68 results on '"Sborov DW"'
Search Results
2. Rapid growth of acquired UBA1 mutations predisposes male patients to low-risk MDS.
- Author
-
Li P, Alnoor FNU, Xie W, Williams M, Feusier J, Ding Y, Zhao X, Zheng G, Zhao C, Zieske AW, Zu Y, Raess PW, Tantravahi S, Osman A, Patel AB, Tashi T, Patel JL, Matynia AP, Menon MP, Miles RR, Jacobsen JR, George TI, Sborov DW, Szankasi P, Rindler P, Close D, and Ohgami RS
- Published
- 2024
- Full Text
- View/download PDF
3. A plain language summary of the final analysis of the GRIFFIN study of daratumumab plus lenalidomide, bortezomib, and dexamethasone for people with newly diagnosed multiple myeloma.
- Author
-
Voorhees PM, Sborov DW, Laubach J, Kaufman JL, Reeves B, Rodriguez C, Silbermann R, Costa LJ, Anderson LD, Nathwani N, Shah N, Bumma N, Efebera YA, Holstein SA, Costello C, Jakubowiak A, Wildes TM, Orlowski RZ, Shain KH, Cowan AJ, Dinner S, Gries KS, Pei H, Cortoos A, Patel S, Lin TS, Usmani SZ, and Richardson PG
- Abstract
What Is This Summary About?: This summary describes the final analysis of the GRIFFIN study. In this study, participants were newly diagnosed with a type of blood and bone marrow cancer called multiple myeloma, had never received any treatment, and were able to undergo an autologous stem cell transplant. The GRIFFIN study looked at adding the drug daratumumab (D) to a combination of standard treatments called RVd (lenalidomide [R], bortezomib [V], and dexamethasone [d]) during the treatment phases induction and consolidation, followed by daratumumab and lenalidomide (D-R) maintenance. Participants also received an autologous stem cell transplant to further help reduce multiple myeloma. The GRIFFIN study looked at whether D-RVd followed by D-R maintenance was better at killing multiple myeloma cells compared with RVd on its own followed by R maintenance on its own, and if treatments were safe. This summary also describes results from 2 other GRIFFIN publications: one that looked at participants with certain multiple myeloma characteristics or demographic factors that are associated with worse outcomes, and another that looked at how treatments impacted the participants' quality of life., What Were the Results?: At the time of the final analysis of GRIFFIN, participants who were treated with D-RVd followed by D-R maintenance had very low (undetectable) levels of multiple myeloma cells and multiple myeloma markers (biological signs) and were more likely to be alive without the multiple myeloma getting worse or coming back compared with participants who received standard RVd treatment followed by R maintenance. There was also a pattern of similar benefits achieved by participants who were at risk for worse outcomes. Additionally, participants who received D-RVd treatment followed by D-R maintenance reported less pain, less fatigue (extreme tiredness), and greater improvements in their ability to conduct daily physical activities. While some side effects (unwanted or unexpected effects of treatment) were higher with D-RVd, side effects in both groups were as expected, and adding daratumumab did not reduce a participant's ability to handle treatment., What Do the Results of the Study Mean?: Results of the GRIFFIN study showed that D-RVd treatment followed by D-R maintenance was better at treating multiple myeloma than the standard treatment of RVd followed by R maintenance in adults with a new diagnosis of multiple myeloma who were able to receive an autologous stem cell transplant, with no unexpected side effects of treatment. Clinical Trial Registration: NCT02874742 (GRIFFIN) (ClinicalTrials.gov).
- Published
- 2024
- Full Text
- View/download PDF
4. Immune effector cell-associated enterocolitis following chimeric antigen receptor T-cell therapy in multiple myeloma.
- Author
-
Fortuna GG, Banerjee R, Savid-Frontera C, Song J, Morán-Segura CM, Nguyen JV, Lekakis L, Fernandez-Pol S, Samraj AN, Naresh KN, Vazquez-Martinez M, Baz RC, Spiegel JY, Mikkilineni L, Gubatan JM, Sidana S, de Menezes Silva Corraes A, Kalariya NM, Patel KK, Shim KG, Fonseca R, Ferreri C, Voorhees PM, Richard S, Valdes CR, Sireesha Asoori, Wolf JL, Cowan AJ, Sborov DW, Locke FL, Lin Y, Wang Y, and Hansen DK
- Subjects
- Humans, Male, Middle Aged, Female, Aged, Adult, Receptors, Chimeric Antigen therapeutic use, Receptors, Chimeric Antigen immunology, Multiple Myeloma therapy, Multiple Myeloma immunology, Multiple Myeloma drug therapy, Enterocolitis etiology, Enterocolitis therapy, Enterocolitis immunology, Immunotherapy, Adoptive adverse effects
- Abstract
We report 14 cases of immune effector cell (IEC)-associated enterocolitis following chimeric antigen receptor T-cell (CAR-T) therapy in multiple myeloma, with a 1.2% incidence overall (0.2% for idecabtagene vicleucel and 2.2% for ciltacabtagene autoleucel). Patients developed acute-onset symptoms (typically non-bloody Grade 3+ diarrhea) with negative infectious workup beginning a median of 92.5 days (range: 22-210 days) after CAR-T therapy and a median of 85 days after cytokine release syndrome resolution. Gut biopsies uniformly demonstrated inflammation, including intra-epithelial lymphocytosis and villous blunting. In one case where CAR-specific immunofluorescence stains were available, CAR T-cell presence was confirmed within the lamina propria. Systemic corticosteroids were initiated in 10 patients (71%) a median of 25.5 days following symptom onset, with symptom improvement in 40%. Subsequent infliximab or vedolizumab led to improvement in 50% and 33% of corticosteroid-refractory patients, respectively. Five patients (36%) have died from bowel perforation or treatment-emergent sepsis. In conclusion, IEC-associated enterocolitis is a distinct but rare complication of CAR-T therapy typically beginning 1-3 months after infusion. Thorough diagnostic workup is essential, including evaluation for potential T-cell malignancies. The early use of infliximab or vedolizumab may potentially hasten symptom resolution and lower reliance on high-dose corticosteroids during the post-CAR-T period., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Safety and Efficacy of Standard of Care Ciltacabtagene Autoleucel for Relapsed/Refractory Multiple Myeloma.
- Author
-
Sidana S, Patel KK, Peres LC, Bansal R, Kocoglu MH, Shune L, Atrash S, Smith K, Midha S, Ferreri CJ, Dhakal B, Dima D, Costello P, Wagner C, Reshef R, Hosoya H, Mikkilineni L, Atanackovic D, Chhabra S, Parrondo RD, Nadeem O, Mann H, Kalariya N, Hovanky V, DeAvila G, Freeman CL, Locke FL, Alsina M, Wong S, Herr MM, Htut M, McGuirk JP, Sborov DW, Khouri J, Martin T, Janakiram M, Lin Y, and Hansen DK
- Abstract
Ciltacabtagene autoleucel (cilta-cel) CAR-T therapy was approved in 2022 for patients with relapsed/refractory multiple myeloma (RRMM). We report outcomes with cilta-cel in the standard-of-care setting. Patients with RRMM who underwent leukapheresis for cilta-cel manufacturing between 3/1/2022-12/31/2022 at 16 US academic medical centers were included. RESULTS: 255 patients underwent leukapheresis and 236 (92.5%) received cilta-cel. Of leukapheresed patients, 56% would not have met CARTITUDE-1 trial eligibility criteria. Manufacturing failure rates at first attempt and overall were 6% and 1%, respectively. Median prior lines of therapy were 6. In treated patients (N=236), cytokine release syndrome was seen in 75% (>= grade 3: 5%), immune effector cell-associated neurotoxicity syndrome in 14% (>= grade 3: 4%), and delayed neurotoxicity in 10%. Best overall and >= CR rates were as follows: infused patients (N=236): 89% and 70%; patients receiving conforming CAR-T product (N=191) 94% and 74%; conforming CAR-T product with fludarabine/cyclophosphamide lymphodepletion (N=152): 95% and 76%, respectively. Non-relapse mortality was 10%, most commonly from infection. After median follow-up of 13 months from CAR-T, median progression-free survival (PFS) was not reached, with 12- month estimate being 68% (95% CI: 62-74%). High ferritin levels, high-risk cytogenetics, and extramedullary disease were independently associated with inferior PFS, with a signal for prior BCMA-TT (p=0.08). Second primary malignancies (SPMs) excluding non-melanoma skin cancers were seen in 5.5% and myeloid malignancies/acute leukemia in 1.7%. We observed a favorable efficacy profile of standard of care cilta-cel in RRMM despite more than half the patients not meeting CARTITUDE-1 eligibility criteria., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
6. Daratumumab with lenalidomide as maintenance after transplant in newly diagnosed multiple myeloma: the AURIGA study.
- Author
-
Badros AZ, Foster L, Anderson LD Jr, Chaulagain CP, Pettijohn EM, Cowan AJ, Costello CL, Larson S, Sborov DW, Shain KH, Silbermann R, Shah N, Chung A, Krevvata M, Pei H, Patel S, Khare V, Cortoos A, Carson R, Lin T, and Voorhees PM
- Abstract
No randomized trial has directly compared daratumumab and lenalidomide (D-R) maintenance therapy versus standard-of-care lenalidomide (R) alone post-transplant. Here, we report the primary results of the phase 3 AURIGA study evaluating D-R versus R maintenance in NDMM patients who were in ≥very good partial response, minimal residual disease (MRD; threshold 10-5) positive, and anti-CD38 naïve post-transplant. Patients were randomized 1:1 to D-R or R maintenance for up to 36 cycles. Two hundred patients were randomized (D-R, n=99; R, n=101). The primary endpoint, MRD-negative (10-5) conversion rate by 12 months from start of maintenance, was significantly higher for D-R versus R (50.5% vs 18.8%; odds ratio [OR], 4.51; 95% confidence interval [CI], 2.37-8.57; P<0.0001). MRD-negative (10-6) conversion rate was similarly higher with D-R (23.2% vs 5.0%; OR, 5.97; 95% CI, 2.15-16.58; P=0.0002). At 32.3 months' median follow-up, D-R achieved a higher overall MRD-negative (10-5) conversion rate (D-R, 60.6% vs R, 27.7%; OR, 4.12; 95% CI, 2.26-7.52; P<0.0001) and ≥complete response rate (75.8% vs 61.4%; OR, 2.00; 95% CI, 1.08-3.69; P=0.0255) versus R alone. Progression-free survival (PFS) favored D-R versus R (hazard ratio, 0.53; 95% CI, 0.29-0.97); estimated 30-month PFS rates were 82.7% for D-R and 66.4% for R. Incidences of grade 3/4 cytopenias (54.2% vs 46.9%) and infections (18.8% vs 13.3%) were slightly higher with D-R versus R. In conclusion, D-R maintenance achieved a higher MRD-negative conversion rate and improved PFS post-transplant versus R alone, with no new safety concerns. This trial was registered at www.ClinicalTrials.gov: #NCT03901963., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
7. Multiple myeloma clinical trials exclude patients with the highest-risk disease: a systematic review of trial exclusion criteria.
- Author
-
Zhukovsky S, White J, Chakraborty R, Costa LJ, Van Oekelen O, Sborov DW, Cliff ERS, and Mohyuddin GR
- Abstract
Patients with certain subsets of multiple myeloma continue to have poor outcomes and are in need of novel treatment approaches. Strict eligibility criteria for randomized controlled trials (RCTs) limit access to clinical trials and limit the external validity of trial results for these patients. We systematically reviewed RCTs in newly diagnosed myeloma from 2006 to 2023 to ascertain the prevalence of 12 key exclusion criteria and trends over time. 80 RCTs were included. Exclusion criteria included: age in 43 (51%) trials; projected life expectancy in 20 (24%); performance status in 74 (87%); non-secretory and/or oligosecretory disease in 47 (55%), hepatic function in 64 (79%), renal function in 63 (74%), hematological thresholds in 50 (59%), prior malignancy in 68 (80%), and neuropathy in 50 (59%). For 53 trials which had detailed exclusion criteria available, plasma cell leukemia was excluded in 21 (40%), extramedullary disease in 5 (9%) and CNS disease in 13 (25%). The percentage of studies invoking each of these exclusion criteria did not significantly improve over time on univariate regression analysis, and exclusion criteria relating to neuropathy have worsened. The restrictive eligibility criteria of most myeloma RCTs perpetuate a cycle where limited data exists to treat challenging myeloma subtypes.
- Published
- 2024
- Full Text
- View/download PDF
8. Risk-adapted treatment in multiple myeloma: Does more make it merrier?
- Author
-
Zanwar S, Galarza Fortuna GM, and Sborov DW
- Subjects
- Humans, Risk Assessment, Practice Guidelines as Topic, Multiple Myeloma therapy, Multiple Myeloma diagnosis
- Abstract
Kaiser et al. offer management recommendations for transplant-eligible, high-risk multiple myeloma (HRMM), derived from recent trials exploring treatment intensification in the various phases of front-line therapy. The definition of HRMM continues to evolve with emergence of novel genomic insights and impact of modern therapies, underscoring the need to expand beyond traditional interphase fluorescence in situ hybridization cytogenetics and International Staging System staging for a precise risk assessment. Despite progress, ongoing challenges in treatment delivery and tolerability underscore the urgency for exploring novel approaches like T-cell redirecting bispecific antibodies and chimeric antigen receptor T-cell to enhance outcomes in this complex patient population. Commentary on: Kaiser et al. Diagnosis and initial treatment of transplant-eligible high-risk myeloma patients: A British Society for Haematology/UK Myeloma Society Good Practice Paper. Br J Haematol 2024; 205:833-839., (© 2024 The Author(s). British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
9. Long term responders in frontline multiple myeloma-exception vs expectation of the modern era.
- Author
-
Baljevic M, Sborov DW, and Kumar SK
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Treatment Outcome, Adult, Multiple Myeloma therapy
- Published
- 2024
- Full Text
- View/download PDF
10. Daratumumab in transplant-eligible patients with newly diagnosed multiple myeloma: final analysis of clinically relevant subgroups in GRIFFIN.
