scholarly journals Interpretation and Application of the International Myeloma Working Group (IMWG) Criteria: Proposal for Uniform Assessment and Reporting in Clinical Trials Based on the First Study Independent Response Adjudication Committee (IRAC) Experience

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3460-3460 ◽  
Author(s):  
Joan Blade ◽  
Stefan Knop ◽  
Adam D. Cohen ◽  
Jatin J. Shah ◽  
Ralph M. Meyer

Abstract Background: In multiple myeloma (MM), reproducible criteria of disease response and progression are critical to ensuring consistency in trial analysis and reporting. Regulatory Agencies responsible for drug approval often require clinical trials use objective endpoints that are evaluated by Independent Response Adjudication Committees (IRACs). The International Myeloma Working Group (IMWG) has developed objective criteria to define disease evaluability, response, and progression (Durie, Leukemia 2006). However, there are scenarios were IMWG criteria are ambiguous, potentially leading to inconsistency amongst IRAC members or between different IRACs when interpreting response data. To address these practical issues, we developed rules for applying IMWG response criteria to the FIRST trial, the largest study in newly-diagnosed MM (Facon, Blood 2013). Patients and Methods: FIRST is a pivotal phase III trial for previously untreated patients with MM not eligible for ASCT that enrolled 1623 patients; the primary endpoint was progression-free survival (PFS). At 12 in-person meetings between 2010-2013, the IRAC assessed eligibility, evaluability and response status of all patients after each cycle until PD or study discontinuation. These evaluations were used in the trial’s primary analysis. Response was based on central laboratory values and assessed using IMWG criteria. For circumstances where IMWG criteria were ambiguous, rules were developed through unanimous consensus of IRAC members and then applied uniformly throughout the study. Results: Rules addressing identified issues on evaluability, response and progressive disease are shown in tables 1-3. Common situations posing a need for rules concerned to measurability, missing laboratory values, timing of BM exam to assess CR, discrepancies between screening and baseline lab values or measurements in the size of extramedullary plasmacytomas Conclusions: These recommendations provide explicit descriptions of response assessment of the FIRST trial, can be used for a more uniform evaluation and reporting in future clinical studies and can assist investigators’ adherence to clinical trial requirements. Table 1. Rules for Use of Data for Evaluation Issue Recommendation Light chain (Bence-Jones) myeloma with “non-measurable” serum light chain Use only 24 hour urine M-spike value for response evaluation, except for complete response (CR) IgG, IgA or IgD myeloma with “non-measurable” serum M-spike values and measurable urine M-spike Use only urine values for response evaluation except for CR or PD Disease with “measurable” values at screening but “non-measurable” at baseline (cycle 1, day 1) All assessments not meeting CR or PD should be “non-evaluable (NE)” Missing data for 2 or more consecutive cycles Consider “NE” for the specific missing cycle assessments M-spike reported as “too small to quantitate” in responding patient Assign value of 0 to allow subsequent calculation of absolute increase to determine PD Plasmacytoma given prior radiation therapy or located only in bone Not used for response assessment, except for potential PD Table 2. Rules for Response Assessment Issue Recommendation Absence of 2 consecutive negative IFE and simultaneous <5% BMPCs CR not assigned, assess as VGPR Extramedullary plasmacytomas (EMPs) - Visits until first EMP assessment Assess as NE - Two consecutive missing EMP assessments Assess as NE - EMPs not assessed as per protocol Assess as NE (consider a sensitivity analysis (ignoring EMPs)) - Patients in serologic VGPR, with ³ 50% decrease in EMP, but still present Assess as PR, until EMPs have disappeared Table 3. Rules for Determining Progressive Disease Issue Recommendation Increase in a previously existing EMP or bone lesion as only source of PD Request verification of radiologist reports before PD is assigned Initiation of a new antimyeloma therapy before documented PD Censor at the time of last assessment before starting the new therapy PD only based on the BMPCs Determine reason for BM exam (anemia? bone pain?) before assigning PD Radiation therapy not for pre-planned reasons Assess as PD PD based on M-protein measurements with no confirmation Censor unless that PD is considered unequivocal by unanimous agreement of IRAC Disclosures Blade: Janssen: Honoraria, Research Funding; Celgene: Honoraria, Research Funding. Knop:Celgene: Honoraria. Cohen:Celgene: Honoraria. Shah:Onyx Pharmaceuticals: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; Millennium Pharmaceuticals: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Array: Consultancy, Research Funding. Meyer:Celgene: Honoraria.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 22-23
Author(s):  
Abdullah S. Al Saleh ◽  
Iuliana Vaxman ◽  
Harsh Parmar ◽  
M Hasib Sidiqi ◽  
Eli Muchtar ◽  
...  

