Chromosome 1q Amplification Is Associated with a History of Prior Malignancies Among Patients Newly Diagnosed with Multiple Myeloma

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2193-2193
Author(s):  
Elizabeth B Lamont ◽  
Andrew J. Yee ◽  
Stuart L. Goldberg ◽  
Andrew D Norden

Background: Over the past 20 years, observational data from usual care clinical oncology settings has been leveraged to inform estimates of cancer treatment-associated benefits and risks among patients not treated on clinical trials. Increasing genomic testing to inform treatment decisions in usual care settings now meaningfully augments traditional observational data, positioning it to provide insights beyond clinical care into tumor biology. We studied patients with newly diagnosed multiple myeloma (MM), comparing cytogenetic test patterns according to history of prior malignancy. Methods: In this retrospective cohort study, we identified 2,380 patients from the COTA real-world database (RWD) who were newly diagnosed with MM in the years 2010-2018. The COTA RWD is a de-identified composite of both abstracted electronic health record and administrative data pertaining to patients receiving their cancer care at one of COTA's clinical oncology practice partners. Among these patients, 1769 (74%) had evidence of MM-associated cytogenetic testing with fluorescent in-situ hybridization (FISH) within the 120 days surrounding their date of diagnosis. The 1,769 patients form the analytic cohort. We compared patients' FISH results for t(4;14), deletion(17p), t(14;16), deletion(13), t(14;20), t(6;14), t(11;14), deletion (1p), and amplification(1q) according to their history of prior malignancy. Results: Within the cohort, 263 prior malignancies were identified in 241 patients (14%, 241/1,769). Two-hundred and twenty-one patients (92%) had one prior malignancy, 28 (7.9%) had two prior malignancies, and one (<1%) had four prior malignancies. The most common prior malignancies were prostate (n=50), breast (n=19), melanoma (n=14), skin (n=13), and cervix (n=6). Amplification of the long arm of chromosome one (amp(1q)) was noted in 31% of patients (75/241) with a prior malignancy vs. 24% of patients (370/1,528) without (chi2 test p=0.02). Overall 25% of patients had amp(1q). No other translocations, amplifications, deletions were associated with prior cancers. A non-parametric test for trend revealed a strong positive association between patients' malignancy count (range 0-4) and amp1q (p<0.01). MM patients with prior lymphomas and prior melanomas also had high rates of amp(1q), though these were not significantly different from patients without these prior malignancies. In a multivariable logistic regression model that adjusted for patient demographic attributes, other known potentially collinear MM poor prognostic factors (i.e., revised ISS stage, IgA sub-type, lambda light chains) and adjusted standard errors for clustering of patients within treatment settings, a history of prostate cancer remained clinically and statistically significantly positively associated with amp(1q) (OR 2.1, 95% CI: 1.9-2.2) as did history of two or more prior malignancies (OR 2.8, 95% CI: 2.3-3.3). Of note, amp(1q) was positively associated with IgA subtype (OR 1.5, 95% CI: 1.3-1.6) and the presence of lambda subtype (OR 1.3, 95%CI: 1.3-1.4). Conclusions: Using RWD, we found that newly diagnosed MM patients with histories of prostate cancer and those with two or more prior malignancies were more likely to have amp(1q), a poor prognostic marker in MM. Gains in 1q have previously been identified among patients with prostate and lymphoid cancers, but to our knowledge this is the first study to identify an association with a prior history of cancer, especially prostate cancer, and amp(1q) in MM. This relationship is worth further exploration of whether there is a common pathway associated with for example risk of prostate cancer and amp(1q) in MM. Clinical trials are less likely to answer this question as patients with prior malignancies are often excluded from enrollment. Overall, the results reported suggest that RWD is an efficient and comparatively inexpensive tool to support research in cancer biology through hypothesis generating and testing analyses of linked real-world phenotypic and genotypic data. Disclosures Lamont: COTA: Employment. Yee:Celgene: Consultancy, Honoraria, Research Funding; Karyopharm: Consultancy; Bristol-Myers Squibb: Consultancy, Research Funding; Takeda: Consultancy; Adaptive: Consultancy; Amgen: Consultancy, Honoraria. Goldberg:Cancer Outcomes Tracking and Analysis (COTA) Inc.: Equity Ownership; COTA: Equity Ownership; Bristol-Myers Squibb: Consultancy. Norden:COTA: Employment, Equity Ownership.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5493-5493
Author(s):  
Yuan Yao ◽  
Dehui Zou ◽  
Aijun Liao ◽  
Xiaoxia Chu ◽  
Wei Wang ◽  
...  