- Author
-
Chari A, Kaufman JL, Laubach J, Sborov DW, Reeves B, Rodriguez C, Silbermann R, Costa LJ, Anderson LD Jr, Nathwani N, Shah N, Bumma N, Holstein SA, Costello C, Jakubowiak A, Wildes TM, Orlowski RZ, Shain KH, Cowan AJ, Pei H, Cortoos A, Patel S, Lin TS, Voorhees PM, Usmani SZ, and Richardson PG
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Bortezomib therapeutic use, Bortezomib administration & dosage, Dexamethasone administration & dosage, Dexamethasone therapeutic use, Lenalidomide therapeutic use, Lenalidomide administration & dosage, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Multiple Myeloma drug therapy, Multiple Myeloma mortality, Multiple Myeloma therapy, Multiple Myeloma diagnosis
- Abstract
The randomized, phase 2 GRIFFIN study (NCT02874742) evaluated daratumumab plus lenalidomide/bortezomib/dexamethasone (D-RVd) in transplant-eligible newly diagnosed multiple myeloma (NDMM). We present final post hoc analyses (median follow-up, 49.6 months) of clinically relevant subgroups, including patients with high-risk cytogenetic abnormalities (HRCAs) per revised definition (del[17p], t[4;14], t[14;16], t[14;20], and/or gain/amp[1q21]). Patients received 4 induction cycles (D-RVd/RVd), high-dose therapy/transplant, 2 consolidation cycles (D-RVd/RVd), and lenalidomide±daratumumab maintenance (≤ 2 years). Minimal residual disease-negativity (10
-5 ) rates were higher for D-RVd versus RVd in patients ≥ 65 years (67.9% vs 17.9%), with HRCAs (54.8% vs 32.4%), and with gain/amp(1q21) (61.8% vs 28.6%). D-RVd showed a trend toward improved progression-free survival versus RVd (hazard ratio [95% confidence interval]) in patients ≥ 65 years (0.29 [0.06-1.48]), with HRCAs (0.38 [0.14-1.01]), and with gain/amp(1q21) (0.42 [0.14-1.27]). In the functional high-risk subgroup (not MRD negative at the end of consolidation), the hazard ratio was 0.82 (0.35-1.89). Among patients ≥ 65 years, grade 3/4 treatment-emergent adverse event (TEAE) rates were higher for D-RVd versus RVd (88.9% vs 77.8%), as were TEAEs leading to discontinuation of ≥ 1 treatment component (37.0% vs 25.9%). One D-RVd patient died due to an unrelated TEAE. These results support the addition of daratumumab to RVd in transplant-eligible patients with high-risk NDMM. Video Abstract., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
11. Health-related quality of life in transplant-eligible patients with newly diagnosed multiple myeloma treated with daratumumab, lenalidomide, bortezomib, and dexamethasone: Patient-reported outcomes from GRIFFIN.
- Author
-
Silbermann R, Laubach J, Kaufman JL, Sborov DW, Reeves B, Rodriguez C, Chari A, Costa LJ, Anderson LD Jr, Nathwani N, Shah N, Bumma N, Holstein SA, Costello C, Jakubowiak A, Orlowski RZ, Shain KH, Cowan AJ, Gries KS, Pei H, Cortoos A, Patel S, Lin TS, Voorhees PM, Usmani SZ, and Richardson PG
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Multiple Myeloma drug therapy, Quality of Life, Bortezomib administration & dosage, Bortezomib therapeutic use, Dexamethasone administration & dosage, Dexamethasone therapeutic use, Lenalidomide administration & dosage, Lenalidomide therapeutic use, Patient Reported Outcome Measures, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal administration & dosage
- Abstract
In the phase 2 GRIFFIN trial (ClinicalTrials.gov identifier: NCT02874742), daratumumab added to lenalidomide, bortezomib, and dexamethasone (D-RVd) improved depth of response and progression-free survival (PFS) versus lenalidomide, bortezomib, and dexamethasone (RVd) alone in transplant-eligible (TE) patients with newly diagnosed multiple myeloma (NDMM). Here, we present patient-reported outcomes (PROs) collected using the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Core 30-item (QLQ-C30), EORTC Quality of Life Questionnaire Multiple Myeloma Module 20-item (QLQ-MY20), and EuroQol 5-Dimension 5-Level (EQ-5D-5L) tools on day 1 of cycles 1, 2, and 3; on day 21 of cycle 4 (end of induction therapy); on day 1 of cycle 5; on day 21 of cycle 6 (end of posttransplant consolidation therapy); and at months 6, 12, 18, and 24 of maintenance therapy. Meaningful improvements from baseline were seen in most of the PRO scales with both treatments after consolidation and were sustained for at least 2 years of maintenance treatment. Large reductions from baseline (~20 points) were especially observed in pain symptoms for both treatment groups, although these were numerically higher for patients receiving D-RVd during the majority of the time points. In addition, improvements in key scales, such as global health status, fatigue symptoms, and physical functioning, were also seen with both D-RVd and RVd. These improvements in health-related quality of life contribute to the totality of evidence supporting the improvement in clinical outcomes such as response rates and PFS with D-RVd in induction, consolidation, and maintenance therapy in TE patients with NDMM., (© 2024 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
12. Sequence not salvage.
- Author
-
Sborov DW, Fortuna GG, and Hayden PJ
- Subjects
- Humans, B-Cell Maturation Antigen, Receptors, Chimeric Antigen therapeutic use, Multiple Myeloma therapy, Immunotherapy, Adoptive methods, Salvage Therapy methods
- Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy for the treatment of multiple myeloma (MM) has fundamentally changed the relapsed and refractory therapeutic landscape, but the disease remains incurable. Two CAR-T products, idecabtagene vicleucel (ide-cel; Abecma) and ciltacabtagene autoleucel (cilta-cel, Carvykti), have been FDA- and EMA-approved for the treatment of relapsed/refractory MM (RRMM); both target B-cell maturation antigen (BCMA), a surface glycoprotein highly expressed on MM cells. Despite deep and durable responses following CAR-T therapy, most patients will need subsequent treatment, and the optimal next-line therapy is presently unclear. Commentary on: Liu et al. Outcomes in patients with multiple myeloma receiving salvage treatment after BCMA-specific CAR-T therapy: A retrospective analysis of LEGEND-2. Br J Haematol 2024;204:1780-1789., (© 2024 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
13. Factors associated with refractoriness or early progression after idecabtagene vicleucel in patients with relapsed/ refractory multiple myeloma: US Myeloma Immunotherapy Consortium real world experience.
- Author
-
Hashmi H, Hansen DK, Peres LC, Puglianini OC, Freeman C, De Avila G, Sidana S, Shune L, Sborov DW, Davis J, Wagner C, Kocoglu MH, Atrash S, Voorhees P, Simmons G, Ferreri C, Kalariya N, Anderson LD Jr, Afrough A, Dima D, Khouri J, McGuirk J, Locke F, Baz R, Patel KK, and Alsina M
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Biological Products therapeutic use, Biological Products administration & dosage, Adult, B-Cell Maturation Antigen immunology, Antigens, CD19 immunology, Antigens, CD19 therapeutic use, Recurrence, Treatment Outcome, Aged, 80 and over, Drug Resistance, Neoplasm, United States epidemiology, Recombinant Fusion Proteins administration & dosage, Recombinant Fusion Proteins therapeutic use, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen therapeutic use, Risk Factors, Multiple Myeloma therapy, Multiple Myeloma immunology, Multiple Myeloma mortality, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects, Disease Progression
- Abstract
While response rates and survival outcomes have been very promising for idecabtagene vicleucel (ide-cel), a proportion of patients do not respond or relapse early after this B-cell maturation antigen (BCMA) targeted chimeric antigen receptor (CAR) T-cell therapy. Understanding the characteristics of these patients is important for patient selection and development of novel strategies to improve outcomes. We evaluated factors associated with early progression (progression or death due to myeloma ≤3 months after CAR T-cell infusion) in patients treated with standard of care ide-cel at 11 US academic centers. Among 211 patients that received ide-cel, 43 patients had a progressive event ≤3 months of infusion. Patients with a history of extramedullary disease, prior BCMA targeted therapy, elevated ferritin at lymphodepletion, use of bridging therapy, Hispanic ethnicity, plasma cell leukemia and t(4;14) were more likely to progress ≤3 months of infusion (P<0.05). Of these risk factors for early progression identified in univariate analyses, history of extramedullary disease, prior BCMA targeted therapy, elevated ferritin at lymphodepletion, plasma cell leukemia, and t(4;14) were associated with worse progression-free survival (PFS) in multivariable analysis. Presence of three or more of these factors had a significant negative impact on PFS (P<0.001; median PFS for ≥3 factors, 3.2 months vs. 0 factors, 14.1 months). This study helps identify patients at high risk of early progression after CAR T-cell therapy who may benefit from specific interventions pre and post CAR T-cell therpy to improve outcomes.
- Published
- 2024
- Full Text
- View/download PDF
14. Patient perspectives on BCMA-targeted therapies for multiple myeloma: a survey conducted in a patient advocacy group.
- Author
-
Hydren JR, Lin D, Sweeney NW, Wu B, Kim N, Patel S, Sborov DW, Berdeja JG, Anderson LD Jr, Huo S, Hurtado Martínez JA, and Ahlstrom JM
- Abstract
Background: Advances in multiple myeloma (MM) treatment have shifted the therapeutic landscape. Understanding patients' perspectives can assist physicians in helping patients make informed decisions. This study aimed to understand the patient decision-making process and gain insights into patient perspectives on B-cell maturation antigen (BCMA)-targeted therapies for MM., Methods: An 18-question survey was completed by patients with MM enrolled in HealthTree® Cure Hub, an online portal helping patients with plasma cell dyscrasias navigate their disease., Results: From October 28, 2022, to January 12, 2023, 325 patients with MM participated in the survey. The mean age (standard deviation) of the respondents was 66 (8) years; 54% were female and 90% were White. Among 218 patients with complete clinical records in the database, the median (min, max) lines of therapy (LOT) was 2 (1,16). Among 61 (28%) patients who had received ≥4 LOTs, 55 (90%) were triple-class exposed. Of the 290 patients who responded to the question about openness to new therapies, 76 (26%) were open to trying a new therapy immediately and 125 (43%) wanted more information on safety and efficacy. Most respondents reported likely or very likely to try a BCMA CAR T-cell therapy (60%) or a bispecific antibody (74%) and some needed more information to decide (16% for CAR T-cell therapy and 13% for bispecific antibody). The most requested information included efficacy, side effects (SEs), eligibility, and administration process for both CAR T-cell and bispecific therapies. When 2 therapies with the same efficacy and duration of response were offered, 69% of respondents would prefer the therapy with a lower risk of severe SEs but requires continuous dosing with no treatment-free interval, and 31% preferred a therapy given once followed by a treatment-free interval but with a potentially higher risk of severe SEs. To receive an effective therapy, the top acceptable trade-offs included frequent monitoring of SEs and initiating a new therapy in a hospital setting, and the least acceptable compromise was caregiver burden., Conclusions: This study found a high level of openness in patients with MM to try BCMA-targeted therapies. Information on efficacy, safety, availability, and eligibility may assist patients on decision-making., Competing Interests: JH received research grant support from Janssen to perform the study and receives research funding from Adaptive Biotechnologies, Sanofi, GlaxoSmithKline, Regeneron, Pfizer, Janssen, and Takeda. DL, BW, NK, SP, SH are all employees of Janssen Scientific Affairs, LLC and may hold stocks of Johnson & Johnson. DS was consultant/advisory for GlaxoSmithKline, Janssen, Sanofi, Abbvie, Bristol Myer Squibb, Pfizer, Bioline, Arcellx, AstraZeneca; research for Gilead, Pfizer, Janssen, Bioline, GlaxoSmithKline, Sanofi, Amgen, Cantex, Arcellx, Roche; steering committee for Janssen (AURIGA); data safety and monitoring for Karyopharm; and independent review committee for Parexel. JB’s institute, Sarah Cannon Research Institute and Tennessee Oncology, receives research funds in his name from Abbvie, Acetylon, Amgen, Astex Pharmaceuticals, Bluebird bio, BMS, C4 Therapeutics, Caribou Biosciences, CARsgen, Cartesian Therapeutics, Celgene, Cellectis, Celularity, CRISPR Therapeutics, Curis, EMD Sorono, Fate Therapeutics, Genentech, GSK, Ichnos Sciences, Incyte, Janssen, Juno Therapeutics, K36 Therapeutics. Karyopharm, Lilly, Novartis, Poseida, Roche, Sanofi, Takeda; as well as, is a consultant for AstraZeneca, Bluebird bio, BMS, Caribou Biosciences, Janssen, K36 Therapeutics, Kite Pharma, Legend Biotech, Pfizer, Roche, Sanofi-Aventis, Takeda. LA has had consulting/advisory board activity with Janssen, Celgene, BMS, Amgen, GSK, AbbVie, Beigene, Cellectar, Sanofi, and Prothena. The authors declare that this study received funding from Janssen Scientific Affairs, LLC. The funder had collaborative involvement in the following steps and earned authorship: study design, interpretation of data, the writing of this article, and the decision to submit it for publication; and not involved in the following steps: data collection and analysis. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© 2024 Hydren, Lin, Sweeney, Wu, Kim, Patel, Sborov, Berdeja, Anderson, Huo, Hurtado Martínez and Ahlstrom.)
- Published
- 2024
- Full Text
- View/download PDF
15. Daratumumab-based quadruplet therapy for transplant-eligible newly diagnosed multiple myeloma with high cytogenetic risk.
