (4) 3/63 (5) 14/79 (18) 14/82 (17) 0.005 Molecular biology, (%) FLT3 ITD 18/105 (17) 18/92 (19) 21/93 (22) 7/77 (9) 0.22 NPM mutation 31/93 (33) 22/89 (25) 24/89 (27) 13/75 (17) 0.3 CEBPA mutation 7/94 (7) 3/86 (3) 5/83 (6) 4/73 (5) 0.6 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 955-955 ◽  
Author(s):  
Lisa Pleyer ◽  
Sonja Burgstaller ◽  
Reinhard Stauder ◽  
Michael Girschikofsky ◽  
Werner Linkesch ◽  
...  

Abstract Background Several studies, including retrospective analyses of patient registries1,2 and a subanalysis of the phase III MDS-AZA-001 trial3 suggest that poor-risk cytogenetics negatively impact overall survival (OS) in patients with myelodysplastic syndrome (MDS) and World Health Organization (WHO)-defined acute myeloid leukemia (AML) treated with azacitidine (AZA). There are few data available to indicate whether AZA has improved clinical activity vs conventional care in AML patients with adverse cytogenetics. However, in a subanalysis of MDS-AZA-001 (MDS and AML [20–30% bone marrow blasts]) patients with –7/–7q abnormalities had better OS with AZA than low-dose cytarabine (21.4 vs 3.5 months, respectively) supporting significant activity of AZA in patients with adverse cytogenetics.4 Methods In this retrospective study of the Austrian AZA Registry (N=346), we compared patients with WHO-AML and intermediate- (n=228) vs high-risk (n=74) cytogenetics according to Medical Research Council (MRC) criteria. Outcomes were also assessed with respect to AZA treatment line. Results The intermediate-risk cytogenetics group comprised 228 patients (AZA 1st line, n=109; AZA ≥2nd line, n=119), and the high-risk cytogenetics group comprised 74 patients (AZA 1st line, n=39; AZA ≥2nd line, n=35; Figure 1). Comparison of baseline characteristics of both groups revealed significant differences with regard to prevalence of males and Eastern Cooperative Oncology Group Performance Status (ECOG PS) >2 for patients with high-risk cytogenetics receiving AZA 1st line, but not in those receiving AZA ≥2nd line. Peripheral blood blasts were present in a significantly larger proportion of high- than intermediate-risk patients (Figure 1). In patients who received AZA 1st line, median number of AZA cycles was 6 for both the intermediate- and high-risk cytogenetic groups (range: 1–46 and 1–25, respectively). Median time from diagnosis to AZA start was <1 month for AZA 1st line and >7.6 months for AZA ≥2nd line. Median time from AZA stop to death was <2 months in all cohorts. In the whole cohort, the overall response rate (ORR) according to International Working Group (IWG) 2003 criteria5 was similar for patients with intermediate- and high-risk cytogenetics (complete response [CR] + CR with incomplete blood count recovery [CRi] + partial response [PR]: 32.0 vs 20.3%; p=0.106; Figure 1). Rates of hematologic improvement (HI) according to IWG 2006 criteria6 were also not significantly different (54.4 vs 75.6; p=0.063), and when ORR and HI were combined, the difference remained non-significant (47.4 vs 46.0%; p=0.885; Figure 1). Median OS was consistently higher in patients with intermediate- than high-risk cytogenetics (9.8 vs 5.4 months for the total cohort; p=0.046 [Figures 1 and 2a]; 13.5 vs 9.5 months for AZA 1st line [not significant]; and 7.6 vs 3.5 months for AZA ≥2nd line; p=0.005 [Figure 1]). However, median OS for responding patients (CR/CRi/PR/HI) was similar for patients with intermediate- and high-risk cytogenetics, irrespective of treatment line (19.9 vs 19.3 months for all responders; 20.5 vs 21.7 months for AZA 1st line; and 18.5 vs 15.0 months for AZA ≥2nd line). Furthermore, presence of a monosomal karyotype had a significant negative impact on OS (Figure 2b). None of the baseline factors analyzed had an impact on OS in patient subgroups with intermediate- or