Background: Multiple Myeloma (MM) is a disease of the elderly, whose prognoses are highly heterogeneous. Hence International Myeloma Working Group (IMWG) proposed geriatric assessment (GA) in 2015, including daily activity and comorbidity status, to better discriminate between fit and frail patients (Palumbo et al, 2015). However, IMWG recruited patients from clinical trials instead of real world practices. Therefore we studied GA in elderly MM patients consecutively in China, along with other perspectives which are known to be problematic in elderly population that were previously left unnoticed, such as nutrition status, risk of cognitive impairment, risk of depression, and quality of life. Aim: Our study centers on the feasibility to perform a more comprehensive geriatric assessment (cGA) in elderly MM patients, current cGA status in elderly MM patients in China, and the cGA difference between Chinese patients and patients in the IMWG study. Method: From August 2017 to April 2019, we continuously recruited 336 newly diagnosed elderly (age ≥ 65) MM patients from 21 centers in China. cGA was performed at diagnosis, after treatment cycle 1, after cycle 4, and 1 year after treatment. cGA includes physical conditions (ECOG), activities of daily living (ADL), instrumental ADL (IADL), mini-nutritional assessment (MNA-SF), geriatric depression scale (GDS), mini-mental state examination (MMSE), quality of life (SF-36) and Charlson comorbidity index (CCI). Staging was assessed at baseline (International Staging System (ISS) & Revised ISS) and hematological responses were evaluated along with each cGA timepoint. Results: We pool-analyzed data of 336 newly-diagnosed elderly MM patients. The median age was 70 (range 65-88) and 25.5% of patients were older than 75 years. 336 (100%) patients were able to complete cGA, and median assessment time was 40 minutes (range 20-70). Upon diagnosis, only 34% and 37.5% of patients had full ADL and IADL respectively. 38.5% of patients had moderate to high risk of depression (GDS ≥ 6). 13.2% of patients were malnourished (MNA-SF ≤ 7), while 46.3% of patients were at risk of malnutrition (8 ≤ MNA-SF ≤ 11). 41% of patients had at least one comorbidity (CCI ≥ 1). 45.7% of patients had moderate to intermediate risk of cognitive impairment (MMSE ≤ 26). Grouping by IMWG-GA index, our study identified 59.9% patients in frail group (vs 39% in IMWG study), 15.8% in intermediate (vs 31% in IMWG) and 24.3% in fit (vs 30% in IMWG). 69% of patients received proteasome inhibitor-containing regimens and 20.7% of patients received lenalidomide-containing regimens. Best hematological responses in fit and intermediate groups were better than responses in frail group (≥ PR rate: 88.5% in fit, 94.4% in intermediate vs 77.5% in frail). Median follow up time was 10 months. To date, 215 (64%) patients have finished the cGA after cycle 1; 164 (48.8%) patients have finished the cGA after cycle 4; 91 (27.1%) patients has finished all 4 planned cGA and improvements in cGA were observed in the majority of these patients. Conclusion: Our study showed significant CGA heterogeneity in elderly MM patients. Even in the IMWG-GA "fit" group, nutrition, depression and cognitive impairment remain problems. Frail patients took up a larger proportion in Chinese elderly MM patients compared to IMWG study. Our study strongly justifies the necessity for cGA in elderly patients with MM, more so in the real world MM patients than in the clinical trials. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5701-5701
Author(s):  
Justin King ◽  
Mark A. Fiala ◽  
Scott R. Goldsmith ◽  
Keith E. Stockerl-Goldstein ◽  
Mark A. Schroeder ◽  
...  

Historically, high-dose therapy in combination with autologous stem cell transplants (ASCT) for multiple myeloma (MM) was reserved for younger patients. In more recent years, the use of ASCT has expanded in the older population. However, there is still limited data on the utilization and efficacy of ASCT in older patients, particularly those over the age of 75. To further evaluate this issue, we retrospectively analyzed all patients with newly diagnosed MM between the ages of 75-78, the institutional cutoff for ASCT eligibility, that were referred to the stem cell transplant unit at our institution for consultation from the years 2012-2018. Baseline characteristics, anti-myeloma treatments, and patient outcomes were abstracted through chart review. Seventy-five patients were referred to our institution. 71% were male, 29% female. 39% patients were considered ineligible for ASCT by the consulting transplant physician. Most patients were considered transplant ineligible due to comorbidities or poor performance status. Of the 46 patients eligible for ASCT, 52% underwent the procedure during their first-line therapy. The majority of those patients received reduced intensity melphalan (140 mg/m2) while 2 patients received conventional dosing (200 mg/m2). The other 22 patients eligible for ASCT declined or elected to defer the procedure and to be treated with conventional therapy. The characteristics of these three groups were similar and are detailed in Table 1. After a median follow-up of 30 months, 25% of the patients had expired. Estimated median overall survival (OS) was 71.3 months (unable to quantitate 95% CI) for all patients. Compared to transplant eligible patients, regardless of transplant receipt, those who were transplant ineligible had a 186% increase risk for death (HR 2.86; 95% CI 1.12-7.35; p = 0.029). There was also a notable trend for longer OS in those who underwent ASCT compared to those who were eligible but declined the procedure, but it was not statistically significant (HR 0.36; 95% CI 0.10-1.28; p = 0.114). At a transplant center, two-thirds of patients referred for newly diagnosed MM between the ages 75-78 were considered eligible for ASCT and one-third underwent the procedure. Outcomes were better for patients eligible for ASCT, regardless of whether they underwent the procedure. There was also a trend for better OS in patients who underwent the procedure compared to those who declined. While small sample sizes and the retrospective nature of the study limit our ability to draw conclusions, it appears that ASCT has an OS benefit among patients age 75-78. Disclosures Fiala: Incyte: Research Funding. Stockerl-Goldstein:AbbVie: Equity Ownership; Abbott: Equity Ownership. Vij:Genentech: Honoraria; Janssen: Honoraria; Bristol-Myers Squibb: Honoraria, Research Funding; Sanofi: Honoraria; Karyopharm: Honoraria; Takeda: Honoraria, Research Funding; Celgene: Honoraria, Research Funding. Wildes:Janssen: Research Funding; Carevive: Consultancy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3787-3787
Author(s):  
Paul B Koller ◽  
Hagop M Kantarjian ◽  
Charles Koller ◽  
Elias Jabbour ◽  
Susan O'Brien ◽  
...  