- Author
-
Callander NS, Silbermann R, Kaufman JL, Godby KN, Laubach J, Schmidt TM, Sborov DW, Medvedova E, Reeves B, Dhakal B, Rodriguez C, Chhabra S, Chari A, Bal S, Anderson LD Jr, Dholaria BR, Nathwani N, Hari P, Shah N, Bumma N, Holstein SA, Costello C, Jakubowiak A, Wildes TM, Orlowski RZ, Shain KH, Cowan AJ, Pei H, Cortoos A, Patel S, Lin TS, Giri S, Costa LJ, Usmani SZ, Richardson PG, and Voorhees PM
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Chromosome Aberrations, Adult, Lenalidomide therapeutic use, Lenalidomide administration & dosage, Lenalidomide adverse effects, Dexamethasone administration & dosage, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma therapy, Multiple Myeloma mortality, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
In the MASTER study (NCT03224507), daratumumab+carfilzomib/lenalidomide/dexamethasone (D-KRd) demonstrated promising efficacy in transplant-eligible newly diagnosed multiple myeloma (NDMM). In GRIFFIN (NCT02874742), daratumumab+lenalidomide/bortezomib/dexamethasone (D-RVd) improved outcomes for transplant-eligible NDMM. Here, we present a post hoc analysis of patients with high-risk cytogenetic abnormalities (HRCAs; del[17p], t[4;14], t[14;16], t[14;20], or gain/amp[1q21]). Among 123 D-KRd patients, 43.1%, 37.4%, and 19.5% had 0, 1, or ≥2 HRCAs. Among 120 D-RVd patients, 55.8%, 28.3%, and 10.8% had 0, 1, or ≥2 HRCAs. Rates of complete response or better (best on study) for 0, 1, or ≥2 HRCAs were 90.6%, 89.1%, and 70.8% for D-KRd, and 90.9%, 78.8%, and 61.5% for D-RVd. At median follow-up (MASTER, 31.1 months; GRIFFIN, 49.6 months for randomized patients/59.5 months for safety run-in patients), MRD-negativity rates as assessed by next-generation sequencing (10
-5 ) were 80.0%, 86.4%, and 83.3% for 0, 1, or ≥2 HRCAs for D-KRd, and 76.1%, 55.9%, and 61.5% for D-RVd. PFS was similar between studies and superior for 0 or 1 versus ≥2 HRCAs: 36-month PFS rates for D-KRd were 89.9%, 86.2%, and 52.4%, and 96.7%, 90.5%, and 53.5% for D-RVd. These data support the use of daratumumab-containing regimens for transplant-eligible NDMM with HCRAs; however, additional strategies are needed for ultra-high-risk disease (≥2 HRCAs). Video Abstract., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
16. Real-world impact of bridging therapy on outcomes of ide-cel for myeloma in the U.S. Myeloma Immunotherapy Consortium.
- Author
-
Afrough A, Hashmi H, Hansen DK, Sidana S, Ahn C, Peres LC, Dima D, Freeman CL, Puglianini OC, Kocoglu MH, Atrash S, Voorhees PM, Shune L, McGuirk JP, Simmons G, Sborov DW, Davis JA, Kaur G, Sannareddy A, Ferreri CJ, Gaballa MR, Goldsmith S, Nadeem O, Midha S, Wagner CB, Locke FL, Patel KK, Khouri J, Anderson LD Jr, and Lin Y
- Subjects
- Humans, Multiple Myeloma therapy, Receptors, Chimeric Antigen
- Published
- 2024
- Full Text
- View/download PDF
17. Idecabtagene vicleucel chimeric antigen receptor T-cell therapy for relapsed/refractory multiple myeloma with renal impairment.
- Author
-
Sidana S, Peres LC, Hashmi H, Hosoya H, Ferreri C, Khouri J, Dima D, Atrash S, Voorhees P, Simmons G, Sborov DW, Kalariya N, Hovanky V, Bharadwaj S, Miklos D, Wagner C, Kocoglu MH, Kaur G, Davis JA, Midha S, Janakiram M, Freeman C, Alsina M, Locke F, Gonzalez R, Lin Y, McGuirk J, Afrough A, Shune L, Patel KK, and Hansen DK
- Subjects
- Humans, Female, Male, Immunotherapy, Adoptive adverse effects, Cell- and Tissue-Based Therapy, Receptors, Chimeric Antigen, Multiple Myeloma complications, Multiple Myeloma therapy, Neoplasms, Plasma Cell, Cytopenia, Renal Insufficiency
- Abstract
We evaluated patients with relapsed multiple myeloma with renal impairment (RI) treated with standard of care idecabtagene vicleucel (ide-cel), as outcomes with chimeric antigen receptor (CAR) T-cell therapy are unknown in this population. RI was defined as creatinine clearance (CrCl) <50 mL/min. CrCl of <30 mL/min or dialysis dependence were defined as severe RI. The study cohort included 214 patients, 28 (13%) patients with RI, including 11 patients severe RI (dialysis, N=1). Patients with RI were older, more likely to be female and had higher likelihood of having Revised International Staging System stage 3 disease. Rates and severity of cytokine release syndrome (89% vs. 84%, grade ≥3: 7% vs. 2%) and immune effector cell-associated neurotoxicity syndrome (23% vs. 20%) were similar in patients with and without RI, respectively. Patients with RI had higher incidence of short-term grade ≥3 cytopenias, although cytopenias were similar by 3 months following CAR T-cell therapy. Renal function did not worsen after CAR T-cell therapy in patients with RI. Response rates (93% vs. 82%) and survival outcomes (median progression-free survival: 9 vs. 8 months; P=0.26) were comparable in patients with and without RI, respectively. Treatment with ide-cel is feasible in patients with RI, with a comparable safety and efficacy profile as patients without RI, with notable exception of higher short-term high-grade cytopenias.
- Published
- 2024
- Full Text
- View/download PDF
18. Racial and ethnic differences in clinical outcomes among patients with multiple myeloma treated with CAR T-cell therapy.
- Author
-
Peres LC, Oswald LB, Dillard CM, De Avila G, Nishihori T, Blue BJ, Freeman CL, Locke FL, Alsina M, Castaneda Puglianini O, Shune L, Sborov DW, Wagner C, Dima D, Hashmi H, Davis JA, Kocoglu MH, Badros AZ, Atrash S, Simmons G, Kalariya N, Ferreri C, Anderson LD Jr, Afrough A, Kaur G, Lin Y, Liu L, Nadeem O, Voorhees P, Khouri J, McGuirk J, Sidana S, Hansen DK, and Patel K
- Subjects
- United States, Humans, Immunotherapy, Adoptive adverse effects, Ethnicity, Minority Groups, Multiple Myeloma therapy, Neoplasms, Plasma Cell
- Abstract
Abstract: Idecabtagene vicleucel (ide-cel) was the first chimeric antigen receptor T-cell therapy to gain US Food and Drug Administration approval for patients with relapsed/refractory multiple myeloma (RRMM). The clinical outcomes of standard of care (SOC) ide-cel in racially and ethnically diverse populations have been understudied. This study pooled data from 207 patients with RRMM (28% patients of racial and ethnic minority groups) treated with SOC ide-cel across 11 institutions to examine racial and ethnic differences in the incidence of toxicities and adverse events, response to ide-cel, and survival. This study included 22 (11%) Hispanic, 36 (17%) non-Hispanic Black, and 149 (72%) non-Hispanic White patients with RRMM. Compared with Hispanic and non-Hispanic White patients, non-Hispanic Black patients had higher median levels of C-reactive protein (1.0, 0.8, and 3.5 mg/dL, respectively; P = .02) and baseline ferritin (362.0 vs 307.0 vs 680.5, respectively; P = .08) and were more likely to develop cytokine release syndrome (77%, 85%, and 97%, respectively; P = .04). Although best overall response rate was lower among Hispanic patients (59%) than among non-Hispanic Black (86%) and White patients (86%; P = .01), there were no racial and ethnic differences in progression-free or overall survival. We provide, to our knowledge, the first and largest investigation of clinical outcomes of SOC ide-cel by race and ethnicity. Despite differences in safety and response to ide-cel, our findings encourage the use of ide-cel in all patients with RRMM. These findings should be confirmed in larger samples of diverse patients with RRMM, with longer follow-up time., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
19. Understanding risks and refining strategies for thromboembolism prophylaxis in hematopoietic stem cell transplant recipients.
- Author
-
Baljevic M and Sborov DW
- Subjects
- Humans, Antifungal Agents therapeutic use, Transplant Recipients, Mycoses drug therapy, Hematopoietic Stem Cell Transplantation, Thromboembolism drug therapy
- Published
- 2023
- Full Text
- View/download PDF
20. Addition of daratumumab to lenalidomide, bortezomib, and dexamethasone for transplantation-eligible patients with newly diagnosed multiple myeloma (GRIFFIN): final analysis of an open-label, randomised, phase 2 trial.
- Author
-
Voorhees PM, Sborov DW, Laubach J, Kaufman JL, Reeves B, Rodriguez C, Chari A, Silbermann R, Costa LJ, Anderson LD Jr, Nathwani N, Shah N, Bumma N, Efebera YA, Holstein SA, Costello C, Jakubowiak A, Wildes TM, Orlowski RZ, Shain KH, Cowan AJ, Dinner S, Pei H, Cortoos A, Patel S, Lin TS, Usmani SZ, and Richardson PG
- Subjects
- Humans, Male, Bortezomib adverse effects, Lenalidomide therapeutic use, Thalidomide adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone, Multiple Myeloma therapy, Thrombocytopenia etiology
- Abstract
Background: Addition of daratumumab to lenalidomide, bortezomib, and dexamethasone (D-RVd) in the GRIFFIN study improved the stringent complete response rate by the end of consolidation in transplantation-eligible patients with newly diagnosed multiple myeloma. Here, we report the findings of the predefined final analysis., Methods: GRIFFIN was an open-label, randomised, active-controlled, phase 2 trial done in 35 research centres in the USA. Patients had newly diagnosed multiple myeloma with measurable disease by M protein or free light chain, were aged 18-70 years, had an ECOG performance score of 0-2, and were eligible for autologous haematopoietic stem-cell transplantation (HSCT). Patients were randomly assigned (1:1) to four D-RVd or RVd induction cycles, autologous HSCT, two D-RVd or RVd consolidation cycles, and lenalidomide with or without daratumumab maintenance therapy for 2 years. Patients received 21-day cycles of oral lenalidomide (25 mg on days 1-14), subcutaneous bortezomib (1·3 mg/m
2 on days 1, 4, 8, and 11), oral dexamethasone (40 mg weekly) with or without intravenous daratumumab (16 mg/kg weekly, cycles 1-4; day 1, cycles 5-6). Maintenance therapy (28-day cycles) was oral lenalidomide (10 mg on days 1-21) with or without daratumumab (16 mg/kg intravenously every 4 or 8 weeks, or 1800 mg subcutaneously monthly). Patients could continue lenalidomide maintenance after study treatment completion. The primary endpoint was stringent complete response rate by the end of consolidation in the response-evaluable population, and has already been reported. Here we report updated stringent complete response rates and secondary outcomes including progression-free survival and overall survival. The trial is registered with ClinicalTrials.gov (NCT02874742) and ended on April 8, 2022., Findings: Between Dec 20, 2016, and April 10, 2018, 104 patients were randomly assigned to the D-RVd group and 103 were randomly assigned to the RVd group; most patients were White (85 [82%] in the D-RVd group and 76 [74%] in the RVd group) and male (58 [56%] in the D-RVd group and 60 [58%] in the RVd group). At a median follow-up of 49·6 months (IQR 47·4-52·1), D-RVd improved rates of stringent complete response (67 [67%] of 100] vs 47 [48%] of 98]; odds ratio 2·18 [95% CI 1·22-3·89], p=0·0079), and 4-year progression-free survival was 87·2% (95% CI 77·9-92·8) for D-RVd versus 70·0% (95% CI 55·9-80·3) for RVd, with a hazard ratio (HR) of 0·45 (95% CI 0·21-0·95, p=0·032) for risk of disease progression or death with D-RVd. Median overall survival was not reached for either group (HR 0·90 [95% CI 0·31-2·56], p=0·84). The most common grade 3-4 treatment-emergent adverse events in the D-RVd versus RVd groups were neutropenia (46 [46%] of 99 vs 23 [23%] of 102), lymphopenia (23 [23%] vs 23 [23%]), leukopenia (17 [17%] vs eight [8%]), thrombocytopenia (16 [16%] vs nine [9%]), pneumonia (12 [12%] vs 14 [14%]), and hypophosphataemia (ten [10%] vs 11 [11%]). Serious treatment-emergent adverse events occurred in 46 (46%) of 99 patients in the D-RVd group and in 53 (52%) of 102 patients in the RVd group. One patient in each treatment group reported a treatment-emergent adverse event that resulted in death (bronchopneumonia in the D-RVd group; cause unknown in the RVd group); neither was related to study treatment. No new safety concerns occurred with maintenance therapy., Interpretation: Addition of daratumumab to RVd improved the depth of response and progression-free survival in transplantation-eligible patients with newly diagnosed multiple myeloma. These results justify further evaluation in phase 3 studies., Funding: Janssen Oncology., Competing Interests: Declaration of interests PMV served as a consultant for, served on an advisory board for, and received honoraria from AbbVie, Amgen, Bristol Myers Squibb, GlaxoSmithKline, Karyopharm Therapeutics, Novartis, Oncopeptides, Pfizer, Sanofi, and Secura Bio. DWS served as a consultant or in an advisory role for GlaxoSmithKline, AbbVie, Sanofi, Bristol Myers Squibb, Janssen, and Pfizer. JLK served as a consultant for AbbVie, Bristol Myers Squibb, Janssen, Roche/Genentech, and Tecnopharma; received research funding from AbbVie, Amgen, Bristol Myers Squibb, Fortis Therapeutics, Heidelberg Pharma, Janssen, Novartis, Roche/Genentech, Sutro Biopharma, and Takeda; received honoraria from AbbVie, Janssen, Roche/Genentech, and Tecnopharma; and served on an advisory board for Incyte and TG Therapeutics. BR received honoraria from Incyte, Bristol Myers Squibb, and PharmaEssentia. CR served as a consultant and on a speakers bureau for Janssen, Takeda, Bristol Myers Squibb, Amgen, and Karyopharm Therapeutics. ACh served as a consultant or in an advisory role for Amgen, Janssen Oncology, Seattle Genetics, Karyopharm Therapeutics, Genzyme, Oncopeptides, Takeda, Antengene, GlaxoSmithKline, Secura Bio, Shattuck Labs, Genentech, AbbVie, and Bristol Myers Squibb/Celgene; and received research funding from Celgene, Janssen, Amgen, Seattle Genetics, Takeda, and Pharmacyclics. RS served as a consultant or in an advisory role for Sanofi/Aventis, Janssen Oncology, and Oncopeptides; and received research funding from Sanofi. LJC served as a consultant or in an advisory role for AbbVie, Amgen, Celgene, Karyopharm Therapeutics, and Sanofi; served on a speakers bureau for Amgen and Sanofi; received honoraria from Amgen, Celgene, Janssen, Karyopharm Therapeutics, and Sanofi; and received research funding from Amgen