Introduction: The bone marrow plasma cell (BMPC) clone burden is typically low in light chain (AL) amyloidosis and some patients do not have a detectable serum monoclonal spike (M-spike). Increased BMPC% and serum free light chain (FLC) are associated with poorer outcomes. However, the outcomes of patients with AL amyloidosis based on the presence or an absence of a measurable serum M-spike before autologous stem cell transplantation (ASCT) has not been explored. Methods: This was a retrospective review of patients who had a diagnosis of AL amyloidosis and received ASCT between March 1996 and September 2017. The serum M-spike was recorded before ASCT and patients were divided according to the presence or absence of a measurable serum M-spike. Progression-free survival (PFS) was defined as time from ASCT to disease progression, relapse or death of any cause. Overall survival (OS) was calculated from time of ASCT to death of any cause. Univariate and multivariate analysis for PFS and OS were done using the following variables: age&gt;65 vs. ≤65 years, Mayo 2012 stage 3/4 vs. 1/2, BMPC ≥ 10% vs. &lt;10%, organs involved &gt;2 vs. ≤2, melphalan conditioning 200mg/m2 vs. 140 mg/m2 ,ASCT year &gt;2010 vs. ≤2010, and presence vs. absence of a measurable serum M-spike. Results: Seven-hundred and sixteen patients were identified and 521 (73%) had a measurable serum M-spike. Patients who had a measurable serum M-spike were more likely to have BMPC≥ 10% than patients without a measurable serum M-spike (46% vs. 34%, P=0.002) and they were more likely to have a difference in FLC ≥18 mg/dl (47% vs. 29%, P=0.0001). Overall, PFS and OS were significantly shorter in patients who had a measurable serum M-spike compared to patients without a measurable serum M-spike (Figure 1 A,B). The shorter survival was irrespective of the administration of induction therapy before ASCT. We also evaluated the difference in survival in patients with Mayo 2012 stage 3/4 based on the presence or absence of a measurable serum M-spike. The PFS and OS were also significantly different in these patients (Figure 1 C,D). Predictors for PFS included Mayo 2012 stage 3/4 vs. 1/2 (hazard ratio (HR): 1.5, P=0.003), BMPC ≥ 10% vs. &lt;10% (HR: 1.4,P=0.004), melphalan conditioning 200mg/m2 vs. 140 mg/m2 (HR:0.6, P=0.001), ASCT year &gt;2010 vs. ≤2010 (HR: 0.8, P=0.03), and the presence vs. absence of a measurable serum M-spike (HR: 1.82, P&lt;0.0001). For OS, Mayo 2012 stage 3/4 vs. 1/2 (HR: 2, P&lt;0.0001), melphalan conditioning 200mg/m2 vs. 140 mg/m2 (HR:0.4, P&lt;0.0001), ASCT year &gt;2010 vs. ≤2010 (HR: 0.6, P=0.002), and the presence vs. absence of a measurable serum M-spike (HR:1.9, P=0.003) were predictive. Conclusion: The presence of a measurable serum M-spike before ASCT is a negative independent predictor for PFS and OS in AL amyloidosis. Figure 1 Disclosures Sidiqi: Amgen: Honoraria; Celgene: Honoraria, Other: Travel grant; Janssen: Honoraria. Dingli:Karyopharm Therapeutics: Research Funding; Apellis: Consultancy; Alexion: Consultancy; Janssen: Consultancy; Bristol Myers Squibb: Research Funding; Rigel: Consultancy; Millenium: Consultancy; Sanofi-Genzyme: Consultancy. Kapoor:Amgen: Research Funding; Celgene: Honoraria; Cellectar: Consultancy; Janssen: Research Funding; Takeda: Honoraria, Research Funding; Sanofi: Consultancy, Research Funding; GlaxoSmithKline: Research Funding. Dispenzieri:Intellia: Research Funding; Alnylam: Research Funding; Pfizer: Research Funding; Takeda: Research Funding; Celgene: Research Funding; Janssen: Research Funding. Kumar:Celgene/BMS: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Janssen Oncology: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; AbbVie: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Takeda: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Tenebio: Other, Research Funding; Genecentrix: Consultancy; MedImmune: Research Funding; Cellectar: Other; Dr. Reddy's Laboratories: Honoraria; Merck: Consultancy, Research Funding; Amgen: Consultancy, Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments, Research Funding; Novartis: Research Funding; Adaptive Biotechnologies: Consultancy; Kite Pharma: Consultancy, Research Funding; Oncopeptides: Consultancy, Other: Independent Review Committee; IRC member; Karyopharm: Consultancy; BMS: Consultancy, Research Funding; Carsgen: Other, Research Funding; Genentech/Roche: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Sanofi: Research Funding. Gertz:Proclara: Other; Abbvie: Other; DAVA oncology: Speakers Bureau; Aurora Bio: Other; Prothena: Other: personal fee; Teva: Speakers Bureau; Physicians Education Resource: Other: personal fee; Annexon: Other: personal fee; Ionis/Akcea: Other: personal fee; Sanofi: Other; Appellis: Other: personal fee; Amgen: Other: personal fee; Janssen: Other: personal fee; Spectrum: Other: personal fee, Research Funding; Medscape: Other: personal fee, Speakers Bureau; Research to Practice: Other; Alnylam: Other: personal fee; Johnson and Johnson: Speakers Bureau; Springer Publishing: Patents & Royalties; Celgene: Other.


BJR|Open ◽  
2019 ◽  
Vol 1 (1) ◽  
pp. 20180035 ◽  
Author(s):  
Yevgeniy Vinogradskiy

A form of lung function imaging is emerging that uses phase-resolved four-dimensional CT (4DCT or breath-hold CT) images along with image processing techniques to generate lung function maps that provide a surrogate of lung ventilation. CT-based ventilation (referred to as CT-ventilation) research has gained momentum in Radiation Oncology because many lung cancer patients undergo four-dimensional CT simulation as part of the standard treatment planning process. Therefore, generating CT-ventilation images provides functional information without burdening the patient with an extra imaging procedure. CT-ventilation has progressed from an image processing calculation methodology, to validation efforts, to retrospective demonstration of clinical utility in Radiation Oncology. In particular, CT-ventilation has been proposed for two main clinical applications: functional avoidance radiation therapy and thoracic dose–response assessment. The idea of functional avoidance radiation therapy is to preferentially spare functional portions of the lung (as measured by CT-ventilation) during radiation therapy with the hypothesis that reducing dose to functional portions of the lung will lead to reduced rates of radiation-related thoracic toxicity. The idea of imaging-based dose–response assessment is to evaluate pre- to post-treatment CT-ventilation-based imaging changes. The hypothesis is that early, imaging-change-based response can be an early predictor of subsequent thoracic toxicity. Based on the retrospective evidence, the clinical applications of CT-ventilation have progressed from the retrospective setting to on-going prospective clinical trials. This review will cover basic CT-ventilation calculation methodologies, validation efforts, presentation of clinical applications, summarize on-going clinical trials, review potential uncertainties and shortcomings of CT-ventilation, and discuss future directions of CT-ventilation research.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1666-1666
Author(s):  
Pinkal Desai ◽  
Niamh Savage ◽  
Spencer Krichevsky ◽  
Tania Curcio ◽  
Sangmin Lee ◽  
...  

