31 results on '"Nizar Bahlis"'
Search Results
2. Doxycycline Plus Bortezomib-Containing Regimens for the Treatment of Light-Chain Amyloidosis in the Frontline Setting: Experience from the Amyloidosis Program of Calgary
- Author
-
Ellen Lewis, Nowell Fine, Sylvia McCulloch, Jason Tay, Peter Duggan, Paola Neri, Nizar Bahlis, and Victor H. Jimenez-Zepeda
- Subjects
light-chain amyloidosis ,bortezomib ,doxycycline ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Pre-clinical and retrospective data suggest that doxycycline added to treatment regimens has benefit in AL amyloidosis. However, a recent multicenter, open-label, randomized controlled trial in AL amyloidosis patients treated with CyBorD did not demonstrate a progression-free survival (PFS) or cardiac PFS benefit with added doxycycline. Objective: The main objective of this study was to explore the role of doxycycline combined with bortezomib-containing regimens (BCRs) for newly diagnosed AL amyloidosis patients with cardiac involvement and to compare them with a cohort of concurrent patients treated with BCR only. Material and Methods: AL amyloidosis patients, newly diagnosed between January 2012 and March 2022, who were treated with BCR at the Amyloidosis Program of Calgary (APC) were evaluated. Results: Sixty-four concurrent patients were identified. Thirty-nine patients received doxycycline in addition to BCR (BCR-D) for a median of 8 months. The overall response rate was similar among the groups. No significant differences in VGPR/CR, dFLC at 1 month, time to first response, time to best response, or organ responses were noted between the BCR alone and BCR-D groups. Summary and Conclusions: Our retrospective study demonstrated that doxycycline combined with BCR failed to prolong OS, PFS, or cardiac responses compared with BCR alone in patients with cardiac AL amyloidosis.
- Published
- 2024
- Full Text
- View/download PDF
3. GPRC5D as a novel target for the treatment of multiple myeloma: a narrative review
- Author
-
Paula Rodriguez-Otero, Niels W. C. J. van de Donk, Kodandaram Pillarisetti, Ingrid Cornax, Deeksha Vishwamitra, Kathleen Gray, Brandi Hilder, Jaszianne Tolbert, Thomas Renaud, Tara Masterson, Christoph Heuck, Colleen Kane, Raluca Verona, Philippe Moreau, Nizar Bahlis, and Ajai Chari
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Multiple myeloma is a genetically complex and heterogenous malignancy with a 5-year survival rate of approximately 60%. Despite advances in therapy, patients experience cycles of remission and relapse, with each successive line of therapy associated with poorer outcomes; therefore, therapies with different mechanisms of action against new myeloma antigens are needed. G protein–coupled receptor class C group 5 member D (GPRC5D) has emerged as a novel therapeutic target for the treatment of multiple myeloma. We review the biology and target validation of GPRC5D, and clinical data from early phase trials of GPRC5D-targeting bispecific antibodies, talquetamab and forimtamig, and chimeric antigen receptor T cell (CAR-T) therapies, MCARH109, OriCAR-017, and BMS-986393. In addition to adverse events (AEs) associated with T-cell–redirection therapies irrespective of target, a consistent pattern of dermatologic and oral AEs has been reported across several trials of GPRC5D-targeting bispecific antibodies, as well as rare cerebellar events with CAR-T therapy. Additional studies are needed to understand the underlying mechanisms involved in the development of skin- and oral-related toxicities. We review the strategies that have been used to manage these GPRC5D-related toxicities. Preliminary efficacy data showed overall response rates for GPRC5D-targeting T-cell–redirecting therapies were ≥64%; most responders achieved a very good partial response or better. Pharmacokinetics/pharmacodynamics showed that these therapies led to cytokine release and T-cell activation. In conclusion, results from early phase trials of GPRC5D-targeting T-cell–redirecting agents have shown promising efficacy and manageable safety profiles, including lower infection rates compared with B-cell maturation antigen- and Fc receptor-like protein 5-targeting bispecific antibodies. Further clinical trials, including those investigating GPRC5D-targeting T-cell–redirecting agents in combination with other anti-myeloma therapies and with different treatment modalities, may help to elucidate the future optimal treatment regimen and sequence for patients with multiple myeloma and improve survival outcomes. Video Summary
- Published
- 2024
- Full Text
- View/download PDF
4. Correction: GPRC5D as a novel target for the treatment of multiple myeloma: a narrative review
- Author
-
Paula Rodriguez-Otero, Niels W. C. J. van de Donk, Kodandaram Pillarisetti, Ingrid Cornax, Deeksha Vishwamitra, Kathleen Gray, Brandi Hilder, Jaszianne Tolbert, Thomas Renaud, Tara Masterson, Christoph Heuck, Colleen Kane, Raluca Verona, Philippe Moreau, Nizar Bahlis, and Ajai Chari
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2024
- Full Text
- View/download PDF
5. 689 Pharmacodynamic results with talquetamab and daratumumab in patients with relapsed/refractory multiple myeloma in TRIMM-2
- Author
-
Lien Vandenberk, Paula Rodríguez-Otero, Maria-Victoria Mateos, Ajai Chari, Bhagirathbhai Dholaria, Deeksha Vishwamitra, Sheri Skerget, M Damiette Smit, Kalpana Bakshi, Sangmin Lee, Nizar Bahlis, Niels WCJ van de Donk, and Raluca Verona
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
6. Profiling Chromatin Accessibility at Single-cell Resolution
- Author
-
Sarthak Sinha, Ansuman T. Satpathy, Weiqiang Zhou, Hongkai Ji, Jo A. Stratton, Arzina Jaffer, Nizar Bahlis, Sorana Morrissy, and Jeff A. Biernaskie
- Subjects
Single-cell ATAC-seq ,Gene regulation ,Epigenetics ,Single-cell multi-omics ,Cis-regulatory elements ,Biology (General) ,QH301-705.5 ,Computer applications to medicine. Medical informatics ,R858-859.7 - Abstract
How distinct transcriptional programs are enacted to generate cellular heterogeneity and plasticity, and enable complex fate decisions are important open questions. One key regulator is the cell’s epigenome state that drives distinct transcriptional programs by regulating chromatin accessibility. Genome-wide chromatin accessibility measurements can impart insights into regulatory sequences (in)accessible to DNA-binding proteins at a single-cell resolution. This review outlines molecular methods and bioinformatic tools for capturing cell-to-cell chromatin variation using single-cell assay for transposase-accessible chromatin using sequencing (scATAC-seq) in a scalable fashion. It also covers joint profiling of chromatin with transcriptome/proteome measurements, computational strategies to integrate multi-omic measurements, and predictive bioinformatic tools to infer chromatin accessibility from single-cell transcriptomic datasets. Methodological refinements that increase power for cell discovery through robust chromatin coverage and integrate measurements from multiple modalities will further expand our understanding of gene regulation during homeostasis and disease.