Abstract Abstract 3787 Background: The natural history of CML has been irrevocably changed since the advent of TKIs with pts living longer than they ever have. Some patients have a history of previous cancers by the time they are diagnosed with CML. These pts' long-term prognosis has never been previously described. Aims: To determine the effect of prior malignancy on patient outcome after diagnosis of CML. Methods: The primary objective was to determine outcome of pts with a previous diagnosis of malignancy (PM) vs a group without (nPM). Patients included in clinical trials of TKI as initial therapy for CML in chronic phase from July 2000 to January 2011 were reviewed. Results: Of the 471 CML pts treated with frontline TKIs 47 (10%) had a PM before their CML diagnosis and 5 (1%) pts PM status could not be obtained. The median age of the patients with a PM was 60 (30–84) compared to 46 (15–86) for those with no PM. There were no significant differences in clinical characteristics between PM and nPM patients. The median from diagnosis of a PM and a diagnosis of CML was 69 months (mo) (7 mo to 707 mo). The five most common PMs were: non-melanoma skin cancer in 14 (30%), breast cancer in 10 (21%), melanoma in 5 (11%), colorectal cancer in 5 (11%), and prostate cancer in 5 (11%). “Six pts (13%) had more than one PM and 2 of those 6 had 3 PM before the diagnosis of CML, the other 4 pts had 2 PM. The PMs were treated prior to the diagnosis of CML in the following ways: 17 (36%) pts received chemotherapy for their PM, 17 (36%) received radiotherapy, and 43 (91%) received surgery. At the time of CML diagnosis 3 (6%) pts had active cancer, while the remainder of the pts with a PM were thought to be in remission. Two (4%) pts were on active therapy for their prior cancer diagnosis at the time they started on CML therapy (Tamoxifen for breast cancer in both). After the diagnosis of CML, 6 (13%) pts had a recurrence of their PM including 2 pts with basal cell cancer, and 1 each with melanoma, breast cancer, prostate cancer, and lymphoma. These recurrent malignancies were treated as follows: 3 with radiation, 2 had surgery, and 2 with chemotherapy. Five of these 6 pts continued TKI while receiving therapy for PM. The median time between diagnosis of CML and relapse of PM was 18 months. Median remission of the PM was 151 months. Also, 6 (13%) pts had a new malignancy after CML diagnosis: 2 pts with squamous skin carcinoma, and 1 each with chronic lymphocytic leukemia, papillary thyroid carcinoma, mucoepidermoid carcinoma, and large B-cell lymphoma. Treatments for the new onset malignancy after the diagnosis of CML include: surgery (4), chemotherapy (2), radiation (1), and observation (1). The median time between diagnosis of CML and a new malignancy was 39 months. Six of the 47 pts are deceased: 2 died of their PM, 2 died secondary to treatment complications for their PM, 2 died of cardiovascular issues unrelated to a cancer diagnosis. None died of CML. Initial treatment for CML was imatinib 400mg in 4/47 (9%) PM and 69/419 (16%) nPM, imatinib 800mg in 15/47 (33%) PM and 189/419 (45%) nPM, dasatinib in 11/47 (24%) PM and 78/419 (19%) nPM, and nilotinib in 16/47 (35%) PM, and 83/419(20%) nPM. The outcome of pts with PM and nPM is presented in Table 1. There was no significant different in patient outcome between pts with PM and nPM. Conclusion: Pts with CML who have prior malignancies have the same excellent outcome as patients with no prior malignancies. In the few instances in which concomitant therapy for other malignancies was required during therapy with TKI this could be accomplished with no significant toxicity. Thus, the history of prior malignancy in a patient who develops CML should not affect the decision to treat with TKI. Disclosures: Cortes: Pfizer Inc: Consultancy, Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2656-2656
Author(s):  
Steven R. Arikian ◽  
Dejan Milentijevic ◽  
Gary Binder ◽  
Mara Silvia Monzini ◽  
X Henry Hu ◽  
...  

Abstract Introduction: As clinical evidence has mounted in support of novel agents and longer treatment (Tx) durations for patients (pts) with newly diagnosed multiple myeloma (NDMM), questions have arisen regarding the economic impact of extending time to progression (TTP) in these pts, and the cost consequences once pts relapse and move to a second line of Tx. Previous analysis showed that relapsed myeloma pts incurred higher monthly costs once they advanced to later lines of Tx (Gaultney, 2013). There is limited information on the cost patterns of MM pts before and after their first relapse. A claims analysis was performed to evaluate the patterns of total direct costs of care, from Tx initiation until progression, for NDMM patients and for newly relapsed patients treated with novel agents, utilizing time to next therapy (TTNT) as a proxy measure for progression. Methods: A retrospective study was conducted using a large US medical and pharmacy claims database, covering > 25 million lives annually. NDMM patients were identified with at least 2 outpatient claims or 1 inpatient medical claim associated with a diagnosis of MM (ICD-9-CM] code 203.0X), with the first such claim used to define the index date. Inclusion criteria required a minimum of 12 months' pre-index enrollment and 6 months' post-index continuous enrollment between 2006 and 2012. Pts with claims for stem cell transplantation (SCT) were excluded, to avoid confounding results from various factors based on timing, costs, and site of care of SCT. The analysis focused on NDMM and relapsed MM pts receiving lenalidomide (LEN)- or bortezomib (BORT)- based Tx, where complete claim history was available from Tx onset to initiation of subsequent Tx. Using methods similar to those described by Gaultney, patients' average monthly costs were determined, including medical (inpatient, ambulatory, and emergency room) and pharmacy (index and other drugs) costs, and total cost patterns over quarterly time periods were calculated. Average Charlson comorbidity scores were determined to compare baseline measures between pt groups. Results: 897 NDMM pts and 280 relapsed MM pts were identified with complete data through initiation of subsequent Tx. Monthly total direct costs for NDMM pts were $15,400 in the first 3 months (mos) of Tx, and declined each quarter, reaching approximately $5,000/mo at 18+ mos. At relapse, monthly costs increased to over $12,000 for the first 3 mos and followed a quarterly pattern of reduction similar to that seen for NDMM pts (Fig 1). Quarterly cost reduction patterns were consistent for patients treated with LEN or BORT for both NDMM and relapsed pts. Pts' total monthly NDMM costs over the full TTNT period averaged $8,942 with LEN vs. $11,139 for BORT (due to 54% higher monthly medical costs for BORT), while monthly drug costs were nearly identical (Table 1). The baseline Charlson comorbidity index was similar between Tx groups in both lines of Tx. Figure 1: Direct monthly costs (medical and pharmacy) for LEN- and BORT-based treatments Figure 1:. Direct monthly costs (medical and pharmacy) for LEN- and BORT-based treatments Table 1: Direct monthly costs for NDMM pts Table 1 Table 1. Conclusions: For a population of NDMM pts receiving either LEN- or BORT-based Tx without SCT, followed until TTNT, total direct monthly costs per pt declined steadily over time, decreasing by 68% from the initial quarter to the period post 18 mos. Costs spiked when pts began 2nd-line therapy, then followed a similar pattern of decline over time. This pattern may suggest that further extending the TTP for NDMM pts may also yield economic benefits for each month extended before relapse. Patterns of cost decline were similar between the LEN and BORT groups, for NDMM and for relapsed patients, although mean monthly total costs were lower for NDMM pts receiving LEN-based Tx due to lower medical costs and similar drug costs. Disclosures Arikian: Genesis Research: Consultancy. Off Label Use: Lenalidomide in newly diagnosed multiple myeloma patients . Milentijevic:Celgene Corporation: Consultancy. Binder:Celgene Corporation: Employment, Equity Ownership. Monzini:Celgene Corporation: Employment, Equity Ownership. Hu:Celgene Corporation: Employment. Nagarwala:Celgene Corporation: Employment. Hussein:Celgene Corporation: Employment. Corvino:Genesis Research LLC: Consultancy. Surinach:Genesis Research LLC: Consultancy. Usmani:Celgene Corporation: Consultancy, Honoraria, Research Funding; Millennium: Consultancy, Honoraria; Onyx: Consultancy, Honoraria, Research Funding; Sanofi: Consultancy; Array BioPharma: Research Funding; Janssen: Research Funding; Pharmacyclics: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4229-4229
Author(s):  
Jatin J. Shah ◽  
Rafat Abonour ◽  
Mohit Narang ◽  
Jayesh Mehta ◽  
Howard R. Terebelo ◽  
...  