Introduction: Philadelphia negative myeloproliferative neoplasms (Ph- MPN) are hematopoietic stem cell malignancies associated with poor median survival of 12.4 months. They are often excluded from clinical trials because there are no accepted standards for treatment or assessment of disease response. SGI-110 (guadecitabine) is a second-generation DNA hypomethylating agent (HMA) that is currently in clinical trials for the treatment of myelodysplastic syndrome and acute myeloid leukemia. Guadecitabine was designed to resist degradation by protein aminases and prolong the exposure of tumor cells to the active metabolite decitabine. The purpose of this study was to test the efficacy and safety of SGI-110 in Philadelphia chromosome negative MPNs (Ph- MPN) and to also test the clinical applicability of the International IWG MDS/MPN response criteria in a prospective trial1. Methods: This is an interim analysis of an open label single-arm, single-institution study to evaluate the efficacy and safety of SGI-110 in Philadelphia chromosome negative (Ph-) myeloproliferative Neoplasms as classified by WHO, including chronic neutrophilic leukemia (CNL), atypical chronic myeloid leukemia (aCML), chronic myelomonocytic leukemia (CMML), myelodysplastic/myeloproliferative neoplasm unclassifiable, accelerated phase myelofibrosis and MPN unclassifiable (defined as peripheral and or bone marrow blasts of 10-19%). PV, ET and primary/secondary myelofibrosis were excluded. Patients were required to complete at least 3 cycles of guadecitabine to be considered evaluable for efficacy. Safety analyses were done on all patients who received any treatment with guadecitabine. Guadecitabine was administered subcutaneously at a dose of 60mg/m2 on days 1-5 repeated every 28 days. The IWG MDS/MPN response classification was used to assess treatment response. Results: Baseline characteristics of the study participants are presented in Table 1. Among the 20 treated patients, 2 (10.0%) were treated with previous HMAs, 3 had progressive disease, 1 transferred care, 7 were not yet evaluable for response, and 1 died after receiving only 2 cycles of treatment. Of the 13 evaluable, protocol specific response was seen in 8 (61.5%) patients: 2 (15.4%) achieved complete remission (CR), 3 (23.1%) with optimal marrow response (OMR), 3 (23.1%) with hematological response/clinical benefit (CB). Stable disease was seen in 4 patients (30.8%). Of the 7 patients that were inevaluable: 3 had progressive disease before completing 3 cycles, 2 received <3 cycles of therapy, 1 discontinued treatment due to personal choice, and 1 patient died from infection after receiving 2 cycles of treatment. The median overall survival (OS) for all evaluable patients was 27.4 months with 25.8 months for responders. Median OS for patients who achieved CR was 27.4 months and 25.0 months for OMR. For patients with CB, mean survival was 21.0 months. There was 1 patient with stable disease with prolonged survival (21 cycles), which elevated the mean survival to 26.0 months for the SD category. The median number of cycles to achieve a response was 3. The median times to first and best response were 3.6 and 3.8 months, respectively. The combination of ASXL1 and EZH2 mutations was associated with rapid progression. The most common AEs and SAEs related to guadecitabine are listed in Tables 2 and 3 respectively. Conclusion: SGI-110 was safe and well tolerated in patients with Ph negative MPN, with encouraging efficacy in this difficult-to-treat patient population. Further investigation of this agent in MDS/MPN overlap syndromes is warranted, and the present trial is ongoing. 1. Savona MR, Malcovati L, Komrokji R, et al. An international consortium proposal of uniform response criteria for myelodysplastic/myeloproliferative neoplasms (MDS/MPN) in adults. Blood. Mar 19 2015;125(12):1857-1865. Disclosures Desai: Cellerant: Consultancy; Astex: Research Funding; Astellas: Honoraria; Sanofi: Consultancy; Celgene: Consultancy. Lee:Helsinn: Consultancy; Jazz Pharmaceuticals, Inc: Consultancy; Roche Molecular Systems: Consultancy; AstraZeneca Pharmaceuticals: Consultancy; Karyopharm Therapeutics: Consultancy; Ai Therapeutics: Research Funding. Ritchie:Celgene, Incyte, Novartis, Pfizer: Consultancy; Ariad, Celgene, Incyte, Novartis: Speakers Bureau; AStella, Bristol-Myers Squibb, Novartis, NS Pharma, Pfizer: Research Funding; Celgene, Novartis: Other: travel support; Jazz Pharmaceuticals: Research Funding; Celgene: Other: Advisory board; Pfizer: Other: Advisory board, travel support; agios: Other: Advisory board; Tolero: Other: Advisory board; Genentech: Other: Advisory board. Roboz:Trovagene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees; Sandoz: Consultancy, Membership on an entity's Board of Directors or advisory committees; Roche/Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astex: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celltrion: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees; Eisai: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; MEI Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; Orsenix: Consultancy, Membership on an entity's Board of Directors or advisory committees; Otsuka: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Agios: Consultancy, Membership on an entity's Board of Directors or advisory committees; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Actinium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amphivena: Consultancy, Membership on an entity's Board of Directors or advisory committees; Argenx: Consultancy, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2063-2063 ◽  
Author(s):  
Helene Caillon ◽  
Alix Irimia ◽  
Jason S. Simon ◽  
Amy Axel ◽  
Kate Sasser ◽  
...  

