Try a new search

Format these results:

Searched for:

in-biosketch:yes

person:morgag04

Total Results:

655


Long-term outcomes after autologous stem cell transplantation for multiple myeloma

Nishimura, Katherine K; Barlogie, Bart; van Rhee, Frits; Zangari, Maurizio; Walker, Brian A; Rosenthal, Adam; Schinke, Carolina; Thanendrarajan, Sharmilan; Davies, Faith E; Hoering, Antje; Morgan, Gareth J
As multiple myeloma (MM) treatments evolve, frequent updates are required to monitor the long-term effect of changes in approach. Traditionally, MM is considered an incurable disease, with most patients eventually relapsing. However, improvements in treatments has raised the possibility that MM might be functionally curable. To examine improvements in long-term survival, we followed 4329 patients with newly diagnosed MM treated with autologous stem cell transplantation (ASCT) at the University of Arkansas for Medical Sciences from 1989 through 2018. Overall survival (OS) and progression-free survival (PFS) were evaluated using Kaplan-Meier analysis, Cox proportional hazards models, relative survival analysis, and cure modeling among different time periods, risk groups, and demographic traits. Steady improvements in OS were found, with patients treated in 2014 or later having superior OS (hazard ratio, 0.35; 95% confidence interval [CI], 0.27-0.45) and reduced excess risk for MM death (relative excess risk, 0.30; 95% CI, 0.22-0.41) compared with patients treated in 1997 or earlier. Patients treated during intervening time periods often had intermediate survival, but trends in OS, PFS, and landmarked analyses were inconsistent. Cure models support the potential for cure, ranging from 6.3% to 31.3%, depending on the year of treatment, with 10.0% to 18.6% of patients achieving their normal life expectancy across multiple periods. There was some evidence of reductions in early mortality within 3 years of diagnosis, longer complete response (CR) duration, and reductions in relapse after achieving CR. However, results differed depending on age, risk group, and cytogenetic characteristics.
PMCID:6988393
PMID: 31990333
ISSN: 2473-9537
CID: 4294052

A meta-analysis of genome-wide association studies of multiple myeloma among men and women of African ancestry

Du, Zhaohui; Weinhold, Niels; Song, Gregory Chi; Rand, Kristin A; Van Den Berg, David J; Hwang, Amie E; Sheng, Xin; Hom, Victor; Ailawadhi, Sikander; Nooka, Ajay K; Singhal, Seema; Pawlish, Karen; Peters, Edward S; Bock, Cathryn; Mohrbacher, Ann; Stram, Alexander; Berndt, Sonja I; Blot, William J; Casey, Graham; Stevens, Victoria L; Kittles, Rick; Goodman, Phyllis J; Diver, W Ryan; Hennis, Anselm; Nemesure, Barbara; Klein, Eric A; Rybicki, Benjamin A; Stanford, Janet L; Witte, John S; Signorello, Lisa; John, Esther M; Bernstein, Leslie; Stroup, Antoinette M; Stephens, Owen W; Zangari, Maurizio; Van Rhee, Frits; Olshan, Andrew; Zheng, Wei; Hu, Jennifer J; Ziegler, Regina; Nyante, Sarah J; Ingles, Sue Ann; Press, Michael F; Carpten, John David; Chanock, Stephen J; Mehta, Jayesh; Colditz, Graham A; Wolf, Jeffrey; Martin, Thomas G; Tomasson, Michael; Fiala, Mark A; Terebelo, Howard; Janakiraman, Nalini; Kolonel, Laurence; Anderson, Kenneth C; Le Marchand, Loic; Auclair, Daniel; Chiu, Brian C-H; Ziv, Elad; Stram, Daniel; Vij, Ravi; Bernal-Mizrachi, Leon; Morgan, Gareth J; Zonder, Jeffrey A; Huff, Carol Ann; Lonial, Sagar; Orlowski, Robert Z; Conti, David V; Haiman, Christopher A; Cozen, Wendy
Persons of African ancestry (AA) have a twofold higher risk for multiple myeloma (MM) compared with persons of European ancestry (EA). Genome-wide association studies (GWASs) support a genetic contribution to MM etiology in individuals of EA. Little is known about genetic risk factors for MM in individuals of AA. We performed a meta-analysis of 2 GWASs of MM in 1813 cases and 8871 controls and conducted an admixture mapping scan to identify risk alleles. We fine-mapped the 23 known susceptibility loci to find markers that could better capture MM risk in individuals of AA and constructed a polygenic risk score (PRS) to assess the aggregated effect of known MM risk alleles. In GWAS meta-analysis, we identified 2 suggestive novel loci located at 9p24.3 and 9p13.1 at P < 1 × 10-6; however, no genome-wide significant association was noted. In admixture mapping, we observed a genome-wide significant inverse association between local AA at 2p24.1-23.1 and MM risk in AA individuals. Of the 23 known EA risk variants, 20 showed directional consistency, and 9 replicated at P < .05 in AA individuals. In 8 regions, we identified markers that better capture MM risk in persons with AA. AA individuals with a PRS in the top 10% had a 1.82-fold (95% confidence interval, 1.56-2.11) increased MM risk compared with those with average risk (25%-75%). The strongest functional association was between the risk allele for variant rs56219066 at 5q15 and lower ELL2 expression (P = 5.1 × 10-12). Our study shows that common genetic variation contributes to MM risk in individuals with AA.
PMCID:6960456
PMID: 31935283
ISSN: 2473-9537
CID: 4334842