high-risk cytogenetics, except number of comorbidities >3. Conclusions Here, we compared outcomes of 302 WHO-AML patients with intermediate- vs high-risk cytogenetics treated with AZA. In line with recent data of MDS patients,1 baseline cytogenetics did not seem to have a significant effect on response to AZA. However, in agreement with other studies of AZA in MDS/WHO-AML patients,1–3 high-risk cytogenetics had a negative impact on survival compared with intermediate-risk cytogenetics in WHO-AML treated with AZA. 1. Sebert M, et al. Oral presentation at ASH 2013. Abstract 389 2. Thepot S, et al. Am J Hematol 2014;89:410–6 3. Fenaux P, et al. J Clin Oncol 2010;28:562–9 4. Fenaux P, et al. Br J Haematol 2010;149:244–9 5. Cheson BD, et al. J Clin Oncol 2003;21:4642–9 6. Cheson BD, et al. Blood 2006;108:419–25 Figure 1 Figure 1. Figure 2 Figure 2. Disclosures Pleyer: AOP Orphan Pharmaceuticals: Honoraria; Novartis: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Celgene: Consultancy, Honoraria. Off Label Use: Vidaza (azacitidine) is indicated for the treatment of adult AML patients who are not eligible for haematopoietic stem cell transplantation with 20–30 % blasts and multi-lineage dysplasia, according to WHO classification. This cohort also includes AML-patients with >30% bone marrow blasts.. Burgstaller:AOP Orphan Pharmaceuticals: Honoraria; Novartis: Honoraria; Mundipharma: Honoraria; Celgene: Consultancy. Stauder:Novartis: Research Funding; Ratiopharm: Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding. Girschikofsky:Pfizer: Honoraria, Research Funding; Mundipharm: Consultancy, Honoraria. Pfeilstöcker:Janssen-Cilag: Honoraria; Novartis: Consultancy, Honoraria; Celgene: Consultancy, Honoraria. Lang:Celgene: Consultancy. Sperr:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria; Phadia: Research Funding. Valent:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees. Greil:Sanofi Aventis: Honoraria; Roche: Honoraria; Pfizer: Honoraria, Research Funding; Boehringer-Ingelheim: Honoraria; Astra-Zeneca: Honoraria; Novartis: Honoraria; Genentech: Honoraria, Research Funding; Janssen-Cilag: Honoraria; Merck: Honoraria; Mundipharma: Honoraria, Research Funding; Eisai: Honoraria; Amgen: Honoraria, Research Funding; Celgene: Consultancy, Research Funding; Cephalon: Consultancy, Honoraria, Research Funding; Bristol-Myers-Squibb: Consultancy, Honoraria; GSK: Research Funding; Ratiopharm: Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2033-2033 ◽  
Author(s):  
Danielle M. Brander ◽  
Michael Y. Choi ◽  
Andrew W. Roberts ◽  
Shuo Ma ◽  
L. Leanne Lash ◽  
...  

Abstract Background: Venetoclax (VEN) is a selective, potent, orally bioavailable BCL-2 inhibitor FDA-approved for patients with del(17p) chronic lymphocytic leukemia (CLL) and who have received ≥1 prior therapy. Based on preclinical evidence of synergy, VEN plus rituximab is being assessed in an ongoing Phase 1b study. Methods: Patients with relapsed/refractory (R/R) CLL received daily VEN with stepwise ramp-up over 3-4 weeks to reach daily doses of 200-600mg. After 1 week at the target dose, monthly rituximab was added for 6 doses. Responses and progression were assessed by iwCLL criteria with CT scan and bone marrow biopsy. Bone marrow assessments were done at screening, completion of combination therapy (month 7), and 2 months after clinical/radiologic criteria of iwCLL response were met. Minimal residual disease (MRD) was assessed in peripheral blood and marrow aspirates using ≥4 color flow cytometry (min sensitivity: 0.01%). Data cutoff was 04March2016, with analysis focusing on updated safety of cytopenias experienced on the course of treatment. Results: Forty-ninepatients enrolled (48 CLL/1 SLL). Patients had received a median of 2 prior therapies (range: 1-5) and disease in 25 (51%) was considered