Abstract Background : Detection and quantification of monoclonal component (M-spike) by serum protein electrophoresis (SPE) and immunofixation (IFE) are essential for response evaluation in multiple myeloma (MM) according to the International Myeloma Working Group (IMWG) criteria. Recent clinical trials on daratumumab, an IgG Kappa anti-CD38 monoclonal antibody, have shown impressive results with deep responses. However daratumumab may be detected on serum protein electrophoresis (SPE) and immunofixation (IFE) assays used for monitoring disease monoclonal immunoglobulins (M protein). This can lead to false positive SPE and IFE assay results for patients with IgG kappa myeloma protein impacting initial assessment of complete responses (CR) by International Myeloma Working Group (IMWG) criteria. Differentiating therapeutic monoclonal antibodies, such as daratumumab, from endogenous monoclonal protein can be challenging when both molecules co-migrate or migrate closely on electrophoresis. The availability of a specific, anti-daratumumab antibody has provided the opportunity to overcome this interference and to correctly assess biochemical response. Indeed, Mc Cudden and al. in collaboration with Janssen developed a technique, the Daratumumab Interference Reflex Assay (DIRA) test, which has been utilised in daratumumab clinical trials. Given the need for a commercially available automated and standardized test, we evaluated a new commercial DIRA kit test being developed by Sebia (Lisses, France): the Hydrashift 2/4 daratumumab. Objective: The aim of this study is to evaluate the Hydrashift 2/4 daratumumab in comparison with our laboratory developed DIRA test for the displacement of daratumumab on IFE. Design and methods: The Hydrashift 2/4 daratumumab assay was prepared by Sebia using the anti-daratumumab antibody produced by Janssen and modified to allow a migration of daratumumab/anti-daratumumab complexes toward the α-globulin fraction on IFE. IFE technical procedures, migration, and staining programs were performed according to the manufacturer instructions, and run on the standard Sebia, Hydrasys plateform, with the HYDRAGEL 4 IF kit. In addition to the regular procedure, an additional applicator to apply the anti-daratumumab antibody was used. Analytical performances including sensitivity, specificity and comparisons with the original DIRA test were assessed on 31 samples from ongoing daratumumab clinical trials. Results: Serum samples from 309/324 (95.4%) patients assessed demonstrated a positive IFE at diagnosis. In 119/309 (38,5%) of cases, the M-spike partially or totally co-migrated with daratumumab detected in serum. Of these, MM cases displayed an isotype other than IgG Kappa or Kappa light chains did not require a DIRA test during follow-up for response assessment as a standard IFE could clearly show if initial monoclonal component was still detectable or not. From our experience, an anti-daratumumab displacement assay was only required for IgG Kappa MM or Kappa Light Chain MM (LCMM) when standard IFE could not distinguish daratumumab from endogenous M-spike. This situation represented 66 cases (21,4 %, i.e. 66 on 309). The Hydrashift 2/4 daratumumab assay showed excellent concordance (100%) with the laboratory developed test on the 31 samples tested (i.e. 17 negative DIRA, 10 positive DIRA and 4 doubtful DIRA). Daratumumab/anti-daratumumab complexes were detected in the α-globulin fraction with a sensibility of 200 mg/L. Daratumumab/anti-daratumumab complex was difficult to visualize when daratumumab concentrations were less than 200 mg/L but daratumumab was shown to be completely removed from the gamma globulin fraction with no trace left for all tested patients. For 48 samples tested on diagnosis, the anti-daratumumab shifted specifically the daratumumab with no effect on the patients' M-spike (100% specificity). Conclusion: With the growing application of monoclonal antibodies such as daratumumab in the treatment of multiple myeloma, the development of validated, widely available assays to overcome antibody interference will become increasingly important. The Hydrashift 2/4 daratumumab test provides the opportunity to automate and standardize the displacement of daratumumab interference and help with the correct interpretation of IFE results for clinical outcome measures. Disclosures Simon: Janssen: Employment. Axel:Janssen Pharmaceuticals Research and Development: Employment. Sasser:Johnson & Johnson: Equity Ownership; Janssen Pharmaceuticals R&D: Employment. Scullion:Janssen: Employment. Ligneel:Sebia: Employment. Nouadje:Sebia: Employment. Moreau:Bristol-Myers Squibb: Honoraria; Amgen: Honoraria; Janssen: Honoraria, Speakers Bureau; Novartis: Honoraria; Takeda: Honoraria; Celgene: Honoraria.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5076-5076 ◽  
Author(s):  
Salomon Manier ◽  
Remy Dulery ◽  
Alain Duhamel ◽  
Eileen Boyle ◽  
Julien Rossignol ◽  
...  