Abstract Introduction: Triplet therapies are used for treatment (Tx) of both transplant-eligible and -ineligible patients (pts) with newly diagnosed multiple myeloma (NDMM). Actual patterns and outcomes of Tx are not fully understood. Connect MM® is the first and largest multicenter, US-based, prospective observational cohort study designed to characterize Tx patterns and outcomes for pts with NDMM. This analysis describes demographic and disease characteristics of pts who received triplet Tx as an induction regimen and for whom transplant was or was not intended. The analysis explores the relationship of these factors with overall survival (OS) and other efficacy endpoints. Patients and Methods: Pts aged ≥ 18 y with NDMM within 60 days of diagnosis were eligible for enrollment regardless of disease severity, medical history, or comorbidities. Data including transplant intent (yes/no) was collected at baseline; follow-up data was collected quarterly thereafter. Based on the initial intent, 2 groups were identified: patients with intent to transplant who received transplant (TT) and pts with no intent to transplant who did not receive a transplant (NT). Triplet Tx was defined as the combination of ≥ 3 concurrent therapeutic agents in the first course of Tx (within 56 days of study entry). KM analysis adjusted for age was conducted for OS. Because decisions on use of transplant and triplet therapy are influenced by multiple factors, a multivariable Cox regression analysis was performed to evaluate the contribution of the triplet therapy (yes/no) to OS and was adjusted for other variables, including age, comorbidities, and ISS staging. Results: Between September 2009 and December 2011, 1493 pts were enrolled. This analysis was on 1436 pts: 650 pts with transplant intent and 786 pts without transplant intent. The data cutoff date was November 30, 2014, and the median follow-up for overall survival (OS) was 33.8 mos. Of pts with transplant intent, 451 (69%) received transplant (TT) and 199 (31%) did not. Of pts without transplant intent, 62 (8%) received transplant and 724 (92%) did not (NT). The abstract focuses on TT and NT groups only. NT pts tended to be older and have more advanced ISS staging and higher β2-microglobulin levels than TT pts (Table). The most common triplet regimen given during the first course treatment (within 56 days) was lenalidomide, bortezomib, and dexamethasone (RVd). RVd was administered to 34% of the NT pts (76/225) and 59% of the TT pts (152/257). The most common non-triplet regimen was bortezomib and dexamethasone (Vd), which was given to 31% of NT pts (156/499) and 38% of TT pts (73/194). Within the NT group, pts given triplet Tx had a lower risk of death than those who did not receive triplet Tx (P = .0013). The multivariable analysis found triplet Tx to be associated with a 36% reduced risk of death (hazard ratio [HR] = 0.64 [95% CI, 0.50-0.82]; P = .001). ISS disease stage (HR = 1.43 [95% CI, 1.21-1.69]; P < .001) and history of diabetes (HR = 1.38 [95% CI, 1.08-1.78]; P = .012) were negative prognostic factors for OS. Within the TT group, pts who received triplet Tx did not attain an OS benefit (P = .8993), and no baseline characteristics were significantly associated with OS. These results may be limited by other factors not considered that may have influenced physicians' choice of treatment, including the use of maintenance therapy and a short follow-up period of 33.8 months. Conclusions: Triplet Tx as a first regimen is associated with longer OS in pts without transplant intent who did not receive a transplant. RVd and Vd were the most common first Tx regimens, respectively. Continued follow-up of these pts and enrollment of an additional cohort will provide additional data with mature follow-up. Table 1. Table 1. Disclosures Shah: Bristol-Myers Squibb: Research Funding; Array: Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Onyx: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Millenium: Research Funding; Merck: Membership on an entity's Board of Directors or advisory committees. Abonour:Celgene: Research Funding, Speakers Bureau. Narang:Celgene: Speakers Bureau. Mehta:Celgene Corporation: Speakers Bureau. Terebelo:Millenium: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pharmacylics: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Gasparetto:Celgene Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Millennium: Honoraria, Other: Export Board Committee, Speakers Bureau. Toomey:Celgene: Consultancy. Hardin:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees. Srinivasan:Celgene Corporation: Employment, Equity Ownership. Larkins:Celgene Corporation: Employment, Equity Ownership. Nagarwala:Celgene Corporation: Employment, Equity Ownership. Rifkin:Onyx Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Millennium Pharmaceuticals, Inc., Cambridge, MA, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited: 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.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3789-3789
Author(s):  
Ben A Derman ◽  
Andrew J. Belli ◽  
Ching-Kun Wang ◽  
Eric Hansen ◽  
Spencer S Langerman ◽  
...  