and Janssen. LDA served as a consultant or in an advisory role for and received honoraria from GlaxoSmithKline, Bristol Myers Squibb, Celgene, Janssen, Amgen, Oncopeptides, Karyopharm Therapeutics, AbbVie, BeiGene, Cellectar, and Sanofi. NS served as a consultant for Amgen, CareDx, CSL Behring, GlaxoSmithKline, Indapta Therapeutics, Karyopharm Therapeutics, Kite, Oncopeptides, and Sanofi; and received research funding from bluebird bio, Bristol Myers Squibb/Celgene, Janssen, Nektar, Poseida, Precision BioSciences, Sutro Biopharma, and TeneoBio. NB received honoraria from OncLive; and served as a consultant or in an advisory role and on a speakers bureau for Janssen, Sanofi Genzyme, and Oncopeptides. YAE received research funding, received honoraria, and served on a speakers bureau for Janssen, Takeda, Oncopeptides, GlaxoSmithKline, Pfizer, Sanofi, and Bristol Myers Squibb. SAH served as a consultant for Bristol Myers Squibb/Celgene, Janssen, Takeda, Oncopeptides, GlaxoSmithKline, Secura Bio, and Sanofi; and received research funding from Oncopeptides. CC received honoraria from Takeda, Bristol Myers Squibb, Pfizer, and Janssen. AJ served as a consultant or in an advisory role for and received honoraria from AbbVie, Amgen, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Karyopharm Therapeutics, and Sanofi. TMW served as a consultant for Janssen, Carevive, and Sanofi. RZO holds stock and other ownership interests in Asylia Therapeutics; received honoraria from and served as a consultant or in an advisory role for AbbVie, Biotheryx, Bristol Myers Squibb, GlaxoSmithKline, Karyopharm Therapeutics, Meridian Therapeutics, Monte Rosa Therapeutics, Neoleukin Corporation, Oncopeptides, Regeneron, Sanofi, and Takeda; received research funding from Asylia Therapeutics, Biotheryx, and Heidelberg Pharma; and holds patents, royalties, or other intellectual property for Asylia Therapeutics. KHS served on an advisory board for AbbVie, Amgen, Bristol Myers Squibb, GlaxoSmithKline, and Janssen; received research funding from AbbVie and Karyopharm Therapeutics; served on a speakers bureau for Adaptive Biotechnologies Corporation, Amgen, Bristol Myers Squibb, Janssen, and Sanofi Genzyme; served as a consultant for Adaptive Biotechnologies Corporation, Novartis, and Sanofi Genzyme; and received honoraria from Karyopharm Therapeutics. AJC served as a consultant for Janssen, Bristol Myers Squibb, EUSA Pharma, AbbVie, Sanofi, Cellectar, GlaxoSmithKline, and Secura Bio; and received research funding from Janssen, AbbVie, Sanofi, Harpoon, Bristol Myers Squibb, Adaptive Biotechnologies, and Nektar. SD is the Executive Officer for Alliance Foundation Trials for Clinical Trials. HP, ACo, and SP are employees of and hold stock and other ownership in Janssen. TSL is an employee of Janssen. SZU served as a consultant or in an advisory role for Celgene, Amgen, Janssen Oncology, Seattle Genetics, Takeda, GlaxoSmithKline, Karyopharm Therapeutics, AbbVie, SkylineDx, Merck, Oncopeptides, Genentech, Gilead Sciences, and Bristol Myers Squibb/Celgene; served on a speakers bureau for Takeda, Amgen, Janssen Oncology, Sanofi, and Bristol Myers Squibb/Celgene; and received research funding from Celgene and Array BioPharma. PGR received research funding from Oncopeptides, Celgene/Bristol Myers Squibb, Takeda, and Karyopharm Therapeutics; and served on an advisory committee for Oncopeptides, Celgene/Bristol Myers Squibb, Takeda, Karyopharm Therapeutics, Janssen, Sanofi, Secura Bio, GlaxoSmithKline, Regeneron, AstraZeneca, and Protocol Intelligence. JL and NN declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
21. Correction: Distinct germline genetic susceptibility profiles identified for common non-Hodgkin lymphoma subtypes.
- Author
-
Berndt SI, Vijai J, Benavente Y, Camp NJ, Nieters A, Wang Z, Smedby KE, Kleinstern G, Hjalgrim H, Besson C, Skibola CF, Morton LM, Brooks-Wilson AR, Teras LR, Breeze C, Arias J, Adami HO, Albanes D, Anderson KC, Ansell SM, Bassig B, Becker N, Bhatti P, Birmann BM, Boffetta P, Bracci PM, Brennan P, Brown EE, Burdett L, Cannon-Albright LA, Chang ET, Chiu BCH, Chung CC, Clavel J, Cocco P, Colditz G, Conde L, Conti DV, Cox DG, Curtin K, Casabonne D, De Vivo I, Diepstra A, Diver WR, Dogan A, Edlund CK, Foretova L, Fraumeni JF Jr, Gabbas A, Ghesquières H, Giles GG, Glaser S, Glenn M, Glimelius B, Gu J, Habermann TM, Haiman CA, Haioun C, Hofmann JN, Holford TR, Holly EA, Hutchinson A, Izhar A, Jackson RD, Jarrett RF, Kaaks R, Kane E, Kolonel LN, Kong Y, Kraft P, Kricker A, Lake A, Lan Q, Lawrence C, Li D, Liebow M, Link BK, Magnani C, Maynadie M, McKay J, Melbye M, Miligi L, Milne RL, Molina TJ, Monnereau A, Montalvan R, North KE, Novak AJ, Onel K, Purdue MP, Rand KA, Riboli E, Riby J, Roman E, Salles G, Sborov DW, Severson RK, Shanafelt TD, Smith MT, Smith A, Song KW, Song L, Southey MC, Spinelli JJ, Staines A, Stephens D, Sutherland HJ, Tkachuk K, Thompson CA, Tilly H, Tinker LF, Travis RC, Turner J, Vachon CM, Vajdic CM, Van Den Berg A, Van Den Berg DJ, Vermeulen RCH, Vineis P, Wang SS, Weiderpass E, Weiner GJ, Weinstein S, Doo NW, Ye Y, Yeager M, Yu K, Zeleniuch-Jacquotte A, Zhang Y, Zheng T, Ziv E, Sampson J, Chatterjee N, Offit K, Cozen W, Wu X, Cerhan JR, Chanock SJ, Slager SL, and Rothman N
- Published
- 2023
- Full Text
- View/download PDF
22. Correction: HDAC inhibitor AR-42 decreases CD44 expression and sensitizes myeloma cells to lenalidomide.
- Author
-
Canella A, Nieves HC, Sborov DW, Cascione L, Radomska HS, Smith E, Stiff A, Consiglio J, Caserta E, Rizzotto L, Zanesi N, Stefano V, Kaur B, Mo X, Byrd JC, Efebera YA, Hofmeister CC, and Pichiorri F
- Published
- 2023
- Full Text
- View/download PDF
23. Efficacy and Safety of CD34+ Stem Cell Boost for Delayed Hematopoietic Recovery After BCMA Directed CAR T-cell Therapy.
- Author
-
Davis JA, Sborov DW, Wesson W, Julian K, Abdallah AO, McGuirk JP, Ahmed N, and Hashmi H
- Subjects
- Adult, Humans, Immunotherapy, Adoptive adverse effects, B-Cell Maturation Antigen therapeutic use, Retrospective Studies, Hematopoietic Stem Cells, Antigens, CD34 therapeutic use, Receptors, Chimeric Antigen therapeutic use, Multiple Myeloma drug therapy, Hematopoietic Stem Cell Transplantation, Thrombocytopenia
- Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized treatment outcomes for patients with relapsed refractory multiple myeloma (RRMM). Despite supportive care with growth factors and thrombopoietin (TPO) mimetics, nearly half of the patients experience severe and prolonged cytopenias after CAR T infusion, which have become a major therapeutic challenge for patients with RRMM. Given the successful use of CD34+ autologous hematopoietic stem cells for treatment of non- or delayed engraftment after allogeneic and autologous stem cell transplantations, there is a need to explore the role of previously stored autologous stem cells as a boost for post-CAR T cytopenias in RRMM. We performed a multicenter retrospective analysis of adult patients with RRMM who received previously collected and stored CD34+ stem cell boosts after CAR T-cell therapy between 2 July 2020 and 18 January 2023. Indications for boost were determined per physician discretion and primarily included cytopenias and related complications. Overall, a total of 19 patients received stem cell boost, at a median dose of 2.75 × 10
6 CD34+ cells/kg (range 1.76-7.38), given at a median of 53 days (range 24-126) after CAR T infusion. Eighteen (95%) patients successfully recovered hematopoiesis after stem cell boost with median time for neutrophil, platelet, and hemoglobin engraftment of 14 (range 9-39), 17 (range 12-39), and 23 (range 6-34) days after the boost, respectively. Stem cell boosts were well tolerated, with no patients experiencing infusion reactions. Although infections were common and severe before stem cell boost, only 1 patient experienced a new infection after boost. All patients had experienced independence from use of growth factors, TPO agonists, and transfusions at the last follow-up. Autologous stem cell boosts can be effectively and safely used to promote hematopoietic recovery for post-CAR T cytopenias in patients with RRMM. Stem cell boosts can be a very effective rescue measure for post-CAR T cytopenias and related complications, as well as supportive care needs., (Copyright © 2023 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
24. Allogeneic stem cell transplant for multiple myeloma & myelofibrosis with split-dose busulfan, fludarabine & cyclophosphamide.
- Author
-
Trunk AD, Patel SS, Prchal JT, Sborov DW, Zander AR, and Lee CJ
- Abstract
Allogeneic stem cell transplant can have high morbidity and mortality in patients with myelofibrosis (MF) and multiple myeloma (MM). This phase 2 study used a novel myeloablative regimen of split-dose busulfan, fludarabine, and then post-transplant cyclophosphamide. Four patients with MF and 2 with MM were enrolled. At 1 year, non-relapse mortality was 33.3%, and overall survival was 50%. Incidence of acute and chronic GVHD was 33.3% and 16.7%, respectively. Those surviving beyond 1 year (MF = 1, MM = 2) had durable remissions with a median follow-up of 42 months. This small study demonstrates relative safety & favorable key outcomes using this novel approach., Competing Interests: None., (© 2023 Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
25. Real-world experience of patients with multiple myeloma receiving ide-cel after a prior BCMA-targeted therapy.
- Author
-
Ferreri CJ, Hildebrandt MAT, Hashmi H, Shune LO, McGuirk JP, Sborov DW, Wagner CB, Kocoglu MH, Rapoport A, Atrash S, Voorhees PM, Khouri J, Dima D, Afrough A, Kaur G, Anderson LD Jr, Simmons G, Davis JA, Kalariya N, Peres LC, Lin Y, Janakiram M, Nadeem O, Alsina M, Locke FL, Sidana S, Hansen DK, Patel KK, and Castaneda Puglianini OA
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Aged, Treatment Outcome, Multiple Myeloma therapy, B-Cell Maturation Antigen antagonists & inhibitors, Immunotherapy, Adoptive methods, Receptors, Chimeric Antigen therapeutic use
- Abstract
Most patients with multiple myeloma experience disease relapse after treatment with a B-cell maturation antigen-targeted therapy (BCMA-TT), and data describing outcomes for patients treated with sequential BCMA-TT are limited. We analyzed clinical outcomes for patients infused with standard-of-care idecabtagene vicleucel, an anti-BCMA chimeric antigen receptor (CAR) T-cell therapy, at 11 US medical centers. A total of 50 patients with prior BCMA-TT exposure (38 antibody-drug conjugate, 7 bispecific, 5 CAR T) and 153 patients with no prior BCMA-TT were infused with ide-cel, with a median follow-up duration of 4.5 and 6.0 months, respectively. Safety outcomes between cohorts were comparable. The prior BCMA-TT cohort had a lower overall response rate (74% versus 88%; p = 0.021), median duration of response (7.4 versus 9.6 months; p = 0.03), and median progression-free survival (3.2 months versus 9.0 months; p = 0.0002) compared to the cohort without prior BCMA-TT. All five patients who received a prior anti-BCMA CAR T responded to ide-cel, and survival outcomes were best for this subgroup. In conclusion, treatment with ide-cel yielded meaningful clinical responses in real-world patients exposed to a prior BCMA-TT, though response rates and durability were suboptimal compared to those not treated with a prior BCMA-TT., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
26. Refractoriness to last line of therapy: how often is it mandated and reported? A systematic review of randomized myeloma trials.
- Author
-
Bindal P, Najjar M, Koehn K, Godara A, Sborov DW, McClune B, Julian K, Wagner C, and Mohyuddin GR
- Subjects
- Humans, Randomized Controlled Trials as Topic, Multiple Myeloma diagnosis, Multiple Myeloma therapy
- Published
- 2023
- Full Text
- View/download PDF
27. Deep Transcriptome Profiling of Multiple Myeloma Using Quantitative Phenotypes.
- Author
-
Griffin R, Hanson HA, Avery BJ, Madsen MJ, Sborov DW, and Camp NJ
- Subjects
- Humans, Gene Expression Profiling, Transcriptome, Phenotype, Progression-Free Survival, Multiple Myeloma genetics, Multiple Myeloma pathology
- Abstract
Background: Transcriptome studies are gaining momentum in genomic epidemiology, and the need to incorporate these data in multivariable models alongside other risk factors brings demands for new approaches., Methods: Here we describe SPECTRA, an approach to derive quantitative variables that capture the intrinsic variation in gene expression of a tissue type. We applied the SPECTRA approach to bulk RNA sequencing from malignant cells (CD138+) in patients from the Multiple Myeloma Research Foundation CoMMpass study., Results: A set of 39 spectra variables were derived to represent multiple myeloma cells. We used these variables in predictive modeling to determine spectra-based risk scores for overall survival, progression-free survival, and time to treatment failure. Risk scores added predictive value beyond known clinical and expression risk factors and replicated in an external dataset. Spectrum variable S5, a significant predictor for all three outcomes, showed pre-ranked gene set enrichment for the unfolded protein response, a mechanism targeted by proteasome inhibitors which are a common first line agent in multiple myeloma treatment. We further used the 39 spectra variables in descriptive modeling, with significant associations found with tumor cytogenetics, race, gender, and age at diagnosis; factors known to influence multiple myeloma incidence or progression., Conclusions: Quantitative variables from the SPECTRA approach can predict clinical outcomes in multiple myeloma and provide a new avenue for insight into tumor differences by demographic groups., Impact: The SPECTRA approach provides a set of quantitative phenotypes that deeply profile a tissue and allows for more comprehensive modeling of gene expression with other risk factors., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
28. Idecabtagene Vicleucel for Relapsed/Refractory Multiple Myeloma: Real-World Experience From the Myeloma CAR T Consortium.