refractory to the most recent therapy. Median time on study was 28 (<1-42) months, with 31 patients active on study. Eighteen patients discontinued: 11 due to disease progression, 3 due to toxicity (peripheral neuropathy [1], MDS [1], and death due to TLS [1]), 3 withdrew consent, and 1 was lost to follow up. Across all doses, the most common AEs of any grade were diarrhea (57%), neutropenia (55%), upper respiratory tract infection (55%), and nausea (51%). Peripheral blood cytopenias were the most common Grade 3/4 AEs (neutropenia [53%], thrombocytopenia [16%], anemia [14%], febrile neutropenia [12%], and leukopenia [12%]). Twenty-seven (55%) patients had a history of neutropenia, of whom 6 were receiving G-CSF support prior to starting VEN. Overall, in the first month of therapy, 15 (31%) experienced an AE of neutropenia (any grade). Thereafter, the rate of new AEs of neutropenia decreased over time. While there was individual patient variability, mean ANC was stable over time. Overall, 26 (53%) patients had Grade 3/4 neutropenia. Neutropenia was generally well tolerated and managed by G-CSF support in 24 patients, in addition to ≥1 dose modification in 11 of the 24 patients. Of 8 (16%) patients who experienced grade 3 infections, 2 were while neutropenic. There were no grade 4 infections. Among the 11 (22%) patients who developed any-grade thrombocytopenia, none occurred within 2 weeks of a reported bleeding-related AE. One patient had thrombocytopenia overlapping with disease progression on therapy. Objective response rate for all patients was 86% (n=42), with 51% (n=25) who had complete response (CR/CRi; 12 achieved CR/CRi by month 7). At the completion of combination therapy (month 7), 39 patients had evaluable bone marrow assessments. Thirty (77%) had no histologic evidence of CLL in the bone marrow and 22 patients (56%) had attained bone marrow MRD-negativity. In longer follow up at any point during treatment for all 49 patients, 37 (75%) patients achieved complete marrow clearance and 28 (57%) achieved marrow MRD-negativity. Conclusions: Transient manageable neutropenia was the most common AE, with first onset usually seen within the first month of treatment and the onset of new neutropenia AEs decreased over time. No patients discontinued the study due to cytopenias. Patients were able to continue on study and high rates of response to treatment were observed. VEN given with rituximab achieved rapid and profound reductions in disease burden in peripheral blood and bone marrow. 77% of evaluable patients achieved morphologic clearance by month 7, and 57% were MRD-negative at any point on study. Figure 1 Figure 1. Disclosures Brander: TG Therapeutics: Research Funding; Gilead: Honoraria. Roberts:AbbVie: Research Funding; Servier: Research Funding; Janssen: Research Funding; Genentech: Research Funding; Genentech: Patents & Royalties: Employee of Walter and Eliza Hall Institute of Medical Research which receives milestone payments related to venetoclax. Ma:Pharmacyclics, LLC, an AbbVie Company: Consultancy, Honoraria, Research Funding, Speakers Bureau; Janssen: Consultancy, Honoraria, Speakers Bureau; Gilead: Consultancy, Honoraria, Research Funding, Speakers Bureau; Genentech: Consultancy, Honoraria, Speakers Bureau; Novartis: Research Funding; Xeme: Research Funding; AbbVie: Research Funding. Lash:AbbVie: Employment. Verdugo:AbbVie: Employment, Other: may own stock. Zhu:AbbVie Inc.: Employment, Other: may own stock. Kim:AbbVie: Employment. Seymour:Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Genentech: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie Inc.: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 7-7
Author(s):  
Carlotta Welters ◽  
Meng-Tung Hsu ◽  
Christian Alexander Stein ◽  
Livius Penter ◽  
María Fernanda Lammoglia Cobo ◽  
...  