Abstract Background: Multiple myeloma (MM) risk stratification schemata such as the International Staging System (ISS) and Revised-ISS (R-ISS) were derived from clinical trial subjects made up predominately of younger White individuals with adequate renal function. It is unknown whether these prognostic indices are applicable to all patients with newly diagnosed (ND) MM, especially among Black individuals, older adults, and those with renal dysfunction. The R-ISS expanded on the ISS by including and serum lactate dehydrogenase (LDH) and high-risk cytogenetic abnormalities (HRCA) identified by fluorescence in-situ hybridization (FISH), but HRCA may not translate into poor prognosis for older adults and for Black individuals. We sought to create an inclusive risk prognostic index for NDMM using real-world data derived from electronic health records. Methods: De-identified NDMM patient-level data in the real-world setting was provided by COTA, Inc. 3000 patients were identified who met the inclusion criteria of NDMM between 2005 and 2020. Baseline diagnostic parameters available within 60 days before or after diagnosis were included. Progression free survival (PFS) was defined as the time from diagnosis to disease progression or death of any cause. Overall survival (OS) was defined as the time from diagnosis to death of any cause. Proportional hazards models were used to calculate hazard ratios (HR) and 95% confidence intervals for all-cause mortality. Age-adjusted univariate analyses identified variables significantly associated with OS, and continuous variables were dichotomized based on accepted cutoffs. Multivariate Cox models to identify the variables with the strongest association with OS were performed adjusting for age, sex, Black race, receipt of proteasome inhibitor and immunomodulatory imide during induction, autologous stem cell transplant within 1 year of diagnosis, ECOG performance status, and creatinine. An additive risk score was created with one point given to each significant variable. The risk score was then validated for PFS using the Multiple Myeloma Research Foundation's (MMRF) CoMMpass database (version IA15). Results: 3000 NDMM pts from the COTA, Inc. real-world database were initially evaluated, and a total of 689 NDMM pts had sufficient level of data to be included. The median follow-up time was 49.9 months (interquartile range (IQR) 29.1-76.2 months). Median age was 64 (IQR 32-86), including 44% age 65+. Of the 607 with reported race, 474 (78%) were White, 86 (14%) Black, 17 (3%) Asian, and 30 (5%) other. Of the 676 pts with reported serum creatinine (mg/dL), the median was 1 mg/dL (IQR 0.8-1.3) with 85 (13%) measuring &gt;2 mg/dL. Examined peripheral blood variables were: calcium (corrected for albumin), albumin&lt;3.5 mg/dL, beta2-microgloublin (B2M) &gt;3.5 mcg/mL, LDH &gt;250 U/L, hemoglobin &lt;10 g/dL, M-spike &gt;3 g/dL, and IgA isotype. Variables with significance using multivariate analysis at p&lt;0.1 were: LDH&gt;250 U/L, B2M &gt;3.5 mcg/mL, hemoglobin &lt;10 g/dL, and IgA isotype. These variables were simultaneously present in 558 patients. Patients were stratified into 3 groups: standard (std score = 0, n=186), intermediate (int score = 1-2, n=295), and high (score 3-4, n=77) risk. For this inclusive risk prognostic index (IRPI), the c-statistic was 0.61 for OS (HR 2.0, 95% CI 1.5-2.6, p&lt;0.001) which compared favorably to the c-statistic for ISS (c=0.64, HR 1.8, 95% CI 1.5-2.2, p&lt;0.001) and for R-ISS (c=0.63, HR 2.0, 95% CI 1.6-2.6). For the IRPI, median OS was 218 (std) vs 121.5 (int) vs 79.5 months (high). In comparison, median OS by ISS was 198.9 (stage I) vs 121.6 (stage II) vs 80.6 months (stage III), and by R-ISS: 198.9 (I) vs 121.6 (II) vs 79.5 months (III). Validation of the inclusive risk prognostic index (IRPI) using the MMRF CoMMpass database in 938 patients with all four criteria showed median PFS was 44 (std) vs 33 (int) vs 20.5 months (high). In comparison, median PFS by ISS was 45.9 (I) vs 31.5 (II) vs 20.5 (III) months. Median PFS by R-ISS was 50.1 (I) vs 32.7 (II) vs 19.1 (III) months. Conclusions: Employing real-world datasets that incorporate a more diverse patient population led to the generation of an inclusive risk prognostic index incorporating beta2-microgloublin, LDH, hemoglobin, and IgA isotype. This IRPI does not require bone marrow sampling, performs similarly to ISS and R-ISS in predicting PFS, and with datasets with longer follow-up may prove to predict OS. Figure 1 Figure 1. Disclosures Derman: Sanofi: Membership on an entity's Board of Directors or advisory committees. Belli: COTA, Inc.: Current Employment, Other: Equity ownership. Wang: COTA, Inc.: Current Employment, Other: Equity ownership. Hansen: COTA, Inc.: Current Employment. Jakubowiak: Amgen: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Gracell: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; GSK: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Janssen: 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. 4327-4327
Author(s):  
James Croft ◽  
Andrew Hall ◽  
Amy L Sherborne ◽  
Katrina Walker ◽  
Sidra Ellis ◽  
...  