Deep sequencing as an approach to understanding the complexity and improving the treatment of multiple myeloma [Review]

Williams, Louis S.; Caro, Jessica; Razzo, Beatrice; Boyle, Eileen M.; Morgan, Gareth J.
ISI:000552123500001
ISSN: 2380-8993
CID: 5389102

International harmonization in performing and reporting minimal residual disease assessment in multiple myeloma trials [Review]

Costa, Luciano J.; Derman, Benjamin A.; Bal, Susan; Sidana, Surbhi; Chhabra, Saurabh; Silbermann, Rebecca; Ye, Jing C.; Cook, Gordon; Cornell, Robert F.; Holstein, Sarah A.; Shi, Qian; Omel, James; Callander, Natalie S.; Chng, Wee Joo; Hungria, Vania; Maiolino, Angelo; Stadtmauer, Edward; Giralt, Sergio; Pasquini, Marcelo; Jakubowiak, Andrzej J.; Morgan, Gareth J.; Krishnan, Amrita; Jackson, Graham H.; Mohty, Mohamad; Mateos, Maria Victoria; Dimopoulos, Meletious A.; Facon, Thierry; Spencer, Andrew; San Miguel, Jesus; Hari, Parameswaran; Usmani, Saad Z.; Manier, Salomon; McCarthy, Phillip; Kumar, Shaji; Gay, Francesca; Paiva, Bruno
ISI:000558138400001
ISSN: 0887-6924
CID: 4573572

Influence of Aging Processes on the Biology and Outcome of Multiple Myeloma [Meeting Abstract]

Boyle, Eileen M.; Williams, Louis; Blaney, Patrick; Ashby, Cody; Bauer, Michael A.; Walker, Brian A.; Choi, Jinyoung; Caro, Jessica; Razzo, Beatrice; Arbini, Arnaldo A.; Kaminetzky, David; Braunstein, Marc; Maura, Francesco; Wang, Yubao; Landgren, Ola; Stoeckle, James; Maclachlan, Kylee H.; Litke, Rachel; Davies, Faith E.; Morgan, Gareth
ISI:000607547201239
ISSN: 0006-4971
CID: 5389132

Response-adapted intensification with cyclophosphamide, bortezomib, and dexamethasone versus no intensification in patients with newly diagnosed multiple myeloma (Myeloma XI): a multicentre, open-label, randomised, phase 3 trial