Multiple myeloma is a malignancy of monoclonal plasma cells accumulating in the bone marrow. The critical influence of tumor-infiltrating T cells on disease control and therapeutic responses has been shown in a variety of malignancies, however, the role of multiple myeloma bone marrow-infiltrating T cells is incompletely understood. Although it has been shown that multiple myeloma neo-antigen-specific T cells can be expanded in vitro, little is known about functions and specificities of clonally expanded multiple myeloma-infiltrating bone marrow T cells. Here we asked at the single cell level whether clonally expanded T cells i) were detectable in multiple myeloma bone marrow and peripheral blood, ii) showed characteristic immune phenotypes, and iii) recognized antigens selectively presented on multiple myeloma cells. A total of 6,744 single bone marrow T cells from 13 treatment-naïve patients were index-sorted and sequenced using our methodologies for determination of paired T cell receptor (TCR) αβ sequences along with immune phenotype, transcription factor and cytokine expression. Clonal T cell expansion occurred predominantly within the CD8+ compartment. Phenotypes of clonally expanded T cells were distinctive of cytolytic effector differentiation and significantly different from non-expanded CD8+ T cells. Less than 25% of expanded CD8+ T cell clones expressed the immune checkpoint molecules programmed death-1 (PD-1), cytotoxic T lymphocyte antigen-4 (CTLA-4), or T cell immunoglobulin and mucin-domain containing-3 (TIM-3), while B and T lymphocyte attenuator (BTLA) was expressed on more than half of the expanded clones. Clonal T cell expansion did not correlate with neo-antigen load as determined by whole exome and RNA sequencing of purified multiple myeloma cells. Furthermore, peripheral blood TCRβ repertoire sequencing from five selected patients with substantial bone marrow T cell expansion identified 90% of expanded bone marrow T cell clones overlapping with peripheral blood. To determine whether clonally expanded bone marrow T cells recognized antigens selectively presented on multiple myeloma cells, 71 dominant TCRs from five selected patients with substantial clonal T cell expansion were re-expressed in 58α-β- T-hybridoma reporter T cells and co-incubated with CD38-enriched multiple myeloma cells from the same patients. Only one of these TCRs recognized antigens selectively presented on multiple myeloma cells and this TCR was not neo-antigen-specific. Hypothesizing that the target antigen was a non-mutated self-antigen, we could show that this TCR also recognized the plasma cell leukemia cell line U-266 in an HLA-A*02:01-restricted manner. In summary, clonally expanded T cells in multiple myeloma bone marrow of newly diagnosed patients show cytolytic effector differentiation. In the majority of patients, clonally expanded bone marrow T cells do not recognize antigens presented on multiple myeloma cells and are not neo-antigen-specific. Our findings are relevant for the design of future therapeutics and clinical trials. The identified TCR, which recognizes a multiple myeloma antigen shared with U-266 in an HLA-A*02:01-restricted manner, could be a promising candidate for T cell therapy. Disclosures Bullinger: Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Hexal: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Menarini: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4332-4332
Author(s):  
Aziz Nazha ◽  
Rami S. Komrokji ◽  
John Barnard ◽  
Najla H Al Ali ◽  
Gail J. Roboz ◽  
...  

Abstract Background Lower-risk (LR) MDS (Low/Int-1 per International Prognostic Scoring System (IPSS)) are a heterogeneous group of disorders characterized mainly by refractory anemia and transfusion dependency. As survival of this patient (pt) population is measured in years. Goals of therapy focus on decreasing blood transfusions, improving quality of life, while minimizing treatment toxicities. While achieving complete remission (CR) in higher-risk MDS correlates with improved overall survival (OS), its relationship to OS in LR MDS is not well defined. We evaluated the impact of achieving CR on OS in LR MDS and defined the clinical characteristics that may predict for this response in this pt population. Method Included pts were diagnosed with MDS (per 2008 WHO criteria) and had LR disease