- Published
- 2021
- Full Text
- View/download PDF
7. Selinexor, daratumumab, and dexamethasone in patients with relapsed or refractory multiple myeloma
- Author
-
Cristina Gasparetto, Suzanne Lentzsch, Gary Schiller, Natalie Callander, Sascha Tuchman, Christine Chen, Darrell White, Rami Kotb, Heather Sutherland, Michael Sebag, Muhamed Baljevic, William Bensinger, Richard LeBlanc, Chris Venner, Nizar Bahlis, Adriana Rossi, Noa Biran, Heidi Sheehan, Jean‐Richard Saint‐Martin, Dane Van Domelen, Kazuharu Kai, Jatin Shah, Sharon Shacham, Michael Kauffman, and Brea Lipe
- Subjects
Selinexor ,Multiple Myeloma ,Daratumumab ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract We assessed the safety, efficacy, maximum tolerated dose (MTD), and the recommended phase 2 dose (RP2D) of selinexor, a first in class oral selective inhibitor of nuclear export (100 mg once weekly [QW] or 60 mg twice weekly), in combination with daratumumab (16 mg/kg per label) and dexamethasone (40 mg QW) (SDd) in patients with relapsed refractory multiple myeloma (RRMM). Thirty‐four patients (median prior therapies, 3 [range, 2‐10]) were enrolled; MM was refractory to proteasome inhibitor (PI) in 85%, immunomodulatory agent (IMiD) in 76%, both in 74%, and daratumumab in 6% of patients. Two dose‐limiting toxicities (DLTs) were reported in the selinexor 60 mg twice‐weekly cohort with no DLTs in the 100 mg QW cohort, making 100 mg QW the MTD and RP2D. Common treatment‐related adverse events included thrombocytopenia (70.6%), nausea (70.6%), fatigue (61.8%), anemia (61.8%), and neutropenia (50.0%). Overall response rate was 73% and median progression‐free survival 12.5 months in daratumumab‐naïve patients. SDd was well tolerated and its promising efficacy suggests that further study of this PI‐ and IMiD‐free regimen in RRMM patients who had at least one prior line of therapy including a PI and an IMiD but whose disease is naïve to daratumumab is warranted.
- Published
- 2021
- Full Text
- View/download PDF
8. CNV Radar: an improved method for somatic copy number alteration characterization in oncology
- Author
-
David Soong, Jeran Stratford, Herve Avet-Loiseau, Nizar Bahlis, Faith Davies, Angela Dispenzieri, A. Kate Sasser, Jordan M. Schecter, Ming Qi, Chad Brown, Wendell Jones, Jonathan J. Keats, Daniel Auclair, Christopher Chiu, Jason Powers, and Michael Schaffer
- Subjects
Copy number variation ,Next generation sequencing ,Multiple myeloma ,Acute myeloid leukemia ,Prostate cancer ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background Cancer associated copy number variation (CNV) events provide important information for identifying patient subgroups and suggesting treatment strategies. Technical and logistical issues, however, make it challenging to accurately detect abnormal copy number events in a cost-effective manner in clinical studies. Results Here we present CNV Radar, a software tool that utilizes next-generation sequencing read depth information and variant allele frequency patterns, to infer the true copy number status of genes and genomic regions from whole exome sequencing data. Evaluation of CNV Radar in a public multiple myeloma dataset demonstrated that CNV Radar was able to detect a variety of CNVs associated with risk of progression, and we observed > 70% concordance with fluorescence in situ hybridization (FISH) results. Compared to other CNV callers, CNV Radar showed high sensitivity and specificity. Similar results were observed when comparing CNV Radar calls to single nucleotide polymorphism array results from acute myeloid leukemia and prostate cancer datasets available on TCGA. More importantly, CNV Radar demonstrated its utility in the clinical trial setting: in POLLUX and CASTOR, two phase 3 studies in patients with relapsed or refractory multiple myeloma, we observed a high concordance rate with FISH for del17p, a risk defining CNV event (88% in POLLUX and 90% in CASTOR), therefore allowing for efficacy assessments in clinically relevant disease subgroups. Our case studies also showed that CNV Radar is capable of detecting abnormalities such as copy-neutral loss of heterozygosity that elude other approaches. Conclusions We demonstrated that CNV Radar is more sensitive than other CNV detection methods, accurately detects clinically important cytogenetic events, and allows for further interrogation of novel disease biology. Overall, CNV Radar exhibited high concordance with standard methods such as FISH, and its success in the POLLUX and CASTOR clinical trials demonstrated its potential utility for informing clinical and therapeutic decisions.
- Published
- 2020
- Full Text
- View/download PDF
9. Impact of COVID-19 on the Diagnosis and Management of Multiple Myeloma: Experience from a Canadian Center
- Author
-
Victor H. Jiménez-Zepeda, Patrick Yau, Douglas Stewart, Jowher Berhan, Carole Chambers, Holly Lee, Jason Tay, Peter Duggan, Sylvia McCulloch, Paola Neri, and Nizar Bahlis
- Subjects
Myeloma. SARS-CoV-2. COVID-19. ,Internal medicine ,RC31-1245 - Abstract
Background: The impact of coronavirus disease-19 on the management of multiple myeloma (MM) has been recognized. However, the real effect on clinical outcomes remains poorly understood. Objective: We describe a local experience of the management of MM patients and report their outcomes during the current pandemic. Methods: All consecutive symptomatic MM patients seen at our center since 03/20 were evaluated. Results: A cohort of 156 patients diagnosed from 01/19 to 12/20 was analyzed to interrogate differences in presentation patterns. A total of 553 MM patients were seen and/or treated at Tom Baker Cancer Center in the year of 2020. From those, 47.1% (n = 261) were tested for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Sixteen patients tested positive and data are presented. In addition, a decrease of 21.7% in the rate of new smoldering MM/MM diagnosis was observed in 2020 as compared to 2019. Further, an increase in deaths was also observed in 2020. Conclusions: Our study confirms an increase lethality for MM patients infected with SARS-CoV-2. A balance between safety and need for cancer control should be emphasized.