Jackson, Graham H; Davies, Faith E; Pawlyn, Charlotte; Cairns, David A; Striha, Alina; Collett, Corinne; Waterhouse, Anna; Jones, John R; Kishore, Bhuvan; Garg, Mamta; Williams, Cathy D; Karunanithi, Kamaraj; Lindsay, Jindriska; Wilson, Jamie N; Jenner, Matthew W; Cook, Gordon; Kaiser, Martin F; Drayson, Mark T; Owen, Roger G; Russell, Nigel H; Gregory, Walter M; Morgan, Gareth J
BACKGROUND:Multiple myeloma has been shown to have substantial clonal heterogeneity, suggesting that agents with different mechanisms of action might be required to induce deep responses and improve outcomes. Such agents could be given in combination or in sequence on the basis of previous response. We aimed to assess the clinical value of maximising responses by using therapeutic agents with different modes of action, the use of which is directed by the response to the initial combination therapy. We aimed to assess response-adapted intensification treatment with cyclophosphamide, bortezomib, and dexamethasone (CVD) versus no intensification treatment in patients with newly diagnosed multiple myeloma who had a suboptimal response to initial immunomodulatory triplet treatment which was standard of care in the UK at the time of trial design. METHODS:subcutaneously or intravenously on days 1, 4, 8, and 11), and dexamethasone (20 mg daily orally on days 1, 2, 4, 5, 8, 9, 11, and 12) up to a maximum of eight cycles of 21 days or no treatment. Patients were stratified by allocated induction treatment, response to induction treatment, and centre. The co-primary endpoints were progression-free survival and overall survival, assessed from intensification randomisation to data cutoff, analysed by intention to treat. Safety analysis was per protocol. This study is registered with the ISRCTN registry, number ISRCTN49407852, and clinicaltrialsregister.eu, number 2009-010956-93, and has completed recruitment. FINDINGS/RESULTS:Between Nov 15, 2010, and July 28, 2016, 583 patients were enrolled to the intensification randomisation, representing 48% of the 1217 patients who achieved partial or minimal response after initial induction therapy. 289 patients were assigned to CVD treatment and 294 patients to no treatment. After a median follow-up of 29·7 months (IQR 17·0-43·5), median progression-free survival was 30 months (95% CI 25-36) with CVD and 20 months (15-28) with no CVD (hazard ratio [HR] 0·60, 95% CI 0·48-0·75, p<0·0001), and 3-year overall survival was 77·3% (95% Cl 71·0-83·5) in the CVD group and 78·5% (72·3-84·6) in the no CVD group (HR 0·98, 95% CI 0·67-1·43, p=0·93). The most common grade 3 or 4 adverse events for patients taking CVD were haematological, including neutropenia (18 [7%] patients), thrombocytopenia (19 [7%] patients), and anaemia (8 [3%] patients). No deaths in the CVD group were deemed treatment related. INTERPRETATION/CONCLUSIONS:Intensification treatment with CVD significantly improved progression-free survival in patients with newly diagnosed multiple myeloma and a suboptimal response to immunomodulatory induction therapy compared with no intensification treatment, but did not improve overall survival. The manageable safety profile of this combination and the encouraging results support further investigation of response-adapted approaches in this setting. The substantial number of patients not entering this trial randomisation following induction therapy, however, might support the use of combination therapies upfront to maximise response and improve outcomes as is now the standard of care in the UK. FUNDING/BACKGROUND:Cancer Research UK, Celgene, Amgen, Merck, Myeloma UK.
PMID: 31624047
ISSN: 2352-3026
CID: 4178382

Phenome-wide association analysis of LDL-cholesterol lowering genetic variants in PCSK9