with clinical and pathologic data collected from MDS Clinical Research Consortium institutions. Only pts with bone marrow blasts of 5-9% who would thus qualify both as having LR MDS and for being eligible to assess CR were included. Responses (including CR, PR, HI, stable disease and progressive disease) were defined per International Working Group 2006 criteria. OS was calculated from the time of achievement of best response to time of death or last follow-up. Cox proportional hazard analysis that included all clinical variables and treatment characteristics was used to identify independent prognostic factors. Results Of 1470 pts included in the database, 999 identified with LR disease, and 174 had bone marrow blasts of 5-9%. The median age was 60 years (range, 22-87), and 37% were female. Median neutrophil count was 1.25 X 109\L (range, .10-51.0), hemoglobin was 9.8 g/dl, platelets were 109 k/ml (range, 18-562), and bone marrow blasts were 6% (range, 5-9%). Best responses to therapy included: 26 pts (15%) with CR, 10 pts (6%) with PR, and 13 pts (7%) with HI. Among pts who achieved CR/PR/HI, 27 received HMA (25 with azacidtine +/- combination and 2 with single agent decitabine), 16 intensive chemotherapy, 2 lenalidomide, and 4 received other therapies. With a median follow up from diagnosis of 31.2 months, the median time from diagnosis to best response was 11.9 months (range, .69-81.0) and was similar in pts who achieved CR compared to PR/HI (11.5 vs. 12.4 months, respectively, p = .74). The median OS from time of CR/PR/HI for the entire cohort was 21.3 months. The median OS for pts who achieved a CR was longer compared to pts with PR/HI (46.5 vs. 18.5 months, respectively, p = .03). In multivariate analyses that included clinical variables and treatment history, achieving CR remained an independent prognostic factor for longer OS (HR .32, p = .03) but no individual demographic, clinical or treatment variables were predictive of CR. Conclusions Similar to pts with higher-risk MDS, LR MDS pts who achieve CR to therapy have improved OS compared to those with PR or HI. CR is thus an important endpoint in LR MDS, though is difficult to predict. As OS is measured in years in LR MDS, CR may be used as a surrogate endpoint for OS in clinical trials in this pt population. Disclosures Komrokji: Novartis: Consultancy, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Roboz:Cellectis: Research Funding; Agios, Amgen, Amphivena, Astex, AstraZeneca, Boehringer Ingelheim, Celator, Celgene, Genoptix, Janssen, Juno, MEI Pharma, MedImmune, Novartis, Onconova, Pfizer, Roche/Genentech, Sunesis, Teva: Consultancy. Sekeres:Millenium/Takeda: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1379-1379
Author(s):  
Gautam M. Borthakur ◽  
Tapan M. Kadia ◽  
Hind Al Azzawi ◽  
Daniella Zheleva ◽  
David Blake ◽  
...  

Cyclin dependent kinases (CDKs) are critical for cell cycle regulation and transcriptional elongation. Dysregulated CDKs have been linked to the cancer hallmarks of uncontrolled proliferation and increased survival. CYC065 is a potent and orally‐available inhibitor of CDK2 and CDK9. CDK9 regulates transcription of genes through phosphorylation of RNA polymerase II (RNAP II) C-terminal domain (CTD). Through inhibition of CDK9, CYC065 suppresses CDK9-dependent gene expression and reduces the level of MCL1, a key anti-apoptotic protein. In the first-in-human study, CYC065 was administered by 4-hour infusion every 3 weeks in patients with advanced cancers. Biomarkers related to CYC065 target inhibition, e.g. phosphorylation of RNAP II CTD Ser2, a direct substrate of CDK9, and protein levels of downstream targets, such as MCL1, were determined in patient's peripheral blood mononuclear cells (PBMCs). Durable MCL1 suppression was observed after a single dose in 11 out of 13 patients treated at the recommended phase 2 dose (RP2D) of 192 mg/m2. Five of these 13 patients achieved stable disease lasting ≥ 6 cycles (Do, KT et al, AACR Annual Meeting 2018 Abs CT037). Acute myeloid leukemia (AML) frequently relapses after initial treatment by intensive or low-intensive therapy. Drug resistance has been attributed to dysregulation in apoptotic pathways. AML cells often upregulate pro-survival members of the BCL2 family, such as BCL2 and MCL1, to avoid apoptosis (Grundy M et al, Oncotarget, 2018). Suppression of