- Published
- 2022
- Full Text
- View/download PDF
10. Plain language summary of the MAIA study of daratumumab plus lenalidomide and dexamethasone for the treatment of people with newly diagnosed multiple myeloma
- Author
-
Thierry Facon, Shaji K Kumar, Torben Plesner, Robert Z Orlowski, Philippe Moreau, Nizar Bahlis, Supratik Basu, Hareth Nahi, Cyrille Hulin, Hang Quach, Hartmut Goldschmidt, Aurore Perrot, Katja Weisel, Noopur Raje, Margaret Macro, Laurent Frenzel, Xavier Leleu, Jianping Wang, Rian Van Rampelbergh, Clarissa M Uhlar, Jessica Vermeulen, Joana Duran, Fredrik Borgsten, and Saad Z Usmani
- Subjects
Cancer Research ,Oncology ,General Medicine - Abstract
What is this summary about? This is a summary of a clinical trial called MAIA. The trial tested 2 combinations of cancer drugs (daratumumab plus lenalidomide and dexamethasone compared with lenalidomide and dexamethasone) in people with newly diagnosed multiple myeloma. None of the participants who took part in the study had been treated before or were eligible to receive stem-cell transplants. How was the study in this summary conducted? A total of 737 participants took part. Half of the participants took daratumumab plus lenalidomide and dexamethasone, while the other half of the participants took only lenalidomide and dexamethasone. Once participants started taking the drugs, the cancer was monitored for improvement (response to treatment), worsening (disease progression), or no change. Participants' blood and urine were tested for myeloma protein to measure response to the treatment. Participants were also monitored for side effects. What were the results of the study? After approximately 56 months of follow-up, more participants who took daratumumab plus lenalidomide and dexamethasone were alive and had decreased myeloma protein levels (indicating improvement of cancer) than participants who took only lenalidomide and dexamethasone. The most common side effects were abnormally low white and red blood cell counts and increased lung infections. What do the results of the study mean? In the MAIA study, participants with multiple myeloma who took daratumumab plus lenalidomide and dexamethasone lived longer and had decreased myeloma protein levels than participants who took only lenalidomide and dexamethasone, indicating survival could be more likely with daratumumab added. Clinical Trial Registration: NCT02252172 (Phase 3 MAIA study)
- Published
- 2023
11. Supplementary Table 2 from Reovirus as a Viable Therapeutic Option for the Treatment of Multiple Myeloma
- Author
-
Don G. Morris, Adnan Mansoor, Douglas Stewart, Mark Pho, Paola Neri, Nizar Bahlis, He Gao, Karen Kopciuk, Joanne Luider, Zhong Qiao Shi, and Chandini M. Thirukkumaran
- Abstract
PDF file, 17K, Reovirus sensitivity and genetic lesions detected in multiple myeloma and MGUS patient tumor.
- Published
- 2023
12. Supplementary Table 1 from Reovirus as a Viable Therapeutic Option for the Treatment of Multiple Myeloma
- Author
-
Don G. Morris, Adnan Mansoor, Douglas Stewart, Mark Pho, Paola Neri, Nizar Bahlis, He Gao, Karen Kopciuk, Joanne Luider, Zhong Qiao Shi, and Chandini M. Thirukkumaran
- Abstract
PDF file, 21K, Reovirus sensitivity and prevalnet genetic lesions of myeloma cell lines used in the current study.
- Published
- 2023
13. Tumor Intrinsic Mechanisms of Antigen Escape to Anti-BCMA and Anti-GPRC5D Targeted Immunotherapies in Multiple Myeloma
- Author
-
Nizar Bahlis, Holly Lee, Sungwoo Ahn, Ranjan Maity, Noémie Leblay, Bachisio Ziccheddu, Monika Chojnacka, Anthony Cirrincione, Michael Durante, Elie Barakat, Remi Tilmont, Sarthak Sinha, Marietta Truger, Shari Kyman, Amrita Krishnan, Ola Landgren, Wencke Walter, Manja Meggendorfer, Claudia Haferlach, Torsten Haferlach, Hermann Einsele, K. Kortüm, Stefan Knop, Jean-Baptiste Alberge, Jonathan Keats, Leo Rasche, Francesco Maura, and Paola Neri
- Abstract
B cell maturation antigen (BCMA) target loss is considered to be a rare event that mediates multiple myeloma (MM) resistance to anti-BCMA chimeric antigen receptor T cell (CAR T) or bispecific T cell engager (TCE) therapies. Emerging data report that downregulation of G protein coupled receptor family C group 5 member D (GPRC5D) protein often occurs at relapse after anti-GPRC5D CAR T. To examine the tumor intrinsic factors that promote MM antigen escape, we performed combined bulk and single cell whole genome sequencing/ copy number variation analysis of 25 patients treated with anti-BCMA and/ or -GPRC5D CAR T/ TCE. In two cases, MM relapse post TCE/CAR T was driven by BCMA negative clones harboring focal biallelic deletions at the TNFRSF17 locus at relapse or by selective expansion of pre-existing subclones with biallelic TNFRSF17 loss. In another three cases of MM relapse, we identified non-truncating missense mutations or in-frame deletions in the extracellular domain of BCMA which negates the efficacies of anti-BCMA TCEs, despite detectable surface BCMA protein expression. With respect to GPRC5D, we report the first four cases of MM relapse with biallelic mutations of GPRC5D following anti-GPRC5D TCE, including two cases with convergent evolution where multiple subclones lost GPRC5D through different somatic events. Our data support that immunoselection of BCMA or GPRC5D negative or mutant clones post CAR T/ TCE therapies may be more prevalent than currently perceived. The engineering and selection of immunotherapies in MM should account for target antigen structural and extracellular domain mutations.