Abstract Abstract 5076 Background. Waldenstrom macroglobulinemia (WM) is a low grade B cell lymphoma characterized by bone marrow infiltration of lymphoplasmacytic tumor cells that secrete monoclonal IgM (M-protein) into the serum. Measurement of the serum M-protein [using serum protein electrophoresis (SPEP)] and measurement of total IgM (using nephelometry) are used to diagnose and monitor WM. There are, however, many limitations with these techniques and new markers are needed. IgG and IgA Hevylite® immunoassays have been reported to be more sensitive than SPEP and nephelometry for identifying monoclonal immunoglobulins in multiple myeloma and unlike immunofixation, provide quantitative information. We hypothesized that serum IgM Hevylite assays (specifically measuring IgMkappa and IgMlambda, separately) would accurately identify serum IgM M-proteins. We also evaluated the association between known tumor burden markers and prognostic factors with IgM Hevylite results in patients with WM. Method. We retrospectively measured IgMkappa and IgMlambda in sera from 59 WM patients: 44 patients were at diagnosis and 15 had relapsed disease. The diagnosis of WM was made according to the current guidelines. All serum samples were kept frozen in the Lille serum bank since collection. All patients gave informed consent prior to the collection and none were treated at time of collection of the serum. Approval of this protocol was obtained from the CHRU Lille and was in accordance with the Declaration of Helsinki. Hevylite measurements were made at The Binding Site Ltd, Birmingham, UK. A normal range was produced from normal (blood donor) sera (n=120), median (and 95%ile ranges) were; IgMkappa 0.634g/L (0.29-1.82), IgMlambda 0.42g/L (0.17-0.94), IgMkappa/IgMlambda ratio 1.6 (0.95-2.3). For ease of comparison IgM hevylite ratios were expressed as the involved monoclonal immunoglobulin/uninvolved polyclonal immunoglobulin (IgMi). To describe the distribution of IgMi Hevylite levels in patients with WM, the median and range (min-max) were reported. Median values were compared using the Wilcoxon rank-sum test and ANOVA. Fisher's exact test was used to compare proportions. All statistical tests were two-sided. All analyses were conducted using SPSSv12 software. Results. The baseline characteristics of the patients were as follows: the median (range) age was 68 years (41-86), male/female 38/21, serum b2M 3.0mg/L (1.2-9.0), hemoglobin 11.8g/dL (7.6-15.4), platelet count 267 ×109/mm3 (55-741), serum M-spike 19g/L (3.0-52.7). In our series, 18 (31%), 22 (37%) and 19 (32%) patients had low, intermediate and high risk disease respectively, in the WM-IPSS scoring system. The median (min-max) IgMkappa ratio was 134 (8.7-2850) and IgMlambda ratio was 0.03 (0.0007-0.39). IgMi Hevylite ratio was 98.07 (2.59-2850). The IgMi Hevylite correlated well with the M-spike measured using SPEP (r=0.601, p<0.0001). In our study, high IgMi Hevylite levels correlated well with markers of high tumor burden and of poor prognosis. The median (range) IgMi Hevylite level was higher in patients with hemoglobin <10g/dL versus ≥10 g/dL, 267 (8.1-1722) and 76 (2.6-2850) respectively (p=0.013). The median (range) IgMi Hevylite ratio level was significantly (p=0.033) higher in the high risk IPSS group 208 (2.6-2850) than in the intermediate 75 (15-1033) and low risk groups, 98 (8-571). The IgMi Hevylite levels also separated WM patients with progressive disease who required therapy. Twenty seven pts were symptomatic and required specific treatment for WM, and 32 pts were left untreated. The median IgMi Hevylite ratio was significantly higher in progressing patients, 210 (8.1-2850) and 60 (2.6-571) in the 2 groups, respectively (p=0.014). Conclusion. In this study we demonstrated that IgM Hevylite measurement is a new and reliable marker for monitoring WM disease. It is related to poor prognostic markers that separate WM patients with progressive disease who require therapy. We are currently expanding the cohort to confirm these observations. These findings have implications in the management of patients with WM. Disclosures: Leblond: Roche: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Mundipharma: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Genzyme: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Leleu:Celgene: Consultancy, Research Funding; Janssen Cilag: Consultancy, Research Funding; Leo Pharma: Consultancy; Amgen: Consultancy; Chugai: Research Funding; Roche: Consultancy, Research Funding; Novartis: Consultancy, Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3087-3087 ◽  
Author(s):  
Stephen J. Schuster ◽  
Jakub Svoboda ◽  
Sunita Dwivedy Nasta ◽  
David L. Porter ◽  
Elise A. Chong ◽  
...  