MCL1 triggered rapid apoptosis in AML, and cured AML-afflicted mice (Glaser SP et al, Genes Dev. 2012). Preclinically, CYC065 has demonstrated potent anti-tumor effect in various AML cell lines, including those with MLL rearrangements, and xenograft models (Frame S et al, AACR, 2010 Abs 3886; Frame S et al, SOHO, 2014 Abs 209). Venetoclax has modest single agent activity in AML. MCL1 dependence appeared to correlate with resistance to venetoclax (Konopleva M et al, Cancer Discovery, 2016). Preclinical study confirmed synergy of CYC065 and venetoclax, suggesting that the suppression of both BCL2 and MCL1 may be more beneficial than inhibiting either one alone (MacKay C et al. AACR-NCI-EORTC 2015 Abs B182). Based on the above rationale, a clinical study (NCT04017546) has been initiated to evaluate a combination of CYC065 with venetoclax in relapsed/refractory AML and MDS. CYC065 will be administered intravenously via 4-hour infusion on Day 1 and Day 15 in combination with daily venetoclax every 4 weeks. Initial dose escalation is 33% and then 25% upon occurrence of the first dose limiting toxicity (DLT). RP2D is the highest dose level at which less than one-third of at least 6 patients experience a DLT during the first treatment cycle. Eligible patients are ≥18 years with previously treated AML or MDS and ≥10% blasts in bone marrow or peripheral blood; adequate bone marrow, renal and liver functions are required. All patients will be asked to participate in the pharmacokinetic and pharmacodynamic studies. Plasma levels of CYC065 and its metabolites as well as venetoclax will be determined. PBMCs will be collected to assess MCL1 levels and the phosphorylation and protein levels of other downstream targets of CDK9 inhibition. Treatment will continue until progression of disease, unacceptable toxicity or changes in patient condition that renders patients ineligible for further treatment. Laboratory tests and bone marrow aspirate/biopsy will be performed to assess response according to standard criteria. Disclosures Borthakur: AbbVie: Research Funding; BioLine Rx: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Research Funding; GSK: Research Funding; Polaris: Research Funding; Tetralogic Pharmaceuticals: Research Funding; Incyte: Research Funding; Cyclacel: Research Funding; Janssen: Research Funding; Bayer Healthcare AG: Research Funding; BioTheryX: Membership on an entity's Board of Directors or advisory committees; Merck: Research Funding; Oncoceutics, Inc.: Research Funding; Strategia Therapeutics: Research Funding; PTC Therapeutics: Consultancy; BMS: Research Funding; Oncoceutics: Research Funding; Eisai: Research Funding; NKarta: Consultancy; Xbiotech USA: Research Funding; Cantargia AB: Research Funding; FTC Therapeutics: Membership on an entity's Board of Directors or advisory committees; Eli Lilly and Co.: Research Funding; Agensys: Research Funding; Argenx: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Research Funding; Arvinas: Research Funding. Kadia:BMS: Research Funding; Bioline RX: Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Research Funding; Jazz: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Research Funding; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding; Genentech: Membership on an entity's Board of Directors or advisory committees. Al Azzawi:Cyclacel LTD: Research Funding. Zheleva:Cyclacel Ltd: Employment, Equity Ownership, Patents & Royalties. Blake:Cyclacel Ltd: Employment, Equity Ownership, Patents & Royalties. Chiao:Cyclacel Ltd: Employment, Equity Ownership, Patents & Royalties.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1203-1203
Author(s):  
Andrew Sochacki ◽  
Shilin Zhao ◽  
Cosmin Adrian Bejan ◽  
Travis Spaulding ◽  
Shannon Stockton ◽  
...  