Background Treatment of relapsed/refractory multiple myeloma (RRMM) remains challenging as durable remissions are achieved in patient sub-groups only. Identifying patients that are likely to benefit prior to or early after starting relapse treatments remains an unmet need. MUKseven is a trial specifically designed to investigate and validate biomarkers for treatment optimization in a 'real-world' RRMM population. Design In the randomized multi-center phase 2 MUKseven trial, RRMM patients (≥2 prior lines of therapy, exposed to proteasome inhibitor and lenalidomide) were randomized 1:1 to cyclophosphamide (500 mg po d1, 8, 15), pomalidomide (4 mg days 1-21) and dexamethasone (40 mg; if ≥75 years 20 mg; d1, 8, 15, 21) (CPomD) or PomD and treated until progression. All patients were asked to undergo bone marrow (BM) and peripheral blood (PB) bio-sampling at baseline, cycle 1 day 14 (C1D14, on-treatment) and relapse. For biomarker discovery and validation, IGH translocations were profiled by qRT-PCR, copy number aberrations by digital MLPA (probemix D006; MRC Holland), GEP by U133plus2.0 array (Affymetrix), PD protein markers by IHC and PB T-cell subsets by flow cytometry for all patients with sufficient material. Primary endpoint was PFS, secondary endpoints included response, OS, safety/toxicity and biomarker validation. Original planned sample size was 250 patients but due to a change in UK standard of care during recruitment with pomalidomide becoming available, a decision was made to stop recruitment early. Results In total, 102 RRMM patients were randomized 1:1 between March 2016 and February 2018. Trial entry criteria were designed to include a real-world RRMM population, permitting transfusions and growth factor support. Median age at randomization was 69 years (range 42-88), 28% of patients had received ≥5 prior lines of therapy (median: 3). Median follow-up for this analysis was 13.4 months (95% CI: 12.0-17.5). 16 patients remained on trial at time of analysis (median number of cycles: 19.5; range 8-28). More patients achieved ≥PR with CPomD compared to PomD: 70.6% (95% CI: 56.2-82.5%) vs. 47.1% (CI: 32.9-61.5%) (P=0.006). Median PFS was 6.9 months (CI: 5.7-10.4) for CPomD vs. 4.6 months (CI: 3.5-7.4) for PomD, which was not significantly different as per pre-defined criteria. Follow-up for OS is ongoing and will be presented at the conference. High-risk genetic aberrations were found at following frequencies: t(4;14): 6%, t(14;16)/t(14;20): 2%, gain(1q): 45%, del(17p): 13%. Non-high risk lesions were present as follows: t(11;14): 22%, hyperdiploidy: 44%. Complete information on all high-risk genetic markers was available for 71/102 patients, of whom 12.7% had double-hit high-risk (≥2 adverse lesions), 46.5% single-hit high-risk (1 adverse lesion) and 40.8% no risk markers, as per our recent meta-analysis in NDMM (Shah V, et al., Leukemia 2018). Median PFS was significantly shorter for double-hit: 3.4 months (CI: 1.0-4.9) vs. single-hit: 5.8 months (CI: 3.7-9.0) or no hit: 14.1 months (CI: 6.9-17.3) (P=0.005) (Figure 1A). GEP was available for 48 patients and the EMC92 high-risk signature, present in 19% of tumors, was associated with significantly shorter PFS: 3.4 months (CI: 2.0-5.7) vs. 7.4 (CI: 3.9-15.1) for EMC92 standard risk (P=0.037). Pharmacodynamic (PD) profiling of cereblon and CRL4CRBN ubiquitination targets (including Aiolos, ZFP91) in BM clots collected at baseline and C1D14 is currently ongoing. Preliminary results for the first 10 patients demonstrate differential change of nuclear Aiolos (Figure 1C), with a major decrease in Aiolos H-scores in 7/10 patients from baseline to C1D14 and reconstitution at relapse. T-cell PB sub-sets were profiled at baseline and C1D14 by flow cytometry. Specific sub-sets increased with therapy from baseline to C1D14, e.g. activated (HLA-DR+) CD4+ T-cells, as reported at last ASH. CD4+ T-cell % at baseline was associated with shorter PFS in these analyses in a multi-variable Cox regression model (P=0.005). PD and T-cell biomarker results will be updated and integrated with molecular tumor characteristics and outcome. Discussion Our results demonstrate that molecular markers validated for NDMM predict treatment outcomes in RRMM, opening the potential for stratified delivery of novel treatment approaches for patients with a particularly high unmet need. Additional immunologic and PD biomarkers are currently being explored. Disclosures Croft: Celgene: Other: Travel expenses. Hall:Celgene, Amgen, Janssen, Karyopharm: Other: Research funding to Institution. Walker:Janssen, Celgene: Other: Research funding to Institution. Pawlyn:Amgen, Janssen, Celgene, Takeda: Other: Travel expenses; Amgen, Celgene, Janssen, Oncopeptides: Honoraria; Amgen, Celgene, Takeda: Consultancy. Flanagan:Amgen, Celgene, Janssen, Karyopharm: Other: Research funding to Institution. Garg:Janssen, Takeda, Novartis: Other: Travel expenses; Novartis, Janssen: Research Funding; Janssen: Honoraria. Couto:Celgene Corporation: Employment, Equity Ownership, Patents & Royalties. Wang:Celgene Corporation: Employment, Equity Ownership. Boyd:Novartis: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria. Pierceall:Celgene: Employment. Thakurta:Celgene: Employment, Equity Ownership. Cook:Celgene, Janssen-Cilag, Takeda: Honoraria, Research Funding; Janssen, Takeda, Sanofi, Karyopharm, Celgene: Consultancy, Honoraria, Speakers Bureau; Amgen, Bristol-Myers Squib, GlycoMimetics, Seattle Genetics, Sanofi: Honoraria. Brown:Amgen, Celgene, Janssen, Karyopharm: Other: Research funding to Institution. Kaiser:Takeda, Janssen, Celgene, Amgen: Honoraria, Other: Travel Expenses; Celgene, Janssen: Research Funding; Abbvie, Celgene, Takeda, Janssen, Amgen, Abbvie, Karyopharm: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3251-3251
Author(s):  
Chang-Ki Min ◽  
Sung-Soo Yoon ◽  
Wee Joo Chng ◽  
Shang-Yi Huang ◽  
Cheng-Shyong Chang ◽  
...  