Schmidt, Amand F; Holmes, Michael V; Preiss, David; Swerdlow, Daniel I; Denaxas, Spiros; Fatemifar, Ghazaleh; Faraway, Rupert; Finan, Chris; Valentine, Dennis; Fairhurst-Hunter, Zammy; Hartwig, Fernando Pires; Horta, Bernardo Lessa; Hypponen, Elina; Power, Christine; Moldovan, Max; van Iperen, Erik; Hovingh, Kees; Demuth, Ilja; Norman, Kristina; Steinhagen-Thiessen, Elisabeth; Demuth, Juri; Bertram, Lars; Lill, Christina M; Coassin, Stefan; Willeit, Johann; Kiechl, Stefan; Willeit, Karin; Mason, Dan; Wright, John; Morris, Richard; Wanamethee, Goya; Whincup, Peter; Ben-Shlomo, Yoav; McLachlan, Stela; Price, Jackie F; Kivimaki, Mika; Welch, Catherine; Sanchez-Galvez, Adelaida; Marques-Vidal, Pedro; Nicolaides, Andrew; Panayiotou, Andrie G; Onland-Moret, N Charlotte; van der Schouw, Yvonne T; Matullo, Giuseppe; Fiorito, Giovanni; Guarrera, Simonetta; Sacerdote, Carlotta; Wareham, Nicholas J; Langenberg, Claudia; Scott, Robert A; Luan, Jian'an; Bobak, Martin; Malyutina, Sofia; Pająk, Andrzej; Kubinova, Ruzena; Tamosiunas, Abdonas; Pikhart, Hynek; Grarup, Niels; Pedersen, Oluf; Hansen, Torben; Linneberg, Allan; Jess, Tine; Cooper, Jackie; Humphries, Steve E; Brilliant, Murray; Kitchner, Terrie; Hakonarson, Hakon; Carrell, David S; McCarty, Catherine A; Lester, Kirchner H; Larson, Eric B; Crosslin, David R; de Andrade, Mariza; Roden, Dan M; Denny, Joshua C; Carty, Cara; Hancock, Stephen; Attia, John; Holliday, Elizabeth; Scott, Rodney; Schofield, Peter; O'Donnell, Martin; Yusuf, Salim; Chong, Michael; Pare, Guillaume; van der Harst, Pim; Said, M Abdullah; Eppinga, Ruben N; Verweij, Niek; Snieder, Harold; Christen, Tim; Mook-Kanamori, D O; Gustafsson, Stefan; Lind, Lars; Ingelsson, Erik; Pazoki, Raha; Franco, Oscar; Hofman, Albert; Uitterlinden, Andre; Dehghan, Abbas; Teumer, Alexander; Baumeister, Sebastian; Dörr, Marcus; Lerch, Markus M; Völker, Uwe; Völzke, Henry; Ward, Joey; Pell, Jill P; Meade, Tom; Christophersen, Ingrid E; Maitland-van der Zee, Anke H; Baranova, Ekaterina V; Young, Robin; Ford, Ian; Campbell, Archie; Padmanabhan, Sandosh; Bots, Michiel L; Grobbee, Diederick E; Froguel, Philippe; Thuillier, Dorothée; Roussel, Ronan; Bonnefond, Amélie; Cariou, Bertrand; Smart, Melissa; Bao, Yanchun; Kumari, Meena; Mahajan, Anubha; Hopewell, Jemma C; Seshadri, Sudha; Dale, Caroline; Costa, Rui Providencia E; Ridker, Paul M; Chasman, Daniel I; Reiner, Alex P; Ritchie, Marylyn D; Lange, Leslie A; Cornish, Alex J; Dobbins, Sara E; Hemminki, Kari; Kinnersley, Ben; Sanson, Marc; Labreche, Karim; Simon, Matthias; Bondy, Melissa; Law, Philip; Speedy, Helen; Allan, James; Li, Ni; Went, Molly; Weinhold, Niels; Morgan, Gareth; Sonneveld, Pieter; Nilsson, Björn; Goldschmidt, Hartmut; Sud, Amit; Engert, Andreas; Hansson, Markus; Hemingway, Harry; Asselbergs, Folkert W; Patel, Riyaz S; Keating, Brendan J; Sattar, Naveed; Houlston, Richard; Casas, Juan P; Hingorani, Aroon D
BACKGROUND:We characterised the phenotypic consequence of genetic variation at the PCSK9 locus and compared findings with recent trials of pharmacological inhibitors of PCSK9. METHODS:Published and individual participant level data (300,000+ participants) were combined to construct a weighted PCSK9 gene-centric score (GS). Seventeen randomized placebo controlled PCSK9 inhibitor trials were included, providing data on 79,578 participants. Results were scaled to a one mmol/L lower LDL-C concentration. RESULTS:The PCSK9 GS (comprising 4 SNPs) associations with plasma lipid and apolipoprotein levels were consistent in direction with treatment effects. The GS odds ratio (OR) for myocardial infarction (MI) was 0.53 (95% CI 0.42; 0.68), compared to a PCSK9 inhibitor effect of 0.90 (95% CI 0.86; 0.93). For ischemic stroke ORs were 0.84 (95% CI 0.57; 1.22) for the GS, compared to 0.85 (95% CI 0.78; 0.93) in the drug trials. ORs with type 2 diabetes mellitus (T2DM) were 1.29 (95% CI 1.11; 1.50) for the GS, as compared to 1.00 (95% CI 0.96; 1.04) for incident T2DM in PCSK9 inhibitor trials. No genetic associations were observed for cancer, heart failure, atrial fibrillation, chronic obstructive pulmonary disease, or Alzheimer's disease - outcomes for which large-scale trial data were unavailable. CONCLUSIONS:Genetic variation at the PCSK9 locus recapitulates the effects of therapeutic inhibition of PCSK9 on major blood lipid fractions and MI. While indicating an increased risk of T2DM, no other possible safety concerns were shown; although precision was moderate.
PMCID:6820948
PMID: 31664920
ISSN: 1471-2261
CID: 5478742