Background:Patients with clonal hematopoiesis (CH) in the absence of WHO-classified myeloid disease are of special interest given their increased prevalence with age, predisposition to morbid cardiovascular complications, and amplified risk of overt hematologic malignancy. Pts are often stratified by normal peripheral blood counts into clonal hematopoiesis of indeterminate potential (CHIP), or those with unexplained cytopenias as clonal cytopenias of undetermined significance (CCUS). However, less is known about pts with elevated counts and clonal hematopoiesis who do not fulfill WHO criteria for any myeloproliferative neoplasia (MPN). We leveraged Vanderbilt University Medical Center's unique biobank, BioVU, to identify the prevalence of JAK2V617Facross 48,000 pts to evaluate the clinical changes in progression from CH to overt myeloid disease. Methods:To develop a reference JAKV617Ftraining set, next generation sequencing via Illumina Trusight Myeloid Panel (NGS) was performed on BioVU samples (N=133) from pts with confirmed myeloproliferative malignancy. Of those pts, 78 harbored JAK2V617Fwith a range of variant allele frequencies (VAF). Matched samples in this training set (N=133) were also analyzed via Infinium® Expanded Multi-Ethnic Genotyping Array (MEGAEX). SNP array JAK2V617Fvariant intensity was extracted (rs77375493; NM_004972.3(JAK2): c.1849G>T (p.Val617Phe). A regression model was built using NGS VAF as a dependent variable and MEGAEX intensity data as independent variable (r2=0.9931).Based on this model, we imputed JAK2V617FVAF for all 48,000 pts in our cohort. Pts with JAK2V617Fwere subdivided into: clinically confirmed myeloid disease, or JAK2V617Fwithout a diagnosis of MPN. Upon review of the EMR, the latter group was further dived into: 1) probable undiagnosedMPN, 2) CHIP, 3) CCUS, or 4) CH with associated elevated peripheral blood counts (CHAPbc). Only lab values after the date of JAK2V617Fdetection were included. Confirmed malignancy was defined by WHO classification of disease. Pts with evidence of possible WHO classified PV or ET with Hgb >18.5g/dl in men, >16.5g/dl in women, or PLT count >450k/mcl regardless of gender were classified as probable undiagnosedMPN. CHIP was defined as JAK2V617Fwithout abnormal counts across a patient's EMR lifetime, except when confounding events, e.g. trauma surgery or overt iron deficiency anemia, incorrectly skewed values. CCUS was defined as JAK2V617Fin the presence of unexplained cytopenias; hemoglobin (Hgb) <13.5g/dl men or <12g/dl women, leukocyte count (WBC) <3.9x10^3/uL or platelet (PLT) <135 x10^3/mcL. We classified pts with elevated blood counts who did not meet the WHO classification of MPN [e.g. WBC >10.7 regardless of gender, Hgb 18-18.5 g/dL in men or 16.0-16.5 g/dL in women with maximum Hgb no greater than 18.5g/dl in men and 16.5g/dl in women, or PLT count between 371-450k/mcl regardless of gender (and no values >450k/mcl)] as CHAPbc. Results:We identified 410 of 48,000 pts who harbored JAK2V617F(0.85% prevalence). Of those, 270/410 had clinically diagnosed hematologic malignancy including primary myelofibrosis (PMF) (79), ET (48), PV (43) and Ph-MPN NOS (29). MDS (29), AML (15), NHL (16), plasma cell dyscrasias (5), CML (3), other (3). There were 19/410 with insufficient clinical data to determine diagnosis. The remaining 121/410 JAK2V617Fpts did not have a related diagnosis. Figure 1a demonstrates imputed VAF differences in JAK2V617Fbetween known MPN vs. CH (including undiagnosed MPN) by age (mean VAF 0.44 vs. 0.17 respectively P<0.001). We identified several undiagnosed MPNs (22), which subdivided into polycythemia predominant (1), thrombocythemia predominant (16) or a combination of polycythemia and thrombocythemia (5). The remaining 99/410 cases were CH,with CHIP (55), CCUS (29), and CHAPbc (15). Ranges of blood counts appear to be a continuous variable among JAK2V617Fpts with CH or undiagnosed MPN (Fig 1b-1d). Summary: We used an unbiased approach to identify the prevalence of JAK2V617Facross all pts at a single institution. In this cohort, hematologic malignancy and CH did stratify by imputed VAF. Further, within JAK2V617FCH, CHAPbc may be differentiated from CHIP by clinical phenotype and further investigation will be required to determine its impact on patient outcomes. Disclosures Savona: Sunesis: Research Funding; TG Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Incyte Corporation: Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Membership on an entity's Board of Directors or advisory committees; Boehringer Ingelheim: Patents & Royalties; Selvita: Membership on an entity's Board of Directors or advisory committees; Karyopharm Therapeutics: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Celgene Corporation: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1750-1750
Author(s):  
Marius E Mayerhoefer ◽  
Alexander Haug ◽  
Ulrich Jaeger ◽  
Lukas Kazianka ◽  
Verena Pichler ◽  
...  