- Author
-
Hansen DK, Sidana S, Peres LC, Colin Leitzinger C, Shune L, Shrewsbury A, Gonzalez R, Sborov DW, Wagner C, Dima D, Hashmi H, Kocoglu MH, Atrash S, Simmons G, Kalariya N, Ferreri C, Afrough A, Kansagra A, Voorhees P, Baz R, Khouri J, Alsina M, McGuirk J, Locke FL, and Patel KK
- Subjects
- Humans, B-Cell Maturation Antigen, Retrospective Studies, Immunotherapy, Adoptive, Cytokine Release Syndrome, Multiple Myeloma, Receptors, Chimeric Antigen
- Abstract
Purpose: Idecabtagene vicleucel (ide-cel) is an autologous B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy approved for relapsed/refractory multiple myeloma (RRMM) on the basis of the phase II pivotal KarMMa trial, which demonstrated best overall and ≥ complete response rates of 73% and 33%, respectively. We report clinical outcomes with standard-of-care (SOC) ide-cel under the commercial Food and Drug Administration label., Methods: Data were retrospectively collected from patients with RRMM who underwent leukapheresis as of February 28, 2022, at 11 US institutions with intent to receive SOC ide-cel. Toxicities were graded per American Society for Transplantation and Cellular Therapy guidelines and managed according to each institution's policies. Responses were graded on the basis of the International Myeloma Working Group response criteria., Results: One hundred fifty-nine of 196 leukapheresed patients received ide-cel by data cutoff. One hundred twenty (75%) infused patients would have been ineligible for participation in the KarMMa clinical trial because of comorbidities at the time of leukapheresis. Any grade and grade ≥ 3 cytokine release syndrome and neurotoxicity occurred in 82/3% and 18/6%, respectively. Best overall and ≥ complete response rates were 84% and 42%, respectively. At a median follow-up of 6.1 months from chimeric antigen receptor T infusion, the median progression-free survival was 8.5 months (95% CI, 6.5 to not reached) and the median overall survival was 12.5 months (95% CI, 11.3 to not reached). Patients with previous exposure to B-cell maturation antigen-targeted therapy, high-risk cytogenetics, Eastern Cooperative Oncology Group performance status ≥ 2 at lymphodepletion, and younger age had inferior progression-free survival on multivariable analysis., Conclusion: The safety and efficacy of ide-cel in patients with RRMM in the SOC setting were comparable with those in the phase II pivotal KarMMa trial despite most patients (75%) not meeting trial eligibility criteria.
- Published
- 2023
- Full Text
- View/download PDF
29. Stem Cell Mobilization Yields with Daratumumab- and Lenalidomide-Containing Quadruplet Induction Therapy in Newly Diagnosed Multiple Myeloma: Findings from the MASTER and GRIFFIN Trials.
- Author
-
Chhabra S, Callander N, Watts NL, Costa LJ, Thapa B, Kaufman JL, Laubach J, Sborov DW, Reeves B, Rodriguez C, Chari A, Silbermann R, Anderson LD Jr, Bal S, Dhakal B, Nathwani N, Shah N, Medvedova E, Bumma N, Holstein SA, Costello C, Jakubowiak A, Wildes TM, Schmidt T, Orlowski RZ, Shain KH, Cowan AJ, Dholaria B, Cornell RF, Jerkins JH, Pei H, Cortoos A, Patel S, Lin TS, Usmani SZ, Richardson PG, and Voorhees PM
- Subjects
- Adult, Humans, Lenalidomide therapeutic use, Hematopoietic Stem Cell Mobilization, Induction Chemotherapy, Transplantation, Autologous, Bortezomib therapeutic use, Dexamethasone therapeutic use, Granulocyte Colony-Stimulating Factor therapeutic use, Multiple Myeloma therapy, Hematopoietic Stem Cell Transplantation, Heterocyclic Compounds pharmacology, Heterocyclic Compounds therapeutic use
- Abstract
For eligible patients with newly diagnosed multiple myeloma (NDMM), standard of care includes induction therapy followed by autologous stem cell transplantation (ASCT). Daratumumab as monotherapy and in combination treatment is approved across multiple lines of therapy for multiple myeloma (MM), and lenalidomide is an effective and commonly used agent for induction and maintenance therapy in MM. However, there is concern that lenalidomide and daratumumab given as induction therapy might impair mobilization of stem cells for ASCT. Therefore, we assessed stem cell mobilization in patients following frontline induction therapy in the MASTER and GRIFFIN phase 2 clinical studies by examining stem cell mobilization yields, apheresis attempts, and engraftment outcomes for patients from each study. Adult transplantation-eligible patients with NDMM received induction therapy consisting of daratumumab plus carfilzomib/lenalidomide/dexamethasone (D-KRd) for four 28-day cycles in the single-arm MASTER trial or lenalidomide/bortezomib/dexamethasone (RVd) with or without daratumumab (D) for four 21-day cycles in the randomized GRIFFIN trial, followed by stem cell mobilization and ASCT in both studies. Institutional practice differed regarding plerixafor use for stem cell mobilization; the strategies were upfront (ie, planned plerixafor use) or rescue (ie, plerixafor use only after mobilization parameters indicated failure with granulocyte colony-stimulating factor [G-CSF] alone). Descriptive analyses were used to summarize patient characteristics, stem cell mobilization yields, and engraftment outcomes. In MASTER, 116 D-KRd recipients underwent stem cell mobilization and collection at a median of 24 days after completing induction therapy. In GRIFFIN, 175 patients (D-RVd, n = 95; RVd, n = 80) underwent mobilization at a median of 27 days after completing D-RVd induction therapy and 24 days after completing RVd induction therapy. Among those who underwent mobilization and collection, 7% (8 of 116) of D-KRd recipients, 2% (2 of 95) of D-RVd recipients, and 6% (5 of 80) of RVd recipients did not meet the center-specific minimally required CD34
+ cell yield in the first mobilization attempt; however, nearly all collected sufficient stem cells for ASCT on remobilization. Among patients who underwent mobilization, plerixafor use, either upfront or as a rescue strategy, was higher in patients receiving D-KRd (97%; 112 of 116) and D-RVd (72%; 68 of 95) compared with those receiving RVd (55%; 44 of 80). The median total CD34+ cell collection was 6.0 × 106 /kg (range, 2.2 to 13.9 × 106 /kg) after D-KRd induction, 8.3 × 106 /kg (range, 2.6 to 33.0 × 106 /kg) after D-RVd induction, and 9.4 × 106 /kg (range, 4.1 to 28.7 × 106 /kg) after RVd induction; the median days for collection were 2, 2, and 1, respectively. Among patients who underwent mobilization, 98% (114 of 116) of D-KRd patients, 99% (94 of 95) of D-RVd patients, and 98% (78 of 80) of RVd patients underwent ASCT using median CD34+ cell doses of 3.2 × 106 /kg, 4.2 × 106 /kg, and 4.8 × 106 /kg, respectively. The median time to neutrophil recovery was 12 days in all 3 treatment groups across the 2 trials. Because both trials used different criteria to define platelet recovery, data on platelet engraftment using the same criteria are not available. Four cycles of daratumumab- and lenalidomide-based quadruplet induction therapy had a minimal impact on stem cell mobilization and allowed predictable stem cell harvesting and engraftment in all patients who underwent ASCT. Upfront plerixafor strategy may be considered, but many patients were successfully collected with the use of G-CSF alone or rescue plerixafor., Competing Interests: Conflict of interest statement SC received honoraria from GlaxoSmithKline, Omeros, and Sanofi and institutional research funding from Janssen, Amgen, Sanofi, Bristol Myers Squibb, Allogene, Ionis, Novartis, Syndax, and GlaxoSmithKline, and is co-chair of the SWOG S1803 clinical trial. NLW received consulting fees from Bristol Myers Squibb and holds stock in Gilead. LJC served as a consultant and received honoraria from Amgen, Janssen, Bristol Myers Squibb, Karyopharm, Pfizer, and Sanofi; received research funding from Amgen, Janssen, and Bristol Myers Squibb; and served on speakers bureaus for Amgen and Sanofi. JLK served as a consultant for Genentech, AbbVie, Janssen, Incyte, Celgene, Bristol Myers Squibb, Roche/Genentech, and Tecnopharma; received research funding from Genentech, AbbVie, Janssen, Amgen, Fortis Therapeutics, Heidelberg Pharma, Novartis, Sutro Biopharma, and Takeda; received honoraria from Tecnopharma, AbbVie, Janssen, Roche/Genentech, and Tecnopharma; and held membership on an entity's board of directors or advisory committees for Incyte and TG Therapeutics. DWS served as a consultant for GlaxoSmithKline, Janssen, and Sanofi; and served as a consultant and held membership on an entity's board of directors or advisory committees for Janssen and AbbVie. BR received honoraria from Incyte, Takeda, and PharmaEssentia; served as a consultant for PharmaEssentia; and served on a speakers bureau for Bristol Myers Squibb. CR served as a consultant and on speakers bureaus for Bristol Myers Squibb, Janssen, Takeda, Amgen, and Karyopharm; and received honoraria and served as a consultant for Oncopeptides. AC served as a consultant and held membership on an entity's board of directors or advisory committees for Janssen Oncology, Bristol Myers Squibb/Celgene, Amgen, Karyopharm, Sanofi Genzyme, Oncopeptides, GlaxoSmithKline, Secura Bio, Shattuck Labs, Genentech, AbbVie, and Antengene; served as a consultant for Takeda, Novartis, and Millennium/Takeda; held membership on an entity's board of directors or advisory committee for Seattle Genetics; and received research funding from Janssen Oncology, Bristol Myers Squibb/Celgene, Amgen, Takeda, Seattle Genetics, Novartis, Pharmacyclics, and Millennium/Takeda. RS held membership on an entity's board of directors or advisory committees for Sanofi Genzyme and Janssen Pharmaceuticals and received research funding from Sanofi Genzyme. LDA served as a consultant, received honoraria, held membership on an entity's board of directors or advisory committee, and received research funding from Celgene, Bristol Myers Squibb, Janssen, GlaxoSmithKline, Karyopharm, Oncopeptides, AbbVie, BeiGene, Prothena, and Amgen. SB received grants from Amyloid Foundation and honoraria from Adaptive Biotechnology. BD served as a consultant for Janssen, Sanofi, Genentech, and AbbVie; received honoraria from Bristol Myers Squibb, GlaxoSmithKline, Sanofi, Karyopharm, and Janssen; and served on an advisory board for Janssen, Natera, Arcellx Takeda, Amgen, and Sanofi. NS served as a consultant for Amgen, CareDx, CSL Behring, GlaxoSmithKline, Indapta Therapeutics, Karyopharm, Kite, Oncopeptides, and Sanofi; and received research funding from bluebird bio, Bristol Myers Squibb/Celgene, Janssen, Nektar, Poseida, Precision BioSciences, Sutro Biopharma, and TeneoBio. NB held membership on an entity's board of directors or advisory committees for Sanofi and Janssen; and served on a speakers bureau for Sanofi and Amgen. SAH served as a consultant for BMS/Celgene, Genetech, GlaxoSmithKline, Janssen, Oncopeptides, Sanofi, Secura Bio, and Takeda and received research funding from Oncopeptides. CC received honoraria and research funding from Takeda, Janssen, Pfizer, Karyopharm, Oncopeptides, and Bristol Myers Squibb; served as a consultant for Takeda, Celgene, and Janssen; and received research funding from Celgene and Poseida. AJ held membership on an entity's board of directors or advisory committees for Sanofi, Karyopharm, Janssen, GlaxoSmithKline, Amgen, AbbVie, Bristol Myers Squibb, Gracell, and Celgene. TMW served as a consultant for Janssen, Carevive, Seattle Genetics, and Sanofi. TS served as a consultant for Sanofi and Janssen. RZO served as a consultant and received honoraria from Amgen, BioTheryX, Inc., Bristol Myers Squibb, Celgene, EcoR1 Capital LLC, Genzyme, GSK Biologicals, Janssen, Karyopharm, Neoleukin Corporation, Oncopeptides AB, Regeneron, Sanofi-Aventis, and Takeda; received research funding from CARsgen Therapeutics, Celgene, Exelixis, Janssen Biotech, Sanofi-Aventis, Takeda, Asylia Therapeutics, Inc., BioTheryX, Inc., and Heidelberg Pharma; holds individual stocks in a privately held company and patents and royalties for Asylia Therapeutics, Inc.; and held membership on an entity's board of directors or advisory committees for Amgen, BioTheryX, Inc., Bristol Myers Squibb, Celg, (Copyright © 2022 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
30. Time-to-event surrogate end-points in multiple myeloma randomised trials from 2005 to 2019: A surrogacy analysis.
- Author
-
Etekal T, Koehn K, Sborov DW, McClune B, Prasad V, Haslam A, Berger K, Booth C, Al Hadidi S, Abdallah AO, Goodman A, and Mohyuddin GR
- Subjects
- Humans, Biomarkers analysis, Progression-Free Survival, Proportional Hazards Models, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Disease-Free Survival, Randomized Controlled Trials as Topic, Multiple Myeloma therapy
- Abstract
Use of surrogate end-points such as progression-free survival (PFS) and other time-to-event (TTE) end-points is common in multiple myeloma (MM) clinical trials. This systematic review characterises all published randomised controlled trials (RCTs) in MM using PFS or other TTE end-points between 2005 and 2019 and assesses strength of surrogacy of PFS for overall survival (OS). The association between OS hazard ratios (HRs) and PFS HRs was evaluated with linear regression, and the coefficient of determination with Pearson's correlation. We identified 88 RCTs of which 67 (76%) used PFS as the primary/co-primary end-point. One trial indicated whether progression was biochemical or clinical. Of the variance in OS, 39% was due to variance in PFS. Correlation between PFS and OS was weak (0.62, 95% confidence interval [CI] 0.38-0.78). In newly diagnosed MM, 43% of the variance in OS was due to changes in PFS. The correlation between PFS and OS was weak (0.65, 95% CI 0.30-0.84). In relapsed/refractory MM, 58% of the variance in OS was due to changes in PFS. Correlation between PFS and OS was medium (0.76, 95% CI 0.42-0.91). We demonstrate that PFS and progression characteristics are characterised poorly in MM trials and that PFS is a poor surrogate for OS in MM., (© 2022 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
31. Evaluating the Impacts of CYP3A4*1B and CYP3A5*3 Variations on Pharmacokinetic Behavior and Clinical Outcomes in Multiple Myeloma Patients With Autologous Stem Cell Transplant.