Circulating cell free DNA is a biomarker for GEP70 risk score and tumor burden in myeloma [Meeting Abstract]

Deshpande, S; Wang, Y; Tytarenko, R; Ashby, C; Boyle, E; Schinke, C; Thanendrarajan, S; Zangari, M; Davies, F; Morgan, G; van, Rhee F; Walker, B
Background: Multiple myeloma (MM) is a heterogeneous disease with variable outcomes. In the past several years, serum proteins, cytogenetics and gene expression profiling (GEP) have been used to predict outcomes of MM patients. Patients identified by the prognostic GEP70 gene signature as high risk (HR) have more aggressive disease and shorter survival compared to patients identified as low risk (LR). We have analyzed cfDNA levels in LR and HR patients and determined its correlation to GEP70 risk score. In a subset of patients, we performed low-pass whole genome sequencing (LP-WGS) and targeted sequencing to determine genomic alterations in cfDNA.
Method(s): GEP was performed on CD138+ plasma cells from bone marrow using Affymetrix U133 Plus 2.0 arrays. Low-risk (n=38) and high-risk MM patients (n=44) as determined by GEP70 score were selected. Plasma cells in bone marrow aspirate were ascertained for tumor burden by flow cytometry. cfDNA was extracted from plasma using QIAamp circulating nucleic acid kit (Qiagen) and was measured using Qubit fluorometer. In a subset of patients, plasma cfDNA, matched tumor DNA and matched white blood cell genomic DNA were sequenced using a targeted panel covering key driver genes and immunoglobulin regions involved in translocations in myeloma. Targeted sequencing was performed on NextSeq500 (Illumina) to a depth of 400-600x. LP-WGS was performed at 0.1X coverage. Sequencing data were analyzed using Strelka and ichor.
Result(s): Total cfDNA (ng/ml plasma) was significantly higher in the HR group compared to the LR group, median cfDNA LR=18.76 ng/ml range 0.2-140 ng/ml plasma vs. HR=33 ng/ml plasma range 7.3-726.66 ng/ml; p=0.02. cfDNA levels among different GEP subgroups did not reach significance, however patients in the PR subgroup had higher cfDNA in plasma compared to other subgroups. Ranked cfDNA levels correlated with GEP risk score r=0.32, p=0.0029 (Spearman's test). cfDNA levels also correlated with tumor burden r=0.41, p<0.0001 (Pearson's test). Additionally, LP-WGS analysis performed in a subset of patients showed that circulating tumor DNA (ctDNA) fraction correlated strongly with GEP70 risk score (Spearman r=0.83, p=0.0012). Monitoring of cfDNA levels in patients before and after chemotherapy showed an increase in cfDNA levels between 3-5 days after chemotherapy and fell to baseline levels a week later. Variant allele frequencies of NRAS and KRAS mutations were higher immediately post-chemotherapy compared to baseline in 3/4 patients.
Conclusion(s): cfDNA levels correlate with GEP70 risk score. In the small subset of sequenced patients, ctDNA fraction also correlated with GEP70 risk score. Molecular monitoring of myeloma using cfDNA can capture the mutational landscape in myeloma and can be a potential prognostic biomarker for MM disease. Keywords: cell-free DNA Gene expression profiling Whole genome sequencing Tracks: Multiple Myeloma Genomics
Copyright
EMBASE:2003393965
ISSN: 2152-2669
CID: 4152932