- Published
- 2023
14. Chimeric Antigen Receptor-T Cell Mediated Bilateral Facial Nerve Palsy: A Case Report
- Author
-
Natalya Patrick, Nizar Bahlis, and Steven Peters
- Subjects
Neurology (clinical) - Abstract
Chimeric antigen receptor (CAR-T) cell therapy is highly effective against hematological cancers but is associated with immune mediated side effects, including neurotoxicity. The most commonly described presentations of immune cell mediated neurotoxicity syndrome (ICANS) include cortical symptoms and generally localize to the central nervous system. In this report, we present a patient with acute onset of bilateral facial nerve palsy following CAR-T cell therapy, followed by a complete clinical recovery. Aside from a temporary anisocoria, he had no other neurologic symptoms and no encephalopathy or seizures. MRI Brain was non-contributory and cerebrospinal fluid revealed a modest increase in lymphocytes without systemic leukocytosis and viral studies were all negative. He was diagnosed with bilateral facial nerve palsy secondary to CAR-T cell therapy and subsequently treated with a course of steroids. Several weeks after presentation he returned to his neurological baseline. The presentation of CAR-T cell mediated facial nerve palsy is both clinically and scientifically relevant for physicians, patients, and researchers.
- Published
- 2023
15. Amyloidosis and COVID-19: experience from an amyloid program in Canada
- Author
-
Ellen Lewis, Nowell Fine, Robert J. H. Miller, Christopher Hahn, Sameer Chhibber, Etienne Mahe, Jason Tay, Peter Duggan, Sylvia McCulloch, Nizar Bahlis, Paola Neri, and Victor H. Jimenez-Zepeda
- Subjects
Amyloid ,Amyloid Neuropathies, Familial ,COVID-19 Testing ,SARS-CoV-2 ,COVID-19 ,Humans ,RNA, Viral ,Hematology ,General Medicine - Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV2) and associated COVID-19 infection continue to impact patients globally. Patients with underlying health conditions are at heightened risk of adverse outcomes from COVID-19; however, research involving patients with rare health conditions remains scarce. The amyloidoses are a rare grouping of protein deposition diseases. Light-chain and transthyretin amyloidosis are the most common disease forms, often present with systemic involvement of vital organs including the heart, nerves, kidneys, and GI tracts of affected individuals. The Amyloidosis Program of Calgary examined 152 ATTR patients and 103 AL patients analyzing rates of vaccination, COVID-19 testing, infection outcomes, influence referrals, and excess deaths. Results showed 15 total PCR-confirmed COVID-19 infections in the tested population of amyloid patients, with a higher frequency of infections among patient with AL compared to the ATTR cohort (26.2% vs 5.1%). Four patients (26.6%) required hospital admission for COVID-19 infection, 2 ATTR, and 2 AL patients. Of the confirmed cases, 1 (0.07%) unvaccinated ATTR patient died of a COVID-19 infection. An excess of deaths was found in both the ATTR and AL cohorts when comparing pre-pandemic years 2018 and 2019 to the pandemic years of 2020 and 2021. The finding suggests that amyloidosis patients are likely at a high risk for severe COVID-19 infection and mortality, especially those of advanced age, those on an active treatment with chemotherapy, and those with concomitant B-cell or plasma cell disorder. The impact of virtual healthcare visits and pandemic measures on the excess of deaths observed requires further research.
- Published
- 2022
16. Suboptimal response for AL amyloidosis: is it time for early switch? Experience from a single amyloid program
- Author
-
Robert Puckrin, Holly Lee, Jason Tay, Peter Duggan, Sylvia McCulloch, Paola Neri, Nizar Bahlis, and Victor H. Jimenez-Zepeda
- Subjects
Amyloid ,Internal Medicine ,Humans ,Amyloidogenic Proteins ,Immunoglobulin Light-chain Amyloidosis ,Amyloidosis - Published
- 2022
17. Teclistamab in Relapsed or Refractory Multiple Myeloma
- Author
-
Philippe Moreau, Alfred L. Garfall, Niels W.C.J. van de Donk, Hareth Nahi, Jesús F. San-Miguel, Albert Oriol, Ajay K. Nooka, Thomas Martin, Laura Rosinol, Ajai Chari, Lionel Karlin, Lotfi Benboubker, Maria-Victoria Mateos, Nizar Bahlis, Rakesh Popat, Britta Besemer, Joaquín Martínez-López, Surbhi Sidana, Michel Delforge, Lixia Pei, Danielle Trancucci, Raluca Verona, Suzette Girgis, Shun X.W. Lin, Yunsi Olyslager, Mindy Jaffe, Clarissa Uhlar, Tara Stephenson, Rian Van Rampelbergh, Arnob Banerjee, Jenna D. Goldberg, Rachel Kobos, Amrita Krishnan, Saad Z. Usmani, Hematology, and CCA - Cancer Treatment and quality of life
- Subjects
General Medicine - Abstract
BACKGROUND Teclistamab is a T-cell-redirecting bispecific antibody that targets both CD3 expressed on the surface of T cells and B-cell maturation antigen expressed on the surface of myeloma cells. In the phase 1 dose-defining portion of the study, teclistamab showed promising efficacy in patients with relapsed or refractory multiple myeloma. METHODS In this phase 1-2 study, we enrolled patients who had relapsed or refractory myeloma after at least three therapy lines, including triple-class exposure to an immunomodulatory drug, a proteasome inhibitor, and an anti-CD38 antibody. Patients received a weekly subcutaneous injection of teclistamab (at a dose of 1.5 mg per kilogram of body weight) after receiving step-up doses of 0.06 mg and 0.3 mg per kilogram. The primary end point was the overall response (partial response or better). RESULTS Among 165 patients who received teclistamab, 77.6% had triple-class refractory disease (median, five previous therapy lines). With a median follow-up of 14.1 months, the overall response rate was 63.0%, with 65 patients (39.4%) having a complete response or better. A total of 44 patients (26.7%) were found to have no minimal residual disease (MRD); the MRD-negativity rate among the patients with a complete response or better was 46%. The median duration of response was 18.4 months (95% confidence interval [CI], 14.9 to not estimable). The median duration of progression-free survival was 11.3 months (95% CI, 8.8 to 17.1). Common adverse events included cytokine release syndrome (in 72.1% of the patients; grade 3, 0.6%; no grade 4), neutropenia (in 70.9%; grade 3 or 4, 64.2%), anemia (in 52.1%; grade 3 or 4, 37.0%), and thrombocytopenia (in 40.0%; grade 3 or 4, 21.2%). Infections were frequent (in 76.4%; grade 3 or 4, 44.8%). Neurotoxic events occurred in 24 patients (14.5%), including immune effector cell-associated neurotoxicity syndrome in 5 patients (3.0%; all grade 1 or 2). CONCLUSIONS Teclistamab resulted in a high rate of deep and durable response in patients with triple-class-exposed relapsed or refractory multiple myeloma. Cytopenias and infections were common; toxic effects that were consistent with T-cell redirection were mostly grade 1 or 2.