BACKGROUND. B-cell receptor (BCR) signaling is a central driver for chronic lymphocytic leukemia (CLL), and its targeting through irreversible inhibition of Bruton's tyrosine kinase (BTK) by ibrutinib has significantly improved the prognosis of CLL patients. Ibrutinib treatment has become standard of care, and, recently, has advanced to the first-line setting. A treatment-induced, sometimes dramatic increase of peripheral lymphocytosis, has emerged as a class effect of BTK inhibitors in CLL. Mechanistically, BTK blocking by ibrutinib might affect adhesion molecules and chemokine receptors, such as CXCR4 and CXCR5, thus interfering with the protective tissue microenvironment and mobilizing tissue-resident CLL cells from the lymph nodes and bone marrow into the peripheral blood. This hypostasized mechanism of a "compartment shift," however, has not yet been demonstrated experimentally or visually. Positron emission tomography (PET) with [68Ga]Pentixafor, a radiotracer that specifically targets the CXCR4 receptor, was recently established as a sensitive approach with which to detect CLL in vivo. As a proof-of-concept, we here present three CLL patients to demonstrate the potential of [68Ga]Pentixafor-PET/MR imaging to functionally track CLL cells along the redistribution induced by ibrutinib. METHODS. Three CLL patients were included: patient 1 was a 74-year-old treatment-naïve male, with unmutated IGHV status and high-risk cytogenetics, including del17p13 and TP-53 mutation; patient 2 was a 53-year-old female with late relapse (12 years after allogeneic stem cell transplantation), IGHV was unmutated, and cytogenetic abnormalities included del11q22 and del13q14; and patient 3 was a 59-year-old treatment-naïve male with unmutated IGHV and del11q22 and del13q14. Whole-body PET/MRI with injection of 150 MBq of [68Ga]Pentixafor was performed pre and on ibrutinib treatment (patient 1: one week; patient 2: two weeks; and patient 3: three weeks after start of the first therapy cycle, respectively). Treatment consisted of continuous oral administration of 420 mg of ibrutinib. Mean standardized [68Ga]Pentixafor uptake values (SUVmean) of involved lymph nodes, the bone marrow, and the spleen were measured. Isolated peripheral blood mononuclear cells (PBMCs) were stained with fluorescence-labeled antibodies, and measurements were performed on flow cytometer. RESULTS. In all three cases - at one week, two weeks, and three weeks on ibrutinib treatment - CXCR4 density, as measured on [68Ga]Pentixafor-PET, shifted from the bone marrow and lymph nodes toward the spleen (Figs. 1-3). In patient 1, the SUVmean decreased in the bone marrow (-9.8%) and lymph nodes (-12.0%), whereas it increased markedly in the spleen (+29.8%). At flow cytometry, this patient exhibited an increase of CXCR4-high (tissue-resident) CLL cells upon ibrutinib treatment (+56.7% relative to baseline). In patient 2, the SUVmean decreased in the bone marrow (-14.7%) and lymph nodes (-27.3%), whereas it more than doubled in the spleen (+133.0%). At flow cytometry, individual CLL cells demonstrated a relative increase (+39.1%) of CXCR4 positivity. In patients 3, the SUVmean decreased in the bone marrow (-27.6% %) and lymph nodes (-41.9%), whereas it increased markedly in the spleen (+26.1%). At flow cytometry, the relative increase of CXCR4-high CLL cells was +58.3%. CONCLUSIONS. We here provide the first pictures of the early functional treatment effects of ibrutinib. While our analyses confirmed a shift of CXCR4 positive CLL cells from lymph nodes to peripheral blood, they also revealed that ibrutinib rapidly released CLL cells from the bone marrow. Also, unexpectedly, CLL cells redistributed to the orthotopic splenic cavernous system. Visualization of CLL on ibrutinib supports the pre-existing clinical hypothesis of a "compartment shift", however it also modified and refined the mechanistic model by describing early clearing of the bone marrow and re-distribution to the peripheral blood and the spleen. Disclosures Jaeger: Celgene, Roche, Janssen, Gilead, Novartis, MSD, AbbVie, Sanofi: Membership on an entity's Board of Directors or advisory committees; Novartis, Roche, Sandoz: Consultancy; AbbVie, Celgene, Gilead, Novartis, Roche, Takeda Millennium: Research Funding; Amgen, AbbVie, Celgene, Eisai, Gilead, Janssen, Novartis, Roche, Takeda Millennium, MSD, BMS, Sanofi: Honoraria. Wester:Scintomics: Other: Spouse CEO of Company; CXCR4-targeted radiopharmaceuticals: Other: Inventor; Scintomics GmbH, Germany: Other: Shareholder. Staber:AbbVie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; MSD: Honoraria, Speakers Bureau; Takeda-Millenium: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Janssen: Honoraria, Speakers Bureau; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.


Sign in / Sign up

Export Citation Format

Share Document