Preclinical evaluation of the new GPRC5DxCD3 (JNJ-7564) bispecific antibody for the treatment of multiple myeloma [Meeting Abstract]

Verkleij, C; Broekmans, M; van, Duin M; Frerichs, K; Kuiper, R; Kaiser, M; Morgan, G; Axel, A; Chiu, C; Sonneveld, P; Zweegman, S; Adams, H; Mutis, T; Van, de Donk N
Background: The prognosis of multiple myeloma (MM) patients who are refractory to all currently available therapies, including anti-CD38 monoclonal antibodies, remains poor, indicating that there is an unmet need for novel treatment strategies. Although data on G protein-coupled receptor 5D (GPRC5D) protein expression is scarce, RNA levels are selectively elevated in MM cell lines and primary MM cells. This makes GPRC5D an attractive target for the novel anti-myeloma JNJ-7564 bispecific antibody.
Method(s): We determined GPRC5D protein expression levels on MM cell lines and bone marrow (BM) mononuclear cells (MNCs) derived from healthy donors (HD) and MM patients by flow cytometry. We also analyzed the impact of differential GPRC5D gene expression in purified CD138+ MM cells derived from patients who participated in 5 large randomized clinical trials (HOVON65, MRC-IX, TT2, TT3 and APEX), on overall survival (OS) and progression free survival (PFS). Furthermore, the activity of a new GPRC5DxCD3 bispecific antibody (JNJ-7564) was evaluated in MM cell lines and patient-derived whole BM samples after 48 hours of incubation (concentrations 0.00064-4mug/ml), as well as biomarkers for in vitro JNJ-7564 response. At baseline, MNCs were characterized for the composition of T-cell subsets.
Result(s): GPRC5D protein expression was significantly higher on MM cells compared to HD plasma cells and immune cells. Gene expression levels of GPRC5D were highly variable in MM, but were significantly higher in patients with t(4;14) or gain 1q. There was no association with OS or PFS in the clinical trials (n=1421). JNJ-7564 effectively killed GPRC5D+ MM cell lines (MM1.S, UM9 and RPMI-8226) in a dose-dependent manner, using HD or patient-derived blood MNCs as effector cells. Co-incubation with patient-derived BM stromal cells resulted in a modest impairment of killing capacity in 2 out of 3 cell lines. In MM patient samples (n=29), the mean lysis of MM cells with 4.0mug/mL JNJ-7564 was 57% (range: -8-97%), while NK-cell and T-cell frequencies were not affected. JNJ-7564 was also active in samples from extensively pretreated and daratumumab (DARA)-refractory patients (n=8; mean lysis with 4.0mug/mL: 59%; range: 22-97%). JNJ-7564-mediated MM cell lysis was associated with activation (CD25+) and degranulation (CD107a+) of CD4+ and CD8+ T-cells. Maximum kill was correlated with GPRC5D expression on MM cells (Spearman r=0.40, p=0.029), % of regulatory T-cells (r=-0.41, p=0.023) and % of PD-1+ T-cells (r=-0.43, p=0.033).
Conclusion(s): GPRC5D is highly and selectively expressed on MM cells, which makes it an attractive therapeutic target. The GPRC5DxCD3 bispecific antibody, JNJ-7564, effectively lysed MM cell lines and primary MM cells in samples derived from both newly diagnosed and heavily pretreated patients, including DARA-refractory patients. Altogether, this strengthens the preclinical rationale for an ongoing phase 1 study with JNJ-7564 in relapsed/refractory MM. Keywords: bispecific antibody immunotherapy Multiple myeloma Tracks: Multiple Myeloma Novel Agents
Copyright
EMBASE:2003392611
ISSN: 2152-2669
CID: 4152962