- Author
-
Li J, Cho YK, Sborov DW, Phelps MA, Hofmeister CC, and Poi MJ
- Subjects
- Humans, Chromatography, Liquid, Melphalan therapeutic use, Tandem Mass Spectrometry, Genotype, Stem Cell Transplantation, Cytochrome P-450 CYP3A genetics, Multiple Myeloma drug therapy, Multiple Myeloma genetics
- Abstract
Background/aim: There exists considerably large interpatient variability in pharmacokinetic exposure of high dose melphalan in multiple myeloma patients with hematopoietic stem-cell transplantation. In this study, we aimed to evaluate the potential impacts of CYP3A4*1B (rs2940574) and CYP3A5*3 (rs776746) variations on pharmacokinetic properties of melphalan and clinical outcomes in multiple myeloma (MM) patients., Patients and Methods: Genotypes of CYP3A4*1B (rs2940574) and CYP3A5*3 (rs776746) were determined by validated gene-specific real-time PCR (RT-PCR) assays using DNA samples from 108 MM patients; plasma concentrations of melphalan at different time points were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS)., Results: CYP3A4*1B/*1B and CYP3A5*3/*3 carriers appeared to have a short median progression-free survival time and a higher maximum melphalan plasma concentration than non-carriers [792 vs. over 950 days, p=0.08; 9.91 (2.67, 34.03) vs. 8.66 (4.46, 17.61) mg/l, p=0.039]., Conclusion: CYP3A4*1B/*1B and CYP3A5*3/*3 variations might influence melphalan therapy in MM patients through yet-to-be-identified mechanisms., (Copyright © 2023, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
32. Impact of academic medical center access on outcomes in multiple myeloma.
- Author
-
Vardell VA, Ermann DA, Tantravahi SK, Haaland B, McClune B, Godara A, Mohyuddin GR, and Sborov DW
- Subjects
- United States epidemiology, Humans, Retrospective Studies, Medicaid, Academic Medical Centers, Survival Analysis, Multiple Myeloma therapy
- Abstract
Treatment at academic cancer centers (ACs) is associated with improved survival across hematologic malignancies, though the benefit in multiple myeloma (MM) has not been examined. This study aims to evaluate survival outcomes at Commission on Cancer accredited ACs compared to non-academic centers (NACs) for patients receiving MM-directed therapy. The National Cancer Database (NCDB) was used to identify demographics and overall survival (OS) of MM patients diagnosed from 2004 to 2017 and to compare outcomes by facility type. Survival analysis was repeated in a propensity score matched cohort, with NACs matched 1:1 to ACs by age, race, comorbidity score, insurance, year of diagnosis, distance traveled, and income. Of 163 375 MM patients, 44.5% were treated at ACs. Patients at ACs were more likely to receive MM-directed therapy compared to NACs (81% vs. 73%, p < .001). For patients receiving treatment, median OS at ACs was 71.3 months versus 41.2 months at NACs (p < .001). When adjusted for baseline demographics, patients treated at ACs had reduced mortality; hazard ratio (HR) 0.79 (95% CI 0.78-0.81, p < .001). The propensity score matched cohort maintained this survival benefit with a median OS of 59.9 months at ACs versus 37.0 months at NACs (p < .001), HR of 0.66 (95% CI 0.64-0.67, p < .001). ACs treated younger patients with fewer comorbidities and were more likely to treat racial minorities and patients with Medicaid or private insurance, and the uninsured. In this analysis, MM patients treated at ACs have significantly improved survival. While potentially related to access to specialized care, socioeconomic factors that drive facility selection may also contribute., (© 2022 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
33. A phase II multi-arm study of magrolimab combinations in patients with relapsed/refractory multiple myeloma.
- Author
-
Paul B, Liedtke M, Khouri J, Rifkin R, Gandhi MD, Kin A, Levy MY, Silbermann R, Cottini F, Sborov DW, Sandhu I, Villarreal L, Murphy M, Gu L, Chen A, Rajakumaraswamy N, and Usmani SZ
- Subjects
- Humans, CD47 Antigen, Dexamethasone therapeutic use, Neoplasm Recurrence, Local pathology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Clinical Trials, Phase I as Topic, Clinical Trials, Phase II as Topic, Multiple Myeloma drug therapy, Multiple Myeloma pathology
- Abstract
Magrolimab is a monoclonal antibody that blocks CD47, a 'do not eat me' signal overexpressed on tumor cells. CD47 is overexpressed in multiple myeloma (MM), which contributes to its pathogenesis. Preclinical studies have shown that CD47 blockade induces macrophage activation, resulting in elimination of myeloma cells, and that there is synergy between magrolimab and certain anticancer therapies. These findings suggest that magrolimab-based combinations may have a therapeutic benefit in MM. This phase II study investigates magrolimab in combination with commonly used myeloma therapies in patients with relapsed/refractory MM and includes a safety run-in phase followed by a dose-expansion phase. Primary end points include the incidence of dose-limiting toxicities and adverse events (safety run-in) and the objective response rate (dose expansion).
- Published
- 2023
- Full Text
- View/download PDF
34. IFITM3 regulates fibrinogen endocytosis and platelet reactivity in nonviral sepsis.
- Author
-
Campbell RA, Manne BK, Banerjee M, Middleton EA, Ajanel A, Schwertz H, Denorme F, Stubben C, Montenont E, Saperstein S, Page L, Tolley ND, Lim DL, Brown SM, Grissom CK, Sborov DW, Krishnan A, and Rondina MT
- Subjects
- Animals, Mice, Clathrin, Endocytosis, Fibrinogen genetics, Membrane Proteins genetics, Membrane Proteins metabolism, Sepsis genetics
- Abstract
Platelets and megakaryocytes are critical players in immune responses. Recent reports suggest infection and inflammation alter the megakaryocyte and platelet transcriptome to induce altered platelet reactivity. We determined whether nonviral sepsis induces differential platelet gene expression and reactivity. Nonviral sepsis upregulated IFN-induced transmembrane protein 3 (IFITM3), an IFN-responsive gene that restricts viral replication. As IFITM3 has been linked to clathrin-mediated endocytosis, we determined whether IFITM3 promoted endocytosis of α-granule proteins. IFN stimulation enhanced fibrinogen endocytosis in megakaryocytes and platelets from Ifitm+/+ mice, but not Ifitm-/- mice. IFITM3 overexpression or deletion in megakaryocytes demonstrated IFITM3 was necessary and sufficient to regulate fibrinogen endocytosis. Mechanistically, IFITM3 interacted with clathrin and αIIb and altered their plasma membrane localization into lipid rafts. In vivo IFN administration increased fibrinogen endocytosis, platelet reactivity, and thrombosis in an IFITM-dependent manner. In contrast, Ifitm-/- mice were completely rescued from IFN-induced platelet hyperreactivity and thrombosis. During murine sepsis, platelets from Ifitm+/+ mice demonstrated increased fibrinogen content and platelet reactivity, which was dependent on IFN-α and IFITMs. Platelets from patients with nonviral sepsis had increases in platelet IFITM3 expression, fibrinogen content, and hyperreactivity. These data identify IFITM3 as a regulator of platelet endocytosis, hyperreactivity, and thrombosis during inflammatory stress.
- Published
- 2022
- Full Text
- View/download PDF
35. Distinct germline genetic susceptibility profiles identified for common non-Hodgkin lymphoma subtypes.
- Author
-
Berndt SI, Vijai J, Benavente Y, Camp NJ, Nieters A, Wang Z, Smedby KE, Kleinstern G, Hjalgrim H, Besson C, Skibola CF, Morton LM, Brooks-Wilson AR, Teras LR, Breeze C, Arias J, Adami HO, Albanes D, Anderson KC, Ansell SM, Bassig B, Becker N, Bhatti P, Birmann BM, Boffetta P, Bracci PM, Brennan P, Brown EE, Burdett L, Cannon-Albright LA, Chang ET, Chiu BCH, Chung CC, Clavel J, Cocco P, Colditz G, Conde L, Conti DV, Cox DG, Curtin K, Casabonne D, De Vivo I, Diepstra A, Diver WR, Dogan A, Edlund CK, Foretova L, Fraumeni JF Jr, Gabbas A, Ghesquières H, Giles GG, Glaser S, Glenn M, Glimelius B, Gu J, Habermann TM, Haiman CA, Haioun C, Hofmann JN, Holford TR, Holly EA, Hutchinson A, Izhar A, Jackson RD, Jarrett RF, Kaaks R, Kane E, Kolonel LN, Kong Y, Kraft P, Kricker A, Lake A, Lan Q, Lawrence C, Li D, Liebow M, Link BK, Magnani C, Maynadie M, McKay J, Melbye M, Miligi L, Milne RL, Molina TJ, Monnereau A, Montalvan R, North KE, Novak AJ, Onel K, Purdue MP, Rand KA, Riboli E, Riby J, Roman E, Salles G, Sborov DW, Severson RK, Shanafelt TD, Smith MT, Smith A, Song KW, Song L, Southey MC, Spinelli JJ, Staines A, Stephens D, Sutherland HJ, Tkachuk K, Thompson CA, Tilly H, Tinker LF, Travis RC, Turner J, Vachon CM, Vajdic CM, Van Den Berg A, Van Den Berg DJ, Vermeulen RCH, Vineis P, Wang SS, Weiderpass E, Weiner GJ, Weinstein S, Doo NW, Ye Y, Yeager M, Yu K, Zeleniuch-Jacquotte A, Zhang Y, Zheng T, Ziv E, Sampson J, Chatterjee N, Offit K, Cozen W, Wu X, Cerhan JR, Chanock SJ, Slager SL, and Rothman N
- Subjects
- Humans, Genome-Wide Association Study, Risk Factors, Germ Cells, Case-Control Studies, Polymorphism, Single Nucleotide, Genetic Predisposition to Disease, Lymphoma, Non-Hodgkin genetics
- Abstract
Lymphoma risk is elevated for relatives with common non-Hodgkin lymphoma (NHL) subtypes, suggesting shared genetic susceptibility across subtypes. To evaluate the extent of mutual heritability among NHL subtypes and discover novel loci shared among subtypes, we analyzed data from eight genome-wide association studies within the InterLymph Consortium, including 10,629 cases and 9505 controls. We utilized Association analysis based on SubSETs (ASSET) to discover loci for subsets of NHL subtypes and evaluated shared heritability across the genome using Genome-wide Complex Trait Analysis (GCTA) and polygenic risk scores. We discovered 17 genome-wide significant loci (P < 5 × 10
-8 ) for subsets of NHL subtypes, including a novel locus at 10q23.33 (HHEX) (P = 3.27 × 10-9 ). Most subset associations were driven primarily by only one subtype. Genome-wide genetic correlations between pairs of subtypes varied broadly from 0.20 to 0.86, suggesting substantial heterogeneity in the extent of shared heritability among subtypes. Polygenic risk score analyses of established loci for different lymphoid malignancies identified strong associations with some NHL subtypes (P < 5 × 10-8 ), but weak or null associations with others. Although our analyses suggest partially shared heritability and biological pathways, they reveal substantial heterogeneity among NHL subtypes with each having its own distinct germline genetic architecture., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
36. Palliative care utilization, transfusion burden, and end-of-life care for patients with multiple myeloma.
- Author
-
McInturf G, Younger K, Sanchez C, Walde C, Abdallah AO, Ahmed N, Shune L, Sborov DW, Godara A, McClune B, Sinclair CT, and Mohyuddin GR
- Subjects
- Humans, Palliative Care, Retrospective Studies, Hospice Care, Multiple Myeloma diagnosis, Multiple Myeloma epidemiology, Multiple Myeloma therapy, Terminal Care
- Abstract
Introduction: Despite treatment advances, multiple myeloma (MM) remains a significant source of morbidity and mortality. We aimed to examine specialist palliative care (SPC) involvement and end-of-life care for patients with MM., Methods: We assessed all deceased patients with a diagnosis of MM who received care at a single institution from January 2010 to December 2019 and assessed SPC involvement., Results: We reviewed 456 deceased patients. Overall, 207 patients (45.4%) received SPC visits by clinicians during their disease, and 153 (33.5%) were on MM treatment in the month before death. Median time from SPC consultation to death was 1 month, with 42 (9.2%) of patients receiving SPC visits 6 or more months before death. Amongst the patients for which a place of death was reported (351), 117 (33.3%) died in the acute care setting. Outpatient SPC did not correlate with a reduction of death in the acute care setting. In the group of patients who received outpatient SPC, 22/84 (26.2%) died in an acute care setting, whereas 95/267 (35.5%) patients who did not receive outpatient SPC also died in an acute care setting, (p = .11)., Conclusion: In our analysis of the entire trajectory of the MM patient experience from diagnosis to death, we found low rates of SPC involvement and a significant proportion of patients receiving aggressive care at end-of-life. While there is no clear correlation that SPC involvement impacted the rate of acute care deaths or decreased utilization of MM treatment in the last month of life, further prospective research on optimal utilization of SPC is required., (© 2022 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
37. Daratumumab plus lenalidomide, bortezomib and dexamethasone in newly diagnosed multiple myeloma: Analysis of vascular thrombotic events in the GRIFFIN study.