Abstract Introduction: Denosumab is a monoclonal antibody targeting receptor activator of nuclear factor-kappa B ligand (RANKL) that has been shown to reduce skeletal-related events (SREs) associated with bone lesions in patients with multiple myeloma. Results from the full primary analysis of an international, double-blind, double-dummy, randomized controlled phase 3 (20090482) study that assessed the efficacy and safety of denosumab vs zoledronic acid for preventing SREs in patients with multiple myeloma (MM) indicated that denosumab was non-inferior to zoledronic acid for time to SREs. Here we present a sub-analysis to evaluate efficacy and safety outcomes in a subgroup of Asian patients enrolled in the 20090482 study. Methods: Adult patients from Asian countries with newly diagnosed MM and ≥1 documented lytic bone lesion were included in this analysis. Patients received subcutaneous denosumab (120 mg) plus intravenous placebo or intravenous zoledronic acid (4 mg) plus subcutaneous placebo in 4-week cycles. The primary endpoint was time to first on-study SRE; the incidence of adverse events (AEs) by preferred term was also examined. Results: Overall, 196 Asian patients (denosumab, n=103; zoledronic acid, n=93) were included in this analysis. Patient demographics were generally well balanced between groups. Median (interquartile range [IQR]) number of months on study was 17.5 (9.8-30.2) for the denosumab group and 20.2 (13.1-29.2) for the zoledronic acid group. Median (IQR) cumulative drug exposure was 15.9 (8.5-24.0) months for denosumab and 17.4 (9.1-26.7) months for zoledronic acid. Fewer patients in the denosumab group developed first on-study SRE compared with the zoledronic acid group; the crude incidence of SREs at the primary analysis cutoff was 38.8% in the denosumab group and 50.5% in the zoledronic acid group. Median (95% CI) time in months to first on-study SRE was not reached (11.2-not reached) for the denosumab group and 12.3 (3.1-not reached) for the zoledronic acid group (hazard ratio [HR], 0.77; 95% CI, 0.48-1.26; Figure 1). Overall, all patients (100%) experienced ≥1 treatment-emergent AE; the AEs reported in ≥20% of patients in either treatment arm are presented in Table 1. The most common AEs reported in either subgroup (denosumab, zoledronic acid) were diarrhea (51.0%, 51.1%), nausea (42.2%, 46.7%), pyrexia (38.2%, 41.3%), upper respiratory tract infection (37.3%, 40.2%), and constipation (33.3%, 31.5%). Renal toxicity (preferred terms of blood creatinine increased, renal failure, urine output decreased, acute kidney injury, renal impairment, and blood urea decreased) occurred in 9 of 102 (8.8%) patients in the denosumab group and 20 of 92 (21.7%) patients in the zoledronic acid group. Adjudicated osteonecrosis of the jaw was reported in 7 (6.9%) patients in the denosumab group and 5 (5.4%) patients in the zoledronic acid group. Hypocalcemia was reported in 19 (18.6%) patients in the denosumab group and 17 (18.5%) patients in the zoledronic acid group. Conclusion: Results from this Asian subgroup analysis were comparable to those of the full analysis set. In addition, in this analysis there were numerically fewer patients in the denosumab arm that developed a first on-study SRE compared with those in the zoledronic acid arm, and the time to first on-study SRE had a trend favoring the denosumab-treated patients. The AE profiles for denosumab and zoledronic acid in the Asian subgroup were comparable to those observed in the full primary analysis, with renal toxicity similarly reported to be higher in the zoledronic acid group. Overall, this analysis supports that denosumab may be an additional treatment option for the standard of care for Asian patients with newly diagnosed MM with bone disease. Disclosures Chng: Merck: Research Funding; Aslan: Research Funding; Celgene: Consultancy, Honoraria, Other: Travel, accommodation, expenses, Research Funding; Janssen: Consultancy, Honoraria, Other: Travel, accommodation, expenses, Research Funding; Takeda: Consultancy, Honoraria, Other: Travel, accommodation, expenses; Amgen: Consultancy, Honoraria, Other: Travel, accommodation, expenses. Chang:BMS: Consultancy, Speakers Bureau; AbbVie: Consultancy; Janssen: Consultancy, Speakers Bureau; Celgene: Consultancy; Takeda: Consultancy; Roche: Consultancy, Speakers Bureau; Novarits: Consultancy, Speakers Bureau. Wong:Amgen: Consultancy, Research Funding, Speakers Bureau; Bayer: Research Funding, Speakers Bureau; Novartis: Research Funding, Speakers Bureau; Archigen: Research Funding; Baxalta: Research Funding; Pfizer: Research Funding; Apellis: Research Funding; Roche: Research Funding; Boehringer: Research Funding; Ingelheim: Research Funding; AbbVie: Research Funding; Alexion: Consultancy; Astellas: Speakers Bureau. Shimizu:Amgen Inc.: Other: Non-remunerative Position of Influence, Denosumab 20090482 Global Steering Committee Member; Fujimoto Pharmacuetical Corp: Consultancy; Daiichi-Sankyo, Co., Ltd: Consultancy. Gao:Amgen Asia Holding Limited: Employment, Equity Ownership. Glennane:Amgen: Employment, Equity Ownership. Guan:Amgen: Employment, Equity Ownership.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3093-3093
Author(s):  
Hearn Jay Cho ◽  
Deepak Perumal ◽  
Adeeb H Rahman ◽  
Donald Jackson ◽  
Michael Robbins ◽  
...  