Abstract BACKGROUND: Autologous T cells genetically modified to express a chimeric antigen receptor consisting of an external anti-CD19 single chain antibody domain with CD3ζ and 4-1BB signaling domains (CTL019 cells) can mediate potent anti-tumor effects in patients (pts) with relapsed or refractory chronic lymphocytic and acute lymphoblastic leukemias. We are conducting a phase IIa clinical trial to evaluate the safety and efficacy of CTL019 cells in pts with relapsed or refractory CD19+ non-Hodgkin lymphomas (NHL). METHODS: 30 evaluable pts are planned for analysis, including at least 8 with follicular lymphoma (FL), 8 with diffuse large B cell lymphoma (DLBCL), and 8 with mantle cell lymphoma. Eligible pts have CD19+ NHL with no available curative treatment options, a limited prognosis of several months to <2 years anticipated survival, and responsive or stable disease with most recent therapy. Pts with FL have progression of lymphoma within 2 years after second or higher line of therapy (not including single agent monoclonal antibody therapy); DLBCL pts have residual disease after primary therapy and are not eligible for autologous stem cell transplant (ASCT) or have relapsed or residual disease after ASCT. After steady state apheresis to collect peripheral blood leukocytes, pts receive lymphodepleting chemotherapy based on disease burden, histology, and past therapies. One to 4 days after chemotherapy, pts receive a single dose of CTL019 cells by intravenous infusion; total CTL019 target dose is 5 x 108 cells. Peripheral blood and marrow samples are collected for immunophenotypic, cytokine, and molecular studies at pre-specified times after T cell infusion. Initial tumor response assessment is performed 3 months after T cell infusion using International Working Group response criteria. Enrollment started in February 2014; data reported here are through July 30, 2014. RESULTS: To date, 23 pts (DLBCL 16; FL 7) have enrolled. The median age is 56 years (range: 25-77), male: female ratio is 14:9, median number of prior therapies is 4 (range: 1-8), and number of pts with prior ASCT is 9 (39%). Ann Arbor stages at enrollment are: stage IV 11 pts (48%); stage III 5 pts (22%); stage II 5 pts (22%); stage IE 2 pts (8%); 5 pts (22%) had bone marrow involvement. LDH was increased in 17 pts (74%). Three pts (DLBCL 2 pts; FL 1 pt) were removed from the trial before therapy due to progressive disease. As of July 30, 2014, 14 pts have received CTL019 T cell infusions. Pre-infusion chemotherapy regimens were EPOCH (1 pt); cyclophosphamide (7 pt); bendamustine (5 pts); cyclophosphamide-fludarabine (1 pt). Median CTL019 T cell dose is 5.8 x 106 cells / kg (range: 3.7 – 8.9 x 106). In vivo expansion of CTL019 cells was brisk; the median peak CTL019% of CD3+ cells in peripheral blood was 6.1% (range: 0.7-43.1%) for all patients, 17.3% (range: 3.9-43.1) for responders, and 4.95% (range: 0.7-7.3) for non-responders. Peak CTL019 cell expansion generally occurred around 7 days after T cell infusion. All patients developed fever following T cell infusion, attributed to cytokine release syndrome (CRS). Severity of CRS according to our novel grading scale (reported separately) was: 12 pts grade 2; 1 pt grade 3; 1 pt grade 4. One pt received steroids and tocilizumab for grade 4 CRS. CRS occurred within the first week of T cell infusion in all pts. Neurologic toxicity was observed in 2 pts (1 pt with grade 3 encephalopathy that resolved with corticosteroids; 1 pt with grade 3 dysarthria and grade 3 ataxia). There was no treatment-related mortality. Eight pts are evaluable for response (DLBCL 6; FL 2). Overall response rate at 3 months is 50% with 3 complete responses (DLBCL 2 pts; FL 1 pt) and 1 partial response (FL); 4 pts with DLBCL had progressive disease before or at initial response assessment. CONCLUSIONS: In this ongoing trial of CTL019 cells in relapsed or refractory NHL, 4 of the first 8 evaluable pts responded to therapy. These early results demonstrate that CTL019 cells can be prepared from previously treated pts with active NHL, can undergo robust in-vivo expansion, and can induce complete responses in pts with advanced, relapsed or refractory DLBCL and FL. Longer follow up will define toxicities, durability of response, and clinical benefit, as well as guide further development of this promising new therapeutic approach. Disclosures Schuster: Novartis: Research Funding. Porter:Novartis: Patents & Royalties, Research Funding; Genentech (spouse employment): Employment. Mahnke:Novartis: Research Funding. Lacey:Novartis: Research Funding. Chew:Novartis: Patents & Royalties, Research Funding. Shah:Novartis: Employment. Hasskarl:Novartis: Employment. Litchman:Novartis: Employment. Frey:Novartis: Research Funding. Zheng:Novartis: Patents & Royalties, Research Funding. Levine:Novartis: Patents & Royalties, Research Funding. June:Novartis: Patents & Royalties, Research Funding.


2019 ◽  
Vol 61 (7) ◽  
pp. 983-991
Author(s):  
Ying-Chieh Lai ◽  
Wen-Cheng Chang ◽  
Chun-Bing Chen ◽  
Chi-Liang Wang ◽  
Yu-Fen Lin ◽  
...  

Background Pseudoprogression is difficult to diagnose in patients undergoing immunotherapy. Subjective response assessment is still common in clinical practice. Purpose To evaluate the differences between response evaluation criteria in solid tumors version 1.1 (RECIST 1.1), immune-related response criteria (irRC), and modified RECIST 1.1 for immunotherapy (iRECIST) through semi-automatic software, and to compare iRECIST-based response evaluation with subjective assessment. Material and Methods The best overall response of each patient based on RECIST 1.1, irRC, and iRECIST was determined on CT scans through semi-automatic software and the differences between the criteria were evaluated. Criteria-based response evaluation through semi-automatic software was compared with subjective assessment on radiology report by correlating the best overall response to overall survival. Results A total of 21 patients were included (five patients with melanoma, 12 patients with non-small-cell lung cancer, and four patients with hepatocellular carcinoma). Two patients with progressive disease by RECIST 1.1 but non-progressive disease by irRC and iRECIST eventually experienced tumor response and had favorable outcomes, indicating pseudoprogression. The survival difference between patients with non-progressive disease and progressive disease was better stratified through iRECIST-based response evaluation ( P = 0.078) than that through subjective assessment ( P = 0.501). Conclusion Pseudoprogression in immunotherapy may be captured through semi-automatic software utilizing irRC or iRECIST criteria. iRECIST-based response evaluation may provide a better survival stratification compared with subjective assessment.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3404-3404
Author(s):  
Sabine Kayser ◽  
Chiara Sartor ◽  
Marlise R. Luskin ◽  
Jonathan Webster ◽  
Fabio Giglio ◽  
...  