- Author
-
Sborov DW, Baljevic M, Reeves B, Laubach J, Efebera YA, Rodriguez C, Costa LJ, Chari A, Silbermann R, Holstein SA, Anderson LD Jr, Kaufman JL, Shah N, Pei H, Patel S, Cortoos A, Bartlett JB, Vermeulen J, Lin TS, Voorhees PM, and Richardson PG
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Aspirin therapeutic use, Bortezomib, Dexamethasone, Lenalidomide, Transplantation, Autologous, Hematopoietic Stem Cell Transplantation, Multiple Myeloma therapy, Venous Thromboembolism prevention & control, Venous Thromboembolism chemically induced
- Abstract
Patients with multiple myeloma are at increased risk of vascular thromboembolic events (VTEs). This post hoc analysis evaluated VTEs in the randomised phase 2 GRIFFIN study (ClinicalTrials.gov Identifier: NCT02874742) that investigated lenalidomide/bortezomib/dexamethasone (RVd) ± daratumumab (D). Patients with newly diagnosed multiple myeloma who were eligible for autologous stem cell transplantation (ASCT) received D-RVd/RVd induction, high-dose therapy and ASCT, D-RVd/RVd consolidation and up to 2 years of lenalidomide maintenance therapy ± D. VTE prophylaxis was recommended (at least aspirin, ≥162 mg daily) in accordance with International Myeloma Working Group guidelines. In the safety population (D-RVd, n = 99; RVd, n = 102), VTEs occurred in 10.1% of D-RVd patients and 15.7% of RVd patients; grade 2-4 VTEs occurred in 9.1% and 14.7%, respectively. Median time to the first onset of VTE was longer for D-RVd versus RVd patients (305 days vs 119 days). Anti-thrombosis prophylaxis use was similar between arms (D-RVd, 84.8% vs RVd, 83.3%); among patients with VTEs, prophylaxis use at time of first VTE onset was 60.0% for D-RVd and 68.8% for RVd. In summary, the addition of daratumumab to RVd did not increase the incidence of VTEs, but the cumulative VTE incidence was relatively high in this cohort and anti-thrombotic prophylaxis use was suboptimal., (© 2022 Janssen Scientific Affairs, LLC. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
38. Global Myeloma Trial Participation and Drug Access in the Era of Novel Therapies.
- Author
-
Fatoki RA, Koehn K, Kelkar A, Al Hadidi S, Mehra N, Mian H, Landgren O, Kazandjian D, Hoffman J, Sborov DW, and Mohyuddin GR
- Subjects
- Ethnicity, Humans, Pharmaceutical Preparations, United States, United States Food and Drug Administration, Drug Approval, Multiple Myeloma drug therapy
- Abstract
Purpose: The globalization of clinical trials has accelerated recent advances in multiple myeloma (MM). However, it is unclear whether trial enrollment locations are reflective of the global burden of MM and whether access to novel therapies is timely and equitable for countries that participate in those trials., Methods: To assess this, we characterized where MM trials that led to US Food and Drug Administration (FDA) approvals were conducted and determined how often and quickly these drug regimens received approval in their participating trial countries on the basis of country income level and geographic region., Results: A systematic review was conducted to identify all MM clinical trials that met their primary endpoint, enrolled patients outside the United States, and resulted in FDA approval from 2005 to 2019. A total of 18 pivotal MM clinical trials were identified. High-income countries enrolled patients in 100% (18/18) of the trials identified, whereas upper-middle and lower-middle-income countries were represented in 61% (11/18) and 28% (5/18) of trials, respectively. No patients from low-income countries were enrolled. One trial enrolled patients in sub-Saharan Africa, and no trials enrolled patients in South Asia/Caribbean. For drugs/regimens that were approved in their participating countries, the median time from FDA approval to approval was 10.9 months. There were no drugs approved in lower-middle-income trial countries. MM trials leading to FDA approval are generally run in high-income, European, and Central Asian countries., Conclusion: There are substantial disparities in where novel therapies are evaluated and where they are ultimately approved for use on the basis of income level and geography., Competing Interests: Douglas W. SborovConsulting or Advisory Role: Sanofi, GlaxoSmithKline, Bristol Myers Squibb/Celgene, Legend Biotech, Janssen, Skyline Diagnostics Dickran KazandjianHonoraria: Arcellx, BMS, SanofiSpeakers' Bureau: Curio Science, Aptitude Health, Cure Educated Patient, Plexus Hira MianHonoraria: Celgene, Janssen, Takeda, Pfizer, Amgen, GlaxoSmithKlineConsulting or Advisory Role: Amgen, TakedaResearch Funding: Janssen Oncology Ola LandgrenHonoraria: Celgene, Bristol Myers Squibb, Medscape, Amgen, Janssen, Karyopharm TherapeuticsConsulting or Advisory Role: Bristol Myers Squibb, Celgene, Janssen, GlaxoSmithKlineResearch Funding: Amgen (Inst), Janssen (Inst), Pfizer (Inst) James HoffmanStock and Other Ownership Interests: SyndaxHonoraria: Bristol Myers Squibb/CelgeneNo potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
39. Toxicity management strategies for next-generation novel therapeutics in multiple myeloma.
- Author
-
Steinbach M, Julian K, McClune B, and Sborov DW
- Abstract
The therapeutic options available for patients with multiple myeloma have greatly expanded over the past decade and incorporating these novel agents into routine clinical practice has significantly improved outcomes. The next generation of therapeutics is available for relapsed and refractory patients either as standard of care or in clinical trial, and these drugs represent a generational paradigm shift. Patients now have access to a multitude of novel immunotherapeutics, including monoclonal antibodies, an antibody-drug conjugate, chimeric antigen receptor T-cells (CAR-T), and bispecific T-cell redirecting antibodies, and novel oral therapies including selinexor (selective inhibitor of nuclear export) and venetoclax (bcl-2 inhibitor). While these drugs have the potential to be highly efficacious in certain subsets of patients when used as single agents or in combination regimens, they are each associated with unique toxicity profiles. It is imperative to understand these potential adverse events to ensure patient safety. Appropriate supportive care management is paramount to maximize drug exposure and therapeutic efficacy. The following review focuses its discussion on drugs and combination regimens that are currently FDA-approved and those that continue to be investigated in clinical trials, highlights the clinically relevant toxicity profiles for each of the different agents, and provides practical considerations for the treatment team., Competing Interests: Conflict of interest statement: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: M.S. is on the speakers’ bureau for GlaxoSmithKline, Karyopharm Therapeutics, and Janssen Pharmaceuticals. K.J. and B.L.M. have no relevant disclosures. D.W.S. is an advisor/consultant for Janssen Pharmaceuticals, Celgene, Abbvie, Sanofi, and GlaxoSmithKline., (© The Author(s), 2022.)
- Published
- 2022
- Full Text
- View/download PDF
40. Heterogeneity of enrolment criteria for ongoing smouldering myeloma trials.
- Author
-
Soomro MA, Hoffman J, Goodman AM, Sborov DW, and Mohyuddin GR
- Subjects
- Humans, Longitudinal Studies, Multiple Myeloma diagnosis, Multiple Myeloma therapy, Smoldering Multiple Myeloma
- Published
- 2022
- Full Text
- View/download PDF
41. Characteristics of clinical trials for haematological malignancies from 2015 to 2020: A systematic review.
- Author
-
Wesson W, Galate VL, Sborov DW, McClune B, Goodman AM, Gyawali B, Prasad V, Abbasi S, and Mohyuddin GR
- Subjects
- Humans, Hematologic Neoplasms therapy, Quality of Life
- Abstract
Background: As the landscape of haematological malignancies dramatically changes due to diagnostic and therapeutic advances, it is important to evaluate trends in clinical trial designs. The objective of our study was to describe the design of clinical trials for five common haematological malignancies with respect to randomisation and end-points. We also aimed to assess trends over time and examine the relationships of funding source and country of origin to proportions of randomisation and utilisation of clinical end-points., Methods: This systematic review identified haematological malignancy clinical trials starting in 2015-2020 registered at ClinicalTrials.gov as of 20th February 2021. Trial-related variables including randomisation status, type of primary end-point, and both projected and actual enrolment numbers were captured. Clinical end-points were defined as overall survival and quality of life, while surrogate end-points included all other end-points., Results: Of 2609 relevant trials included in this analysis, only one-fifth were randomised (538, 21%), with a significant decrease in the proportion of randomised clinical trials from 26% of trials in 2015 to 19% in 2020 (p < 0.00001). Between the years 2015 and 2020, the proportion of randomised trials for all haematological malignancies using primary surrogate end-points remained relatively consistent, ranging from 84% in 2015 to 78% in 2020 (p = 0.352). Overall, only 15% of trials utilised primary end-points of overall survival or quality of life in a randomised design., Conclusions: This systematic review of haematological malignancy trials found that the majority of trials are non-randomised and that there has been an increase in the ratio of non-randomised to randomised studies over time. The vast majority of randomised haematological malignancy trials use surrogate primary end-points., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests, although the study did not recieve any funding from these sources: Vinay Prasad reports royalties from Johns Hopkins Press, Medscape, MedPage, consulting for UnitedHealthcare, and speaking fees for Evicore. Vinay Prasad has a plenary session podcast that has Patreon backers. Vinay Prasad is funded to study low value drugs through a grant from Arnold Ventures. The funder had no role in the design of this study. Aaron Goodman reports consulting for Seattle Genetics and EUSA Pharma. Douglas Sborov reports consulting for Janssen, SkylinDx, GlaxoSmithKline, Legend Biotech, Amgen and Celgene. None of the authors have any other conflicts of interest., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
42. A Single Nucleotide Polymorphism (SNP) in the SLC22A3 Transporter Gene Is Associated With the Severity of Oral Mucositis in Multiple Myeloma Patients Receiving Autologous Stem Cell Transplant Followed by Melphalan Therapy.
- Author
-
Li J, Persaud AK, Johnson JA, Sborov DW, Phelps MA, Hofmeister CC, and Poi MJ
- Subjects
- Adult, Aged, Genetic Association Studies, Genotype, Humans, Male, Melphalan adverse effects, Melphalan therapeutic use, Middle Aged, Multiple Myeloma complications, Multiple Myeloma genetics, Multiple Myeloma pathology, Polymorphism, Single Nucleotide genetics, Stem Cell Transplantation adverse effects, Stomatitis epidemiology, Stomatitis pathology, Transplantation, Autologous adverse effects, Genetic Predisposition to Disease, Multiple Myeloma therapy, Organic Cation Transport Proteins genetics, Stomatitis genetics
- Abstract
Background: It has been reported that expression of OCT3 enhanced the sensitivity to melphalan in cells, indicative of potential roles of OCT3 in melphalan transport. Herein we investigated the association of select single nucleotide polymorphisms in SLC22A3 (gene encoding OCT3) with clinical outcomes in multiple myeloma (MM) patients with hematopoietic autologous stem cell transplants followed by high-dose melphalan therapy., Materials and Methods: Melphlan concentrations in blood samples from 108 MM patients were measured using liquid chromatography-tandem mass spectrometry (LC-MS/ΜS); genotypes of rs2048327, rs1810126, and rs3088442 in these patients were determined using quatitive RT-PCR assays., Results: Rs3088442 A variant-carriers had a significantly increased risk of severe oral mucositis in comparison with homozygous rs3088442 G-carriers with adjusted odds ratio of 4.00 (95% CI=1.25-14.7; p=0.027). Rs3088442 A carriers tended to have lower creatinine clearance (p=0.10) and higher maximum plasma concentration of melphalan (p=0.07)., Conclusion: OCT3 might be involved in melphalan transport in MM patients., (Copyright © 2022 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
43. Optimizing Thromboembolism Prophylaxis for the Contemporary Age of Multiple Myeloma.
- Author
-
Baljevic M, Sborov DW, Lim MY, Hillengass J, Martin T, Castillo JJ, Streiff MB, Kumar SK, and Callander NS
- Subjects
- Anticoagulants adverse effects, Humans, Lenalidomide adverse effects, Risk Factors, Thalidomide adverse effects, Multiple Myeloma complications, Multiple Myeloma drug therapy, Venous Thromboembolism epidemiology, Venous Thromboembolism etiology, Venous Thromboembolism prevention & control
- Abstract
Venous thromboembolism (VTE) is a major complication in all patients with cancer. Compared with the general population, patients with multiple myeloma (MM) have a 9-fold increase in VTE risk, likely because of their malignancy, its treatments, and other additional patient-related factors. In MM, thromboembolism events tend to occur within 6 months of treatment initiation, regardless of treatment regimen; however, the use of immunomodulatory agents such as thalidomide or lenalidomide, especially in combination with dexamethasone or multiagent chemotherapy, is known to create a significant risk for VTE. Currently, official recommendations for VTE prophylaxis in MM outlined in various national guidelines or multidisciplinary society panels are based on expert opinion, because data from randomized controlled trials are scarce. Large studies which have mainly focused on the efficacy of thromboprophylaxis in patients with cancer at higher risk for VTE either had a very low representation of patients with MM, or excluded them all together, limiting our ability to draw evidence-based conclusions on how to effectively protect MM population from VTE. In this brief perspective, we highlight some of the greatest challenges that have hampered the field concerning the availability of high-quality clinical trial data for the formulation of best VTE prophylaxis strategies in patients with newly diagnosed MM, as well as the rationale for the latest updates in the NCCN Guidelines on this topic.
- Published
- 2022
- Full Text
- View/download PDF
44. The evaluation and management of monoclonal gammopathy of renal significance and monoclonal gammopathy of neurological significance.