The advent of therapeutic monoclonal antibodies (elotuzumab, daratumumab) for multiple myeloma (MM) heralded a new era of immunologic therapy for this disease. Laboratory and clinical data suggest that immune checkpoint inhibitors (ICI) targeting the PD-1/PD-L1 axis are a promising novel strategy, as PD-L1 is commonly expressed on MM cells and a phase 2 clinical trial demonstrated encouraging responses to pembrolizumab (anti-PD-1) in combination with pomalidomide and dexamethasone. However, two late stage MM clinical trials of ICI combined with either lenalidomide or pomalidomide were halted due to safety concerns and lack of efficacy, although subgroup analysis suggested that the overall response rate in subjects with immune related adverse events was higher in subjects who received pembrolizumab + pomalidomide and dexamethasone vs the control group. As a result, further investigation of ICI in MM has largely discontinued. This discrepancy with the positive findings in the early phase trial suggests that genetic or immunologic features of the tumors or immune microenvironment of the patients may influence outcome to ICI, but these factors are as yet undefined. In solid tumors, high mutation burden as assessed by microsatellite sequence instability-high (MSI-high) or mismatch repair genes deficiency (dMMR) was correlated to response to anti-PD-1 ICI, and is now an indication for these agents. Newly diagnosed MM is considered an intermediate-low mutation burden disease (<10 muts/Mb), but a subset of newly diagnosed subjects, often characterized by overexpression of cMAF, have two-to-ten-fold higher average mutation burden. In addition, clonal evolution, characterized by accumulation of somatic mutations, is a common feature of MM and increases the mutation burden through its natural history. We hypothesized that cMAF/hyper-mutated (HM) MM patients may be more likely to have a tumor microenvironment (TME) that is amenable to ICI therapy, analogous to MSI-high/dMMR solid tumor patients. To investigate this, we identified cMAF/HM (n=25) and standard mutation burden (n=20) subjects from the CoMMPass database of newly-diagnosed MM patients. We performed bulk RNA sequencing and mass cytometry (CyTOF) analysis of CD138- bone marrow mononuclear cells, representing the TME, and proteomic (O-link) and grand serology (ELISA-based) analysis of peripheral blood plasma. CyTOF analysis of the TME demonstrated that cMAF/HM subjects had relatively decreased cycling myeloid cells and increased CD4+, CD73low T cells compared to standard. cMAF/HM subjects had significantly higher frequencies of antibody titers against the MM-associated autologous antigens SOX2 and NY-ESO-1. These results suggest that cMAF/HM patients are more likely to feature immune activation in the TME and generate adaptive immune responses to MM-associated antigens, but these responses are not sufficient to control the disease, possibly due to inhibition by checkpoint ligands such as PD-L1. These results support further investigation of ICI-based combination therapies in cMAF/HM MM patients. Disclosures Cho: The Multiple Myeloma Research Foundation: Employment; Celgene: Honoraria, Research Funding; Takeda: Research Funding; Agenus: Research Funding; Genentech: Honoraria, Research Funding; BMS: Consultancy; GSK: Consultancy. Jackson:Bristol-Myers Squibb: Employment, Equity Ownership. Robbins:Bristol-Myers Squibb: Employment, Equity Ownership. Parekh:Karyopharm Inc.: Research Funding; Foundation Medicine Inc.: Consultancy; Celgene Corporation: Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4517-4517 ◽  
Author(s):  
Shaji K. Kumar ◽  
Brian G M Durie ◽  
Zhuo Su ◽  
Joris Diels ◽  
Brian Hutton ◽  
...  

Abstract Objective: To fully contextualize the benefit of novel agents such as daratumumab (DARA) monotherapy for the treatment of patients with heavily pre-treated and highly refractory multiple myeloma (MM), it is critical to understand the real-world outcomes of this patient population on current standard of care (SOC) therapies. The objective of this study was to perform adjusted comparisons to determine the comparative effectiveness of DARA monotherapy versus real-world SOC therapies. Methods: Data for patients treated with DARA 16 mg/kg monotherapy were available from clinical trials MMY2002 (n=106) and GEN501 (n=42), while patients treated with SOC therapies were derived from the International Myeloma Foundation (IMF) chart review of patients with MM who had ≥3 prior lines of therapy and were double refractory to a proteasome inhibitor (PI) and an immunomodulatory drug (IMiD) (n=543) (Kumar et al., ASH 2016; submitted). The pooled DARA studies demonstrated a median overall survival (OS) of 20.1 months versus 13.0 months for SOC based on the IMF cohort (Usmani et al., Blood 2016; Kumar et al., ASH 2016; submitted). The relative treatment effect of DARA versus SOC was estimated using two adjusted comparison methodologies, propensity score matching (PSM) and multivariate Cox regression analyses. Both methodologies utilized individual patient data to compare OS. Modeled covariates for the PSM were age, gender, prior lines of therapy, albumin, and refractory status to bortezomib (BOR), carfilzomib (CAR), lenalidomide (LEN), and pomalidomide (POM). PSM was performed using caliper matching with a caliper width 25% of the standard deviation of the logit-transformed propensity score, using sampling without replacement. For the regression analysis, the covariates included in the multivariate proportional hazards regression model were age, gender, prior lines of therapy, albumin, beta-2 microglobulin, prior exposure to POM and CAR, and PI/IMiD refractory status. Clustering of observations at the treatment-line level within patients was controlled for using the robust sandwich estimate for the covariance matrix, making confidence intervals (CIs) more conservative. For both PSM and regression, statistical significance testing was performed using a two-tailed p-value of <0.05, and all comparisons between treatment groups were reported with hazard ratios (HRs) and 95% CIs. Results: Prior to PSM, imbalances between the DARA and SOC groups were significant for prior lines of therapy and proportions of patients refractory to POM, CAR, BOR, and LEN. After PSM, the DARA and SOC groups were well balanced for all covariates included in propensity score calculations. After PSM, comparisons found significant improvement in favor of DARA relative to SOC for OS (HR=0.44 [95% CI 0.31-0.63]) (Figure 1). Regression analyses revealed consistent results. After adjustment for differences in all covariates included in regression between the DARA and SOC groups, results showed significant improvement in favor of DARA compared with SOC for OS (HR=0.43 [95% CI 0.32-0.59]) (Figure 2). Conclusions: Findings from both PSM and regression analyses were consistent and suggest that DARA is associated with significant gains in OS compared with SOC therapies for patients with heavily pre-treated and highly refractory MM. References: 1. Usmani SZ, Weiss BM, Plesner T, Bahlis NJ, Belch A et al. (2016) Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma. Blood 128 (1): 37-44. 2. Kumar SK, et al. (2016) Natural history of relapsed myeloma, refractory to immunomodulatory drugs and proteasome inhibitors: a multicenter IMWG study. The 58th Annual Meeting of the American Society of Hematology: submitted. Disclosures Kumar: Celgene: Consultancy, Research Funding; Noxxon: Consultancy, Honoraria; Janssen: Research Funding; AbbVie: Research Funding; BMS: Consultancy; Amgen: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Sanofi: Consultancy, Research Funding; Skyline: Consultancy, Honoraria. Durie:Amgen: Consultancy; Takeda: Consultancy; Janssen: Consultancy. Su:Janssen: Research Funding. Diels:Johnson & Johnson: Employment, Equity Ownership. Hutton:Essai Canada: Consultancy; Cornerstone Research Group: Consultancy; Janssen: Research Funding. Lam:Janssen: Employment. Tetsuro:Johnson & Johnson: Equity Ownership; Janssen: Employment.


Sign in / Sign up

Export Citation Format

Share Document