- Published
- 2022
18. OAB-039: Treatment duration and long-term outcomes with daratumumab in transplant-ineligible newly diagnosed multiple myeloma from the phase 3 MAIA study
- Author
-
Philippe Moreau, Thierry Facon, Saad Usmani, Shaji Kumar, Torben Plesner, Hartmut Goldschmidt, Robert Orlowski, Aurore Perrot, Ajai Chari, Gordon Cook, Huiling Pei, Rian Van Rampelbergh, J Blake Bartlett, Clarissa Uhlar, Robin Carson, and Nizar Bahlis
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
19. P-232: Adverse event patterns and management with pomalidomide, dexamethasone, and daratumumab in patients with relapsed or refractory multiple myeloma: a safety analysis of the phase 2 MM-014 study
- Author
-
Nizar Bahlis, Gary Schiller, Christy J. Samaras, Michael Sebag, Jesus G. Berdeja, Siddhartha Ganguly, Jeffrey Matous, Kevin Song, Christopher Seet, Michael Bar, Donald Quick, Gustavo Fonseca, Donna E. Reece, Weiyuan Chung, Christian Gentili, Kim Lee, and David S. Siegel
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
20. OAB-002: The preexisting T cell landscape determines response to T cell-engagers therapy in multiple myeloma
- Author
-
Mirco Friedrich, Niklas Kehl, Paola Neri, Julius Michel, Noemi Leblay, Ranjan Maity, Michael Kilian, Holly Lee, Elie Barakat, Sungwoo Ahn, Simon Steiger, Roman Sankowski, Niels Weinhold, Karsten Rippe, Lukas Bunse, Michael Platten, Carsten Mülle-Tidow, Hartmut Goldschmidt, Marc S. Raab, and Nizar Bahlis
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
21. P-260: Daratumumab (DARA) plus bortezomib and dexamethasone (D-Vd) or lenalidomide and dexamethasone (D-Rd) in relapsed or refractory multiple myeloma (RRMM): subgroup analyses of CASTOR and POLLUX
- Author
-
María-Victoria Mateos, Paul Richardson, Katja Weisel, Philippe Moreau, Jesus San-Miguel, Hartmut Goldschmidt, Robert Orlowski, Pieter Sonneveld, Donna E. Reece, Kenshi Suzuki, Nizar Bahlis, Sung-Soo Yoon, Andrew Spencer, Ajay Nooka, Vania Hungria, Torben Plesner, Dina Ben Yehuda, Huiling Pei, Wendy Garvin Mayo, Xue Gai, Jodi Carey, Robin Carson, and Meletios A. Dimopoulos
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
22. OAB-005: Point mutations in BCMA extracellular domain mediate resistance to BCMA targeting immune therapies
- Author
-
Holly Lee, Ranjan Maity, Sungwoo Ahn, Noemi Leblay, Remi Tilmont, Elie Barakat, Paola Neri, and Nizar Bahlis
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
23. P-230: Preclinical and translational biomarker analysis to support further clinical development and dose optimization of mezigdomide (MEZI; CC-92480) in combination with either bortezomib or carfilzomib
- Author
-
Michael Amatangelo, Chad C. Bjorklund, Patrick Hagner, Phillip Koo, Tiziana Civardi, Alessandro Ghiddi, Jessica Katz, Paul Richardson, Nizar Bahlis, and Anita Gandhi
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
24. P-263: Matching-adjusted indirect treatment comparison (MAIC) of teclistamab vs approved therapies for the treatment of patients with triple-class exposed (TCE) relapsed/refractory multiple myeloma (RRMM)
- Author
-
Philippe Moreau, Saad Usmani, Niels W.C.J. van de Donk, Alfred Garfall, Michel Delforge, Albert Oriol, Ajay Nooka, Laura Rosiñol, Nizar Bahlis, Paula Rodríguez-Otero, Thomas Martin, Joris Diels, Suzy Van Sanden, Lixia Pei, Eric Ammann, Rachel Kobos, Alexander Marshall, Mary Slavcev, Jennifer Smit, Anil Londhe, and Amrita Krishnan
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
25. The 2020 BMT CTN Myeloma Intergroup Workshop on Immune Profiling and Minimal Residual Disease Testing in Multiple Myeloma
- Author
-
Sarah A. Holstein, Nizar Bahlis, P. Leif Bergsagel, Manisha Bhutani, Niccolo Bolli, Carrie Brownstein, Pierre Demolis, David Foureau, Francesca Gay, Irene M. Ghobrial, Nicole Gormley, Jens Hillengass, Martin Kaiser, Marcela V. Maus, J. Joseph Melenhorst, Maximilian Merz, Michael O. Dwyer, Bruno Paiva, Marcelo C. Pasquini, Nina Shah, Sandy W. Wong, Saad Z. Usmani, and Philip L. McCarthy
- Subjects
Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,Cellular therapy ,CAR T-cell ,Article ,Immune profiling ,Cell therapy ,Rare Diseases ,Bone Marrow ,Multiple myeloma ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Endpoint, CAR T-cell ,Minimal residual disease ,Cancer ,Transplantation ,Hematology ,business.industry ,Clinical study design ,High-Throughput Nucleotide Sequencing ,Cell Biology ,medicine.disease ,Endpoint ,Chimeric antigen receptor ,Clinical trial ,Good Health and Well Being ,Residual ,Neoplasm ,Molecular Medicine ,Diterpenes ,business - Abstract
The fifth annual Blood and Marrow Transplant Clinical Trials Network Myeloma Intergroup Workshop on Immune Profiling and Minimal Residual Disease Testing in Multiple Myeloma was conducted as one of the American Society of Hematology Annual Meeting Scientific Workshops on Thursday December 3, 2020. This workshop focused on four main topics: (1) integrating minimal residual disease into clinical trial design and practice; (2) the molecular and immunobiology of disease evolution and progression in myeloma; (3) adaptation of next-generation sequencing, next-generation flow cytometry, and cytometry by time of flight techniques; and (4) chimeric antigen receptor T-cell and other cellular therapies for myeloma. In this report, we provide a summary of the workshop presentations and discuss future directions in the field.