Quadruplet KCRD (Carfilzomib, Cyclophosphamide, Lenalidomide and Dexamethasone) Induction for Newly Diagnosed Myeloma Patients [Meeting Abstract]

Pawlyn, C; Davies, F; Cairns, D; Striha, A; Hockaday, A; Kishore, B; Garg, M; Williams, C; Karunanithi, K; Lindsay, J; Jones, J; Jenner, M; Russell, N; Cook, G; Kaiser, M; Drayson, M; Owen, R; Gregory, W; Morgan, G; Jackson, G
Background: Multiple myeloma (MM) has significant spatial and temporal clonal heterogeneity suggesting therapeutic agents with different mechanisms of action, delivered in combination or sequentially, are required to maximize the depth of response and improve outcomes. The UK NCRI Myeloma XI phase III randomized trial compared induction with the second generation proteasome inhibitor carfilzomib and lenalidomide containing quadruplet, KCRD, vs a response-adapted approach of sequential triplet therapies in newly diagnosed transplant eligible patients.
Method(s): 1056 patients were randomized between KCRD (28 day cycles of carfilzomib (K) 36mg/m2 IV d1-2,8-9,15-16, cyclophosphamide (C) 500mg PO d1,8, lenalidomide (R) 25mg PO d1-21, dexamethasone (D) 40mg PO d1-4,8-9,15-16) and immunomodulatory drug (IMiD) triplet CTD/CRD prior to ASCT. Patients with a suboptimal response to CTD/CRD underwent response-adapted intensification randomization to a proteasome inhibitor (bortezomib, CVD) containing triplet or no CVD. A maintenance randomization at 3 months post ASCT compared lenalidomide to observation. Molecular high-risk (HiR) was classified by t(4;14), t(14;16), t(14;20), del(17p) or gain(1q) with ultra-high risk (UHiR) the presence of >1 lesions.
Result(s): KCRD was associated with a significantly longer PFS than IMiD triplet therapy (HR 0.63, 95%CI 0.51, 0.76, 3yr PFS KCRD 64.5% vs CTD/CRD 50.3%, p<0.0001). PFS2 was also significantly improved with KCRD (HR 0.75, 95% CI 0.56, 0.99, 3yr PFS2 KCRD 81.8% vs CTD/CRD 75.1%). Deeper response rates were seen in patients treated with KCRD vs CTD/CRD pre and post-transplant (p<0.0001). All regimens were well tolerated with no significant additional toxicity due to the quadruplet regimen. A higher proportion of patients receiving KCRD induction were able to undergo ASCT than those who received response-adapted induction and in an analysis restricted to those who had completed ASCT, KCRD induction was still associated with a significantly longer PFS. There was no significant heterogeneity in PFS outcome between molecular risk groups with a benefit for KCRD over triplets in all. In patients receiving KCRD there was no difference in response rate at the end of initial induction by risk group but UHiR disease was associated with significantly shorter PFS than both SR and HiR, whilst there was no difference in outcome between patients with HiR (one adverse lesion only) and SR. An exploratory analysis compared the patients receiving KCRD to patients in the CTD/CRD arm who had received the optimum response-adapted approach (i.e. excluding those with a suboptimal response randomized to no CVD). KCRD was associated with significantly longer PFS than using a response adapted sequential triplet approach (HR 0.64, 95% CI 0.52, 0.78, p<0.0001).
Conclusion(s): KCRD was well tolerated with deep responses pre- and post-transplant and a significant PFS benefit compared to triplet therapy across all risk groups. Keywords: carfilzomib carfilzomib-lenalidomide-dexamethasone Lenalidomide Tracks: Treatment of Newly Diagnosed Myeloma Transplant Eligible
Copyright
EMBASE:2003393915
ISSN: 2152-2669
CID: 4152952