- Author
-
Castillo JJ, Callander NS, Baljevic M, Sborov DW, and Kumar S
- Subjects
- Animals, Disease Management, Disease Progression, Humans, Kidney Diseases diagnosis, Kidney Diseases etiology, Kidney Diseases pathology, Kidney Diseases therapy, Monoclonal Gammopathy of Undetermined Significance complications, Monoclonal Gammopathy of Undetermined Significance diagnosis, Monoclonal Gammopathy of Undetermined Significance therapy, Nervous System Diseases diagnosis, Nervous System Diseases etiology, Nervous System Diseases pathology, Nervous System Diseases therapy, Kidney pathology, Monoclonal Gammopathy of Undetermined Significance pathology, Nervous System pathology
- Abstract
Despite the benign nature of monoclonal gammopathy of undetermined significance (MGUS), mounting data are associating MGUS with the development of organ dysfunction, specifically monoclonal gammopathy of renal significance (MGRS) and monoclonal gammopathy of neurological significance (MGNS), which could be associated with substantial morbidity. Emerging evidence suggests that patients with MGRS and MGNS could benefit from treatments used for myeloma, Waldenström macroglobulinemia, or chronic lymphocytic leukemia, depending on the underlying pathology. However, the treatment of MGRS and MGNS is not standardized, and potentially effective therapies might not be reimbursed because these conditions do not formally meet the criteria for malignant processes. The present review aims at establishing standards for the evaluation and management of MGRS and MGNS, which can facilitate the diagnosis of and provide therapeutic options for treating practitioners and patients affected by these conditions. The careful design and execution of clinical trials for patients with MGRS and MGNS are positively encouraged., (© 2021 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
45. Sequencing at lymphoid neoplasm susceptibility loci maps six myeloma risk genes.
- Author
-
Waller RG, Klein RJ, Vijai J, McKay JD, Clay-Gilmour A, Wei X, Madsen MJ, Sborov DW, Curtin K, Slager SL, Offit K, Vachon CM, Lipkin SM, Dumontet C, and Camp NJ
- Subjects
- Acyl-CoA Oxidase genetics, Female, Genetic Predisposition to Disease, Hodgkin Disease genetics, Hodgkin Disease pathology, Humans, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Lymphocytes pathology, Lymphoma, Follicular genetics, Lymphoma, Follicular pathology, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Male, Multiple Myeloma pathology, Polymorphism, Single Nucleotide genetics, Risk Factors, Tetraspanins genetics, Transmembrane Activator and CAML Interactor Protein genetics, Exome Sequencing, Butyrophilins genetics, Genome-Wide Association Study, Interferon Regulatory Factors genetics, Multiple Myeloma genetics, T-Box Domain Proteins genetics
- Abstract
Inherited genetic risk factors play a role in multiple myeloma (MM), yet considerable missing heritability exists. Rare risk variants at genome-wide association study (GWAS) loci are a new avenue to explore. Pleiotropy between lymphoid neoplasms (LNs) has been suggested in family history and genetic studies, but no studies have interrogated sequencing for pleiotropic genes or rare risk variants. Sequencing genetically enriched cases can help discover rarer variants. We analyzed exome sequencing in familial or early-onset MM cases to identify rare, functionally relevant variants near GWAS loci for a range of LNs. A total of 149 distinct and significant LN GWAS loci have been published. We identified six recurrent, rare, potentially deleterious variants within 5 kb of significant GWAS single nucleotide polymorphisms in 75 MM cases. Mutations were observed in BTNL2, EOMES, TNFRSF13B, IRF8, ACOXL and TSPAN32. All six genes replicated in an independent set of 255 early-onset MM or familial MM or precursor cases. Expansion of our analyses to the full length of these six genes resulted in a list of 39 rare and deleterious variants, seven of which segregated in MM families. Three genes also had significant rare variant burden in 733 sporadic MM cases compared with 935 control individuals: IRF8 (P = 1.0 × 10-6), EOMES (P = 6.0 × 10-6) and BTNL2 (P = 2.1 × 10-3). Together, our results implicate six genes in MM risk, provide support for genetic pleiotropy between LN subtypes and demonstrate the utility of sequencing genetically enriched cases to identify functionally relevant variants near GWAS loci., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
46. A Phase 1 dose-escalation study of disulfiram and copper gluconate in patients with advanced solid tumors involving the liver using S-glutathionylation as a biomarker.
- Author
-
Kelley KC, Grossman KF, Brittain-Blankenship M, Thorne KM, Akerley WL, Terrazas MC, Kosak KM, Boucher KM, Buys SS, McGregor KA, Werner TL, Agarwal N, Weis JR, Sharma S, Ward JH, Kennedy TP, Sborov DW, and Shami PJ
- Subjects
- Adult, Aged, Aged, 80 and over, Disulfiram adverse effects, Dose-Response Relationship, Drug, Female, Gluconates adverse effects, Humans, Male, Middle Aged, Neoplasms metabolism, Disulfiram administration & dosage, Gluconates administration & dosage, Glutathione metabolism, Liver Neoplasms secondary, Neoplasms drug therapy
- Abstract
Background: Disulfiram and metals inactivate key oncoproteins resulting in anti-neoplastic activity. The goal of this study was to determine the maximum tolerated dose of copper when administered with disulfiram in patients with advanced solid tumors and liver involvement., Methods: Disulfiram 250 mg was administered daily in 28-day cycles. Four doses of copper gluconate were tested (2, 4, 6, and 8 mg of elemental copper) in a standard 3 + 3 dose escalation design. Patients were evaluated for dose limiting toxicities and response. Protein S-glutathionylation was evaluated as a pharmacodynamic marker., Results: Twenty-one patients were enrolled and 16 patients were evaluable for dose limiting toxicities. Among the 21 patients, there was a median of 4 lines of prior chemotherapy. Five Grade 3 toxicities were observed (anorexia, elevated aspartate aminotransferase or AST, elevated alkaline phosphatase, fever, and fatigue). Response data was available for 15 patients. Four patients had stable disease with the longest duration of disease control being 116 days. The median duration of treatment for evaluable patients was 55 days (range 28-124). Reasons for discontinuation included functional decline, disease progression, and disease-associated death. Increased S-glutathionylation of serum proteins was observed with treatment., Conclusion: Disulfiram 250 mg daily with copper gluconate (8 mg of elemental copper) was well-tolerated in patients with solid tumors involving the liver and was not associated with dose limiting toxicities. While temporary disease stabilization was noted in some patients, no objective responses were observed. Treatment was associated with an increase in S-glutathionylation suggesting that this combination could exert a suppressive effect on cellular growth and protein function., Trial Registration: NCT00742911 , first posted 28/08/2008.
- Published
- 2021
- Full Text
- View/download PDF
47. A phase 1 trial of the histone deacetylase inhibitor AR-42 in patients with neurofibromatosis type 2-associated tumors and advanced solid malignancies.
- Author
-
Collier KA, Valencia H, Newton H, Hade EM, Sborov DW, Cavaliere R, Poi M, Phelps MA, Liva SG, Coss CC, Wang J, Khountham S, Monk P, Shapiro CL, Piekarz R, Hofmeister CC, Welling DB, and Mortazavi A
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Neoplasms mortality, Neurofibromatosis 2 mortality, Phenylbutyrates adverse effects, Phenylbutyrates pharmacokinetics, Young Adult, Histone Deacetylase Inhibitors therapeutic use, Neoplasms drug therapy, Neurofibromatosis 2 drug therapy, Phenylbutyrates therapeutic use
- Abstract
Purpose: Given clinical activity of AR-42, an oral histone deacetylase inhibitor, in hematologic malignancies and preclinical activity in solid tumors, this phase 1 trial investigated the safety and tolerability of AR-42 in patients with advanced solid tumors, including neurofibromatosis type 2-associated meningiomas and schwannomas (NF2). The primary objective was to define the maximum tolerated dose (MTD) and dose-limiting toxicities (DLTs). Secondary objectives included determining pharmacokinetics and clinical activity., Methods: This phase I trial was an open-label, single-center, dose-escalation study of single-agent AR-42 in primary central nervous system and advanced solid tumors. The study followed a 3 + 3 design with an expansion cohort at the MTD., Results: Seventeen patients were enrolled with NF2 (n = 5), urothelial carcinoma (n = 3), breast cancer (n = 2), non-NF2-related meningioma (n = 2), carcinoma of unknown primary (n = 2), small cell lung cancer (n = 1), Sertoli cell carcinoma (n = 1), and uveal melanoma (n = 1). The recommended phase II dose is 60 mg three times weekly, for 3 weeks of a 28-day cycle. DLTs included grade 3 thrombocytopenia and grade 4 psychosis. The most common treatment-related adverse events were cytopenias, fatigue, and nausea. The best response was stable disease in 53% of patients (95% CI 26.6-78.7). Median progression-free survival (PFS) was 3.6 months (95% CI 1.2-9.1). Among evaluable patients with NF2 or meningioma (n = 5), median PFS was 9.1 months (95% CI 1.9-not reached)., Conclusion: Single-agent AR-42 is safe and well tolerated. Further studies may consider AR-42 in a larger cohort of patients with NF2 or in combination with other agents in advanced solid tumors., Trial Registration: NCT01129193, registered 5/24/2010.
- Published
- 2021
- Full Text
- View/download PDF
48. Chimeric antigen receptor T-cell therapy in multiple myeloma: a systematic review and meta-analysis of 950 patients.
- Author
-
Mohyuddin GR, Rooney A, Balmaceda N, Aziz M, Sborov DW, McClune B, and Kumar SK
- Subjects
- Cell- and Tissue-Based Therapy, Humans, Immunotherapy, Adoptive, T-Lymphocytes, Multiple Myeloma therapy, Receptors, Chimeric Antigen genetics
- Published
- 2021
- Full Text
- View/download PDF
49. Oncolytic herpes simplex virus infects myeloma cells in vitro and in vivo .
- Author
-
Ghose J, Dona A, Murtadha M, Gunes EG, Caserta E, Yoo JY, Russell L, Jaime-Ramirez AC, Barwick BG, Gupta VA, Sanchez JF, Sborov DW, Rosen ST, Krishnan A, Boise LH, Kaur B, Hofmeister CC, and Pichiorri F
- Abstract
Because most patients with multiple myeloma (MM) develop resistance to current regimens, novel approaches are needed. Genetically modified, replication-competent oncolytic viruses exhibit high tropism for tumor cells regardless of cancer stage and prior treatment. Receptors of oncolytic herpes simplex virus 1 (oHSV-1), NECTIN-1, and HVEM are expressed on MM cells, prompting us to investigate the use of oHSV-1 against MM. Using oHSV-1-expressing GFP, we found a dose-dependent increase in the GFP
+ signal in MM cell lines and primary MM cells. Whereas NECTIN-1 expression is variable among MM cells, we discovered that HVEM is ubiquitously and highly expressed on all samples tested. Expression of HVEM was consistently higher on CD138+ /CD38+ plasma cells than in non-plasma cells. HVEM blocking demonstrated the requirement of this receptor for infection. However, we observed that, although oHSV-1 could efficiently infect and kill all MM cell lines tested, no viral replication occurred. Instead, we identified that oHSV-1 induced MM cell apoptosis via caspase-3 cleavage. We further noted that oHSV-1 yielded a significant decrease in tumor volume in two mouse xenograft models. Therefore, oHSV-1 warrants exploration as a novel potentially effective treatment option in MM, and HVEM should be investigated as a possible therapeutic target., Competing Interests: C.C.H. reports grants from Janssen, Bristol-Meyers Squibb, and Oncolytics Biotech, grants and personal fees for membership of a marketing advisory board from Celgene, personal fees for membership of a research advisory board from Karyopharm, personal fees for membership of a research advisory board from Oncopeptides and Imbrium Therapeutics, and personal fees for membership of a regulatory advisory board from Adaptive Biotechnologies, all outside the submitted work. D.W.S. reports grants from Oncolytics Biotech, Bristol-Meyers Squibb, GlaxoSmithKline, and Janssen, all outside the submitted work. L.H.B reports research funding from AstraZeneca and personal fees for membership of advisory boards from AstraZeneca and Genentech., (© 2021 The Authors.)- Published
- 2021
- Full Text
- View/download PDF
50. Duo Shared Genomic Segment analysis identifies a genome-wide significant risk locus at 18q21.33 in myeloma pedigrees.
- Author
-
Griffin Waller R, Madsen MJ, Gardner J, Sborov DW, and Camp NJ
- Abstract
Aim: High-risk pedigrees ( HRPs ) are a powerful design to map highly penetrant risk genes. We previously described Shared Genomic Segment (SGS) analysis, a mapping method for single large extended pedigrees that also addresses genetic heterogeneity inherent in complex diseases. SGS identifies shared segregating chromosomal regions that may inherit in only a subset of cases. However, single large pedigrees that are individually powerful (at least 15 meioses between studied cases) are scarce. Here, we expand the SGS strategy to incorporate evidence from two extended HRPs by identifying the same segregating risk locus in both pedigrees and allowing for some relaxation in the size of each HRP., Methods: Duo-SGS is a procedure to combine single-pedigree SGS evidence. It implements statistically rigorous duo-pedigree thresholding to determine genome-wide significance levels that account for optimization across pedigree pairs. Single-pedigree SGS identifies optimal segments shared by case subsets at each locus across the genome, with nominal significance assessed empirically. Duo-SGS combines the statistical evidence for SGS segments at the same genomic location in two pedigrees using Fisher's method. One pedigree is paired with all others and the best duo-SGS evidence at each locus across the genome is established. Genome-wide significance thresholds are determined through distribution-fitting and the Theory of Large Deviations. We applied the duoSGS strategy to eleven extended, myeloma HRPs., Results: We identified one genome-wide significant region at 18q21.33 (0.85 Mb, P = 7.3 × 10
-9 ) which contains one gene, CDH20 . Thirteen regions were genome-wide suggestive: 1q42.2, 2p16.1, 3p25.2, 5q21.3, 5q31.1, 6q16.1, 6q26, 7q11.23, 12q24.31, 13q13.3, 18p11.22, 18q22.3 and 19p13.12., Conclusion: Our results provide novel risk loci with segregating evidence from multiple HRPs and offer compelling targets and specific segment carriers to focus a future search for functional variants involved in inherited risk formyeloma., Competing Interests: Conflicts of interest All authors declared that there are no conflicts of interest.- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.