- Published
- 2021
26. Poster: MM-379: MagnetisMM-1: A Study of Elranatamab (PF-06863135), a B-Cell Maturation Antigen (BCMA)-Targeted, CD3-Engaging Bispecific Antibody, for Patients with Relapsed or Refractory Multiple Myeloma (MM)
- Author
-
Moshe Levy, Nizar Bahlis, Noopur Raje, Caitlin Costello, Bhagirathbhai Dholaria, Melhem Solh, Michael Tomasson, Harman Dube, Michael Damore, Hoi Kei Lon, Cynthia Basu, Athanasia Skoura, Edward Chan, Suzanne Trudel, Andrzej Jakubowiak, Michael Chu, Cristina Gasparetto, Andrew Dalovisio, Michael Sebag, and Alexander Lesokhin
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2021
27. A Randomized Phase II, Open Label, Study of Daratumumab, Weekly Low-Dose Oral Dexamethasone and Cyclophosphamide with or without Pomalidomide in Patients with Relapsed and Refractory Multiple Myeloma
- Author
-
Michael Sebag, Nizar Bahlis, Christopher P. Venner, Arleigh McCurdy, C. Tom Kouroukis, Jesse Shustik, Darrell J White, Rami Kotb, Julie Stakiw, Nicole B. Laferriere, Tony Reiman, Fernando Camacho, Julia Dobbin, Molei Fu, Engin Gul, and Donna E Reece
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Introduction As lenalidomide (Len) has become an integral part of therapy for newly diagnosed MM patients, most will have been either exposed or refractory to Len at the time of first or second relapse. The monoclonal antibody, Daratumumab, in combination with the more potent IMID pomalidomide (Pom) demonstrates good responses in patients previously exposed to lenalidomide. Low dose weekly cyclophosphamide has been shown to enhance the potency of pomalidomide in association with dexamethasone. In this clinical trial, we set out to compare the combination of daratumumab, weekly low dose cyclophosphamide, dexamethasone and pomalidomide (DCdP) to daratumumab, cyclophosphamide and dexamethasone (DCd) with pomalidomide added only at disease progression. Although we expected that a four-drug regiment would give superior clinical results, we hypothesized that a significant number of patients would not necessarily need all four drugs but could benefit from the addition of pomalidomide at treatment failure. Patients/Methods In this phase II clinical trial 120 patients with relapsed refractory myeloma, after at least one line of therapy, were randomized to receive either daratumumab (16mg/kg) weekly IV C1-2, every 2 weeks C3-6, monthly C7+, dexamethasone 40mg po weekly, cyclophosphamide 400mg po weekly and pomalidomide 4mg po days 1-21 of 28 day cycles (Arm A) or the same doses and dosing regimen of daratumumab, cyclophosphamide and dexamethasone but with pomalidomide added only after confirmed disease progression (Arm B). All patients had to be exposed to proteasome inhibitors and len prior to study entry. The primary endpoint of this study is the comparison of the PFS of Arm A and the PFS of Arm B after the addition Pom (PFS2) at 36 months while secondary endpoints included overall responses, duration of responses, survival and safety. Correlative laboratory studies are also planned. Results As of 1 April 2019 all 120 patients have been enrolled in 11 sites across Canada. The patient characteristics were: median age 65 (range 39-82); median 2 prior lines of therapy (range 1-8); 70% had a previous ASCT; 95% lenalidomide exposed; 93% proteasome inhibitor exposed; 90% lenalidomide and PI exposed; 25% carfilzomib exposed, len was the last line of therapy in 65%. Median follow-up was 8.2 months (range 1-15.6), median number of cycles 8 (range 1-17). The overall response rates (ORR) were 88.5% for arm A compared with 50.8% for arm B, with 57.4% and 25.4% of patients achieving ≥VGPR in arm A and B respectively. Among the 20 patients in Arm B that had progressed by data cutoff, the ORR after adding pomalidomide was 40% albeit with the short follow up time of 3.4 months. Although the median PFS of Arm A has not yet been reached, it was 10.9 mo. in Arm B prior to the addition of pom and 14.3 mo. from trial entry in the smaller group in whom pom was added after first progression (PFS2). In Arm A the 9- month PFS was 83%. Rates of grade 3/4 hematologic toxicities included a high incidence of neutropenia, 74% in Arm A and 30% in Arm B; however the rates of febrile neutropenia were low at 8.2% and 6.8% respectively. Grade 3/4 thrombocytopenia were 4.9% and 13.6%, respectively. The most common non-hematologic toxicity was pneumonia in 18% and 16.9% in arms A and B, respectively. Conclusions The results of this randomized phase II trial demonstrate that in a moderately pretreated MM population (median 2 lines of therapy but range 1-8) that the four-drug combination (DCdP) confers impressive response rates (ORR 88.5%) and a 9-month PFS of 83%. Although the three-drug combination (DCd) showed an inferior response rate of 50%, this is superior to Daratumumab used as a single agent in a similar patient population and so far at least 40% of patients who have progressed appear salvageable showing responses upon addition of Pom. Moreover, the addition of low dose cyclophosphamide, an alkylator with recognized immune properties, appears to enhance ORR and produce a durable PFS even when compared to Dara-pom-dex combinations used after two lines of therapy. Toxicities were principally hematologic and few resulted in treatment discontinuations. Disclosures Sebag: Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Bahlis:Amgen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria. Venner:Celgene: Research Funding; Amgen: Honoraria, Research Funding; Janssen: Honoraria; Takeda: Honoraria. McCurdy:Celgene: Honoraria; Janssen: Honoraria. Shustik:Takeda: Honoraria; Amgen: Honoraria; Janssen: Honoraria; Celgene: Honoraria. White:Sanofi: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Amgen: Consultancy, Honoraria. Kotb:Karyopharm: Equity Ownership; Amgen: Honoraria; Merck: Honoraria, Research Funding; Celgene: Honoraria; Janssen: Honoraria; Takeda: Honoraria. Stakiw:Roche: Research Funding; Lundbeck: Honoraria; BMS: Honoraria; Novartis: Honoraria, Speakers Bureau; Amgen: Honoraria, Speakers Bureau; Celgene: Honoraria, Speakers Bureau; Janssen: Honoraria, Research Funding, Speakers Bureau; Sanofi: Honoraria. Laferriere:Celgene: Honoraria; Taiho: Honoraria; Teva Pharm: Honoraria; ROCHE: Honoraria; Pfizer: Honoraria; Janssen: Honoraria; Takeda: Honoraria; Novartis: Honoraria; Astra Zeneca: Honoraria; Amgen: Honoraria. Camacho:Abbvie: Consultancy; Janssen: Consultancy; Baush-Health: Consultancy. Reece:Otsuka: Research Funding; BMS: Research Funding; Merck: Research Funding; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Karyopharm: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Consultancy, Honoraria, Research Funding.