Abstract Background: Extramedullary B-cell acute lymphoblastic leukemia (EM-ALL) is a rare occurrence characterized by dismal outcome and no defined therapeutic approach. The monoclonal antibody inotuzumab ozogamicin (InO) is approved for treatment of CD22+ B-ALL. Aims: To characterize a series of relapsed/refractory (r/r) adult EM-ALL patients (pts) and evaluate outcome after treatment with InO. Methods: We studied 31 r/r pts (median age, 31 years; range, 19-81 years), who were treated with InO between 2015 and 2021 within a compassionate use program (n=7) or on-label after FDA or EMA approval (n=24) All pts were CD22 positive at relapse/progressive disease. Up to 6 InO cycles (≤2 cycles, n=19; 3-4 cycles, n=7; 5-6 cycles, n=5) were administered according to the previously approved regimen. EM response assessment was performed by CT or PET-CT. Prior therapy consisted of intensive chemotherapy +/- tyrosine kinase inhibitors. Allogeneic hematopoietic stem cell transplantation (allo-SCT) was performed in 18 pts (first line or at relapse, n=9, each). Prior to InO, blinatumomab was administered in n=14 and local irradiation in n=5 pts. Results: Overall, pts had in median 2 EM manifestations (range, 1-9). Localization of EM disease is shown in Table 1. In addition to EM disease, n=16 (52%) pts had a relapse in bone marrow. At the time of r/r EM-ALL median white blood cell and platelet counts were 5.1/nl (range, 0.04-24.7/nl) and 110.5/nl (range, 6-337/nl), respectively. Fifteen pts (48%) were female; ECOG was ≤ 2 in 29 pts and 3 in 2 pts. Cytogenetic analysis at the time of r/r EM-ALL was available in 13 (42%) pts. Of those, 6 pts had a normal karyotype, 4 were complex, 2 pts displayed a t(9;22) and 1 had an additional X-chromosome. Seven (23%) of the 31 pts had no response assessment after the first induction cycle including 1 patient who died at day 11 of the first InO cycle due to cerebral hemorrhage. Complete remission assessed by PET-CT (CR; including EM and hematological/bone marrow CR) after the first InO cycle was achieved in 10 of 24 assessed pts (42%), 9 pts (37.5%) had a partial remission (PR), 2 (8%) had stable disease (SD) and 3 (12.5%) showed resistant/progressive disease (RD/PD). After 2 InO cycles, CR was achieved in 17 of 31 pts (55%), PR in 9 (29%); 1 patient (3%) experienced early death and 4 pts with SD+RD/PD did not receive further InO treatment (13%). Median follow-up was 29 months (95%-CI, 21 months - not reached) and median overall survival (OS) 12.8 months (95%-CI, 9.9-16.2 months; Figure 1). One-year and 2-years OS rates were 53% (95%-CI, 37-76%) and 18% (95%-CI, 8-43%), respectively. In Cox regression analysis age as a continuous variable had no impact on OS (P=0.83). This was also true when using 60 years as cut-off (P=0.2). Twelve pts went on to allo-SCT (CR, n=6; PR, n=3; PD, n=3). Prior to allo-SCT 8 pts received ≤2 InO cycles and 4 pts ≤4 cycles. Sinusoidal obstruction syndrome (SOS) was reported in 1 patient after transplant; conditioning in this patient consisted of treosulfan/fludarabine/thiotepa. In pts achieving a CR after InO treatment (n=16), median OS was 10 months with no difference (P=0.80) in relapse-free survival (RFS) if an allo-SCT was performed (n=6) or not (n=10). There was no difference on OS (P=0.08) or RFS (P=0.2) if pts had EM manifestations only as compared to EM disease and bone marrow involvement. In patients with CR/PR after InO treatment, relapse occurred in 10 of 26 pts (38%; after allo-SCT, n=3); of those, all except than one succumbed of their disease. Two pts died in remission (sepsis, SOS/multi-organ failure, n=1; each). One patient experienced a molecular relapse, which could be successfully treated with InO again. Ten pts are in ongoing CR (n=9) or PR (n=1), including the patient with prior molecular relapse and InO re-exposure. Conclusions: This outcome analysis demonstrates that treatment with InO is an effective and promising approach in r/r-ALL patients with EM disease. However, allo-SCT alone seems not to be effective in maintaining disease control. Thus, autologous chimeric antigen receptor T-cells or advanced bi-specific antibodies as consolidation therapy should be evaluated in the future. Figure 1 Figure 1. Disclosures Webster: Pfizer: Consultancy; AmGen: Consultancy. Brunner: Keros Therapeutics: Consultancy; GSK: Research Funding; AstraZeneca: Research Funding; Aprea: Research Funding; Agios: Consultancy; Acceleron: Consultancy; Janssen: Research Funding; Takeda: Consultancy, Research Funding; BMS/Celgene: Consultancy, Research Funding; Novartis: Consultancy, Research Funding. Levis: AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen, Astellas Pharma, Daiichi-Sankyo, FujiFilm, and Menarini: Honoraria; Pfizer: Consultancy, Honoraria; Takeda: Honoraria; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Jazz: Consultancy, Honoraria; Astellas and FujiFilm: Research Funding. Schlenk: Agios: Honoraria; Astellas: Honoraria, Research Funding, Speakers Bureau; Celgene: Honoraria; Daiichi Sankyo: Honoraria, Research Funding; Hexal: Honoraria; Neovio Biotech: Honoraria; Novartis: Honoraria; Pfizer: Honoraria, Research Funding, Speakers Bureau; Roche: Honoraria, Research Funding; AstraZeneca: Research Funding; Boehringer Ingelheim: Research Funding; Abbvie: Honoraria. Papayannidis: Pfizer, Amgen, Novartis: Honoraria.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5237-5237 ◽  
Author(s):  
Chin-Hin NG ◽  
Liang Piu Koh ◽  
Melissa Ooi ◽  
Wei Ying Jen ◽  
Wee Joo Chng