- Published
- 2019
28. Four-Year Follow-up of the Phase 3 Pollux Study of Daratumumab Plus Lenalidomide and Dexamethasone (D-Rd) Versus Lenalidomide and Dexamethasone (Rd) Alone in Relapsed or Refractory Multiple Myeloma (RRMM)
- Author
-
Jonathan L. Kaufman, Saad Z. Usmani, Jesús San-Miguel, Nizar Bahlis, Darrell J White, Lotfi Benboubker, Gordon Cook, Merav Leiba, P Joy Ho, Kihyun Kim, Naoki Takezako, Philippe Moreau, Maria Krevvata, Huiling Pei, Jon Ukropec, Thomas Renaud, Sonali Trivedi, Rachel Kobos, and Meletios A. Dimopoulos
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Introduction: Daratumumab, a human IgGκ monoclonal antibody targeting CD38 with a direct on-tumor and immunomodulatory mechanism of action, is approved in combination with standard-of-care regimens for the treatment of newly diagnosed multiple myeloma (NDMM) and RRMM. When combined with standard of care regimens across four phase 3 studies, daratumumab demonstrated ≥44% reductions in the risk of progression or death, nearly doubled complete response (CR) or better rates, and tripled minimal residual disease (MRD)-negative rates at the 10-5 sensitivity threshold in pts with RRMM or NDMM (Palumbo A, et al. N Engl J Med 2016. 375[8]:754-766; Dimopoulos MA, et al. N Engl J Med 2016. 375[14]:1319-1331; Mateos MV, et al. N Engl J Med 2018. 378[6]:518-528; Facon T, et al. N Engl J Med 2019. 380[22]2104-2015). In the phase 3 POLLUX study (median follow-up 44.3 months), D-Rd reduced the risk of disease progression or death by 56% and significantly increased the overall response rate (ORR) versus Rd alone (93% vs 76%; P 4 years of median follow-up. Methods: Pts who received ≥1 prior line of therapy were randomized (1:1) to Rd (lenalidomide 25 mg PO on Days 1-21 of each 28-day cycle; dexamethasone 40 mg per week) ± daratumumab (16 mg/kg IV QW for Cycles 1-2, Q2W for Cycles 3-6, then Q4W until disease progression). Cytogenetic risk was determined by local fluorescence in situ hybridization or karyotyping; pts with high cytogenetic risk had t(4;14), t(14;16), and del17p abnormalities. PFS on subsequent line of therapy (PFS2), an exploratory endpoint, was defined as time from randomization to progression after next line of subsequent therapy or death. Results: A total of 569 pts were randomized (D-Rd, n = 286; Rd, n = 283). At a median follow-up of 51.3 months, D-Rd significantly prolonged progression-free survival (PFS) versus Rd (median 45.8 vs 17.5 months; hazard ratio [HR], 0.43; 95% confidence interval [CI], 0.35-0.54; P The median duration of treatment was 34.3 months in the D-Rd arm versus 16.0 months in the Rd arm. The most common (≥10%) grade 3/4 treatment-emergent adverse events (TEAEs) observed with D-Rd versus Rd included neutropenia (56% vs 42%), anemia (18% vs 22%), thrombocytopenia (15% vs 16%), pneumonia (16% vs 10%), and diarrhea (10% vs 4%). Similar rates of discontinuations due to TEAEs were observed for D-Rd versus Rd (16% vs 15%). The incidence of invasive second primary malignancies was 4.9% and 5.7% for the D-Rd and Rd groups, respectively. Additional efficacy data, including minimal residual disease, and safety analyses will be presented at the meeting. Conclusion: After >4 years of median follow-up, D-Rd continues to demonstrate a significant PFS benefit and higher rates of deeper responses versus Rd alone in pts with RRMM. Although significant PFS benefit was observed with D-Rd in RRMM pts regardless of prior lines of therapy or cytogenetic risk status, the greatest benefit was observed when used in patients treated earlier with D-Rd. The significant improvement in PFS2 suggests a potential survival benefit, but OS data is still immature. No new safety concerns were identified with this additional follow-up. Disclosures Kaufman: Celgene: Consultancy; Winship Cancer Institute of Emory University: Employment; Takeda: Consultancy; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria; AbbVie: Consultancy; Bristol-Myers Squibb: Consultancy; Incyte: Consultancy; Karyopharm: Membership on an entity's Board of Directors or advisory committees; TG Therapeutics: Consultancy; Amgen: Consultancy. Usmani:Amgen, Bristol-Myers Squibb, Celgene, Janssen, Merck, SkylineDX, Takeda: Other: Consultant/Advisor; Amgen, Celgene, Janssen, Sanofi, Takeda: Speakers Bureau; Amgen Array Biopharma, Bristol-Myers Squibb, Celgene, Janssen, Merck, Pharmacyclics, Sanofi, Takeda: Other: Research Grant. San-Miguel:Amgen, Bristol-Myers Squibb, Celgene, Janssen, MSD, Novartis, Roche, Sanofi, and Takeda: Consultancy, Honoraria. Bahlis:Celgene: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; Janssen: Consultancy, Honoraria. White:Janssen: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria. Cook:Amgen, Bristol-Myers Squib, GlycoMimetics, Seattle Genetics, Sanofi: Honoraria; Janssen, Takeda, Sanofi, Karyopharm, Celgene: Consultancy, Honoraria, Speakers Bureau; Celgene, Janssen-Cilag, Takeda: Honoraria, Research Funding. Ho:Novartis: Other: Trial Investigator meeting travel costs; La Jolla: Other: Trial Investigator meeting travel costs; Janssen: Other: Trial Investigator meeting travel costs; Celgene: Other: Trial Investigator meeting travel costs. Moreau:Takeda: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Amgen: Consultancy, Honoraria. Krevvata:Janssen: Employment. Pei:Janssen: Employment, Equity Ownership. Ukropec:Janssen: Employment, Equity Ownership. Renaud:Janssen: Employment, Equity Ownership. Trivedi:Janssen: Employment, Equity Ownership. Kobos:Janssen: Employment. Dimopoulos:Sanofi Oncology: Research Funding.