Abstract Background: Midostaurin is a non-specific FLT3 inhibitor which showed anti-leukemic activity against both FLT3 mutated as well as unmutated acute myeloid leukemia (AML) in early phase studies. Panobinostat, a potent oral pandeacetylase inhibitor had also showed significant anti-leukemic signal in early phase studies. In the pre-clinical study, our group had previously reported synergistic anti-leukemic activity when a FLT3 inhibitor was used in combination with a HDAC inhibitor in vitro as well as in vivo animal study. The current study is our first bench-to-bedside study to further evaluate this combination therapy in AML patients. Method: Elderly AML who were either newly diagnosed and decline hypomethylating agent (HMA) or who failed HMA, MDS-RAEB-2, and relapsed refractory AML who were unfit for intensive chemotherapy or have no other treatment option were eligible regardless of FLT3 mutation status. The classical 3+3 dose escalation method was applied. Dose level 1 for Midostaurin was 50mg bd and Panobinostat was 10mg 3 times per week. Dose escalations were planned with only one drug escalation at a time on every level. Midostaurin was administered daily as continuous cycle while Panobinostat was given for the first 3 weeks of a 4 weeks cycle. Dose-limiting toxicities were defined as grade 3 or more non-haematologic toxicities occurring within the first cycle. Chronic toxicity is defined as recurrent or persistent adverse events that are possibly related to investigational products. Response assessments were done at post cycle 2 and cycle 4. Concomitant treatment with hydroxyurea was allowed up to cycle 1 day 14 to control leukocytosis if needed. Results: A total 7 patients have been recruited with a median age of 71 years old (range: 47 to 82). 2 patients had MDS-RAEB2, one with Acute myelomonocytic leukemia, 3 had AML with myelodysplasia-related changes (AML-MRC) and one with de novo FLT3-ITD AML. One of the AML-MRC had FLT3-ITD mutation and another had FLT3-TKD mutation. 4 out of 7 had prior HMA therapy, the remaining three were treatment naïve. 2 patients were not evaluable for DLT and response assessment due to progressive disease. 2 completed 2 cycles, the other 3 completed 4, 5 and 9 cycles respectively. No DLT was observed within cycle 1. Significant grade 1-2 treatment emerging adverse events were anorexia (4/5), fatigue (3/5), nausea (2/5), and dysgeusia (2/5). These toxicities persist beyond cycle 1 with increasing frequency. Grade 2 fatigue were observed in 4 patients, grade 2 anorexia in 4 patients, grade 1 dysgeusia in 4 patients, and grade 3 lipase elevation in one patient. 4 out of 5 patients developed severe grade 4 thrombocytopenia requiring regular platelet transfusion. Two had baseline grade 4 thrombocytopenia but the severity worsen after 1 cycle of treatment. These were thought to be chronic toxicities because the symptoms were largely resolved or improved during dose interruptions. In view of significant chronic toxicities, no further dose escalation was done and dose level 1 would be evaluated further in expansion cohort. 5 out 7 had post cycle 2 response assessment. One patient with FLT3-TKD achieved CRi, one AML-MRC showed 50% blast reduction, and the remaining 3 had stable disease. The one who achieved CRi eventually withdrew from study after completing 6 cycles due to fatigue, anorexia, severe grade 4 thrombocytopenia with gastrointestinal bleeding. As of report date, he remains alive and well after 9 months off study. His transfusion requirement has also reduced. One succumbed to intracranial bleeding during his cycle 10. He was in stable disease. The remaining 5 demised due to progressive disease. Conclusion: Even though no DLT was observed within cycle 1, significant chronic toxicities such as fatigue and anorexia were observed. Objective anti-leukemic activity was observed in 5 evaluable patients. Further expansion cohort would provide a clearer picture on the anti-leukemic effect as well as the chronic toxicities. Disclosures Chng: Janssen: Consultancy, Honoraria, Other: Travel, accommodation, expenses, Research Funding; Amgen: Consultancy, Honoraria, Other: Travel, accommodation, expenses; Merck: Research Funding; Aslan: Research Funding; Takeda: Consultancy, Honoraria, Other: Travel, accommodation, expenses; Celgene: Consultancy, Honoraria, Other: Travel, accommodation, expenses, Research Funding.


2009 ◽  
Vol 27 (3) ◽  
pp. 404-410 ◽  
Author(s):  
Simon J. Crabb ◽  
Demetris Patsios ◽  
Eric Sauerbrei ◽  
Peter M. Ellis ◽  
Andrew Arnold ◽  
...  

PurposeWe have observed cavitation of lesions in clinical trials of an angiogenesis inhibitor combined with chemotherapy for non–small-cell lung cancer (NSCLC). We hypothesized that cavitation might alter response assessment in such clinical trials.Patients and MethodsWe performed a retrospective radiologic review of patients with NSCLC enrolled onto three National Cancer Institute of Canada Clinical Trials Group (NCIC CTG) trials of platinum-based chemotherapy with or without a small-molecule angiogenesis inhibitor (vascular endothelial growth factor receptor inhibitor [VEGFRI]). Response was assessed both by Response Evaluation Criteria in Solid Tumors (RECIST) guidelines and a novel alternate method in which the longest diameter of any cavity was subtracted from the overall longest diameter of that lesion to measure target lesions. Rates of cavitation were documented.ResultsMarked cavitation of pulmonary lesions was seen in 24% of 33 patients treated with VEGFRI combined with platinum-based chemotherapy but in none of 18 patients treated with platinum-based chemotherapy alone. Use of the alternate method for response assessment resulted in an alteration of response assessment, time to best response, duration of response, and time of disease progression in a minority of patients compared with RECIST.ConclusionCavitation of target and nontarget lesions is common in NSCLC patients treated with VEGFRIs and platinum-based chemotherapy. Impact on response and time to event outcomes occurred but seems to be less common. Response assessment might be improved by incorporating cavitation into volume assessment for target lesions, potentially altering outcomes of key efficacy parameters in clinical trials. This should be prospectively assessed in clinical trials of angiogenesis inhibitors.


Sign in / Sign up

Export Citation Format

Share Document