- Published
- 2019
29. Abstract 3015: IMiDs and BET inhibitors target distinct pathways of MYC dysregulation by super-enhancers in multiple myeloma
- Author
-
Daniel L. Riggs, Camille Herzog, Victoria M. Garbitt, Niamh Keane, Courtney J. Hillukka, Zachary J. Hammond, Julia E. Wiedmeier, Seth J. Welsh, Shulan Tian, Huihuang Yan, Ranjan Maity, Nizar Bahlis, Paola Neri, W Michael Kuehl, Marta Chesi, and P Leif Bergsagel
- Subjects
Cancer Research ,Oncology - Abstract
MYC dysregulation, the most common genetic aberration in multiple myeloma, is frequently due to the translocation of super-enhancers to the MYC locus. Several drugs target proteins that are enriched at many of these enhancers including BRD4 (BET inhibitors, BETi) and Ikaros (IMiDs), although their mechanism of action remains poorly understood. Here we present a characterization of the responses to these drugs in a collection of over sixty myeloma cell lines having a diversity of MYC rearrangements. We found that the anti-proliferative effects of these drugs significantly correlated with changes in MYC protein levels, consistent with both drugs targeting MYC expression. Despite this common target, there was no statistically significant correlation between the individual BETi and IMiD responses, suggesting that they act through different mechanisms. Of those lines having extremes of sensitivity or resistance, there were two major groups (BETiS/IMiDS and BETiS/IMiDR), a smaller group of four lines resistant to both drugs individually (BETiR/IMiDR) and only one line was BETiR/IMiDS. In the BETiR/IMiDR group, resistance to BETi was mediated by a BRD4-independent mechanism as BRD4 was efficiently released from the MYC-associated enhancers. In all three of BETiR/IMiDR cell lines that we examined, treatment with BETi and IMiD together abolished proliferation and down-regulated MYC, consistent with parallel BRD4- and Ikaros-dependent pathways driving MYC expression. These resistant lines all expressed high levels of the transcription factor ETV4 and knocking out its gene sensitized a line to each drug individually. Thus ETV4 appears to be necessary for the parallel pathways driving MYC expression. There were nine lines in the BETiS/IMiDR group. Sensitivity to BETi in these lines could be explained by either low ETV4 expression or by BETi repressing Ikaros levels (which was only observed in BETiS lines, suggesting that BRD4 drives IKZF1 expression in these lines). Thus, in ETV4-containing lines, BETi sensitivity is due to the simultaneously targeting of the BRD4- and Ikaros-dependent pathways. IMiD resistance likely was due to several reasons. In one cell line, OCIMY5, IMiD had little effect on Ikaros levels, likely due to the previously reported low levels of Cereblon. Seven of the eight remaining lines expressed high levels of either ETV4, or the other potential super-enhancer binding factors IRF4 or RUNX1. In the eight BETiS/IMiDS cell lines, IMiD strongly reduced both Ikaros and Aiolos protein levels, which likely caused IMiD sensitivity. As with the BETiS lines described above, the lines in this group either lacked ETV4 or BETi repressed Ikaros levels. In conclusion, by examining drug response in a collection of genetically annotated myeloma cell lines we have been able to identify factors that contribute the broad range of responses to BETi and IMiDs in myeloma cells. Citation Format: Daniel L. Riggs, Camille Herzog, Victoria M. Garbitt, Niamh Keane, Courtney J. Hillukka, Zachary J. Hammond, Julia E. Wiedmeier, Seth J. Welsh, Shulan Tian, Huihuang Yan, Ranjan Maity, Nizar Bahlis, Paola Neri, W Michael Kuehl, Marta Chesi, P Leif Bergsagel. IMiDs and BET inhibitors target distinct pathways of MYC dysregulation by super-enhancers in multiple myeloma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3015.
- Published
- 2019
30. Expression, adverse prognostic significance and therapeutic small molecule inhibition of Polo-like kinase 1 in multiple myeloma
- Author
-
Raymond Lai, Robert P. Evans, Greg Dueck, Inka Toman, Alexander C. Klimowicz, Tony Reiman, Sunita Ghosh, Nizar Bahlis, Andrew R. Belch, Joyce Fung, Jonathan M. Loree, Linda M. Pilarski, Roger Sidhu, and Michelle Jung
- Subjects
Adult ,Male ,Cancer Research ,Myeloma protein ,Cell ,Antineoplastic Agents ,Cell Cycle Proteins ,Mice, SCID ,Polo-like kinase ,Nod ,Protein Serine-Threonine Kinases ,Biology ,PLK1 ,Small Molecule Libraries ,Mice ,Mice, Inbred NOD ,immune system diseases ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Multiple myeloma ,Aged ,Retrospective Studies ,Aged, 80 and over ,Regulation of gene expression ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,Immunology ,Cancer research ,Female ,Bone marrow ,Multiple Myeloma - Abstract
The amplified myeloma centrosome has been identified as a therapeutic target. The present study explored the expression and prognostic significance of the centrosome-associated protein PLK1 in myeloma and the effect of BI 2536, a potent and selective inhibitor of PLK1, on myeloma cells. High plasma cell expression of PLK1 protein in myeloma patient bone marrow biopsies is an independent adverse prognostic factor (HR=2.3, p=0.003 unadjusted; HR=1.9, p=0.03 in multivariable model). BI 2536 inhibits myeloma cell lines at nanomolar concentrations, and is therapeutic for xenografts in NOD/SCID mice. PLK1 inhibition is a potential new strategy for the treatment of multiple myeloma.
- Published
- 2011
31. A Vaccination Technique to Combat Presently Untreatable Chronic Ailments
- Author
-
Arpad Barabas, Chad Cole, Nizar Bahlis, and Rene Lafreniere
- Subjects
Vaccination ,medicine.medical_specialty ,business.industry ,General Earth and Planetary Sciences ,Medicine ,business ,Intensive care medicine ,General Environmental Science - Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.