1
|
Fotiou D, Badelita SN, Katodritou E, Beksac M, Bila J, Spanoudakis E, Batinić J, Coriu D, Barbu S, Danaila C, Dalampira D, Sevastoudi A, Seval GC, Toprak SK, Sretenovic A, Markovic O, Valkovic T, Cvetkovic Z, Theodorakakou F, Gavriatopoulou M, Terpos E, Dimopoulos MA, Kastritis E. Disease Characteristics and Treatment Outcomes of Myeloma Patients Under 50 Years of Age: An Analysis of the Balkan Myeloma Study Group. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e310-e318. [PMID: 39890518 DOI: 10.1016/j.clml.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND Multiple myeloma (MM) is predominantly a disease of the elderly, but approximately 10% of patients are younger than 50 years at diagnosis. METHODS: This study aimed to investigate the clinical characteristics, treatment outcomes, and prognostic factors in younger MM patients using retrospective data from the Balkan Myeloma Study Group registry. RESULTS: A total of 350 patients under 50 years old were included, comprising 10.4% of the overall cohort. The study found that younger patients had lower rates of renal impairment and anemia but a higher incidence of lytic bone disease and adverse cytogenetics. Treatment regimens, including proteasome inhibitors and immunomodulatory agents, were comparable between younger and older patients, but younger patients had significantly better complete response rates and overall survival (OS). The 5- and 10-year OS rates were 76% and 64%, respectively, with a projected median OS exceeding 15 years. Factors such as anemia, hypercalcemia, and high-risk cytogenetics were associated with worse survival outcomes. Autologous stem cell transplantation (ASCT) emerged as a key contributor to improved progression-free survival (PFS) and OS. CONCLUSION In conclusion, younger MM patients exhibit distinct disease features and benefit from intensified treatment approaches, underscoring the need for tailored therapies to achieve potential disease cure.
Collapse
Affiliation(s)
- Despina Fotiou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Eirini Katodritou
- Department of Hematology, Theagenio Cancer Hospital, Thessaloniki, Greece
| | - Meral Beksac
- Ankara Liv Hospital, Istinye University, Ankara, Turkey
| | - Jelena Bila
- Clinic of Hematology, University Clinical Center of Serbia, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Emmanouil Spanoudakis
- Department of Hematology, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Josip Batinić
- Department of internal medicine, Division of Hematology, University Hospital Centre Zagreb; Zagreb, Croatia
| | - Daniel Coriu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Sinziana Barbu
- Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania
| | - Catalin Danaila
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Dimitra Dalampira
- Department of Hematology, Theagenio Cancer Hospital, Thessaloniki, Greece
| | | | | | - Selami Koçak Toprak
- Department of Hematology, Ankara University School of Medicine, Ankara, Turkey
| | - Aleksandra Sretenovic
- Clinic of Hematology, University Clinical Center of Serbia, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Olivera Markovic
- Clinic of Hematology, Clinical Hospital Center "Bezanijska Kosa", Medical Faculty, University of Belgrade
| | - Toni Valkovic
- Special Hospital Medico, Rijeka, Croatia; Faculty of Health Studies, University of Rijeka, Croatia
| | - Zorica Cvetkovic
- Department of Hematology Clinical Hospital Center Zemun, Medical Faculty University of Belgrade, Belgrade, Serbia
| | - Fenia Theodorakakou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
2
|
Fotiou D, Dimopoulos MA. Dr. Raymond Alexanian: Pioneering Contributions to Multiple Myeloma Research, Treatment, and the Concept of Curability. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:156-161. [PMID: 39424475 DOI: 10.1016/j.clml.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Multiple myeloma is a challenging hematological malignancy, with ongoing efforts toward finding a cure. Dr. Raymond Alexanian has been instrumental in advancing the understanding and treatment of multiple myeloma through his pioneering research. Trained at Dartmouth College and Harvard Medical School, Dr. Alexanian MD Anderson Cancer Center career spanned nearly 5 decades. He developed the highly effective MP (melphalan-prednisone) regimen, which became a standard treatment for years. Dr. Alexanian's exploration of steroids, particularly high-dose dexamethasone, and the collaboration with Dr. Bart Barlogie led to the development of the VAD (vincristine, doxorubicin, and dexamethasone) regimen, significantly improving outcomes for refractory cases. He also contributed to the establishment of high-dose melphalan with autologous stem cell transplantation. Dr. Alexanian's work identified critical prognostic factors and contributed understanding indolent and localized myeloma. His efforts in evaluating new agents, including thalidomide and bortezomib, further enhanced treatment options. Beyond research, his compassionate patient care and advocacy have had a profound impact. Dr. Alexanian's legacy continues to inspire advancements in multiple myeloma treatment, with his innovative approaches reshaping the field and fostering the pursuit of a cure.
Collapse
Affiliation(s)
- Despina Fotiou
- Department of Clinical Therapeutics, University of Athens, School of Medicine, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, University of Athens, School of Medicine, Athens, Greece.
| |
Collapse
|
3
|
Dang M, Wang R, Lee HC, Patel KK, Becnel MR, Han G, Thomas SK, Hao D, Chu Y, Weber DM, Lin P, Lutter-Berka Z, Berrios Nolasco DA, Huang M, Bansal H, Song X, Zhang J, Futreal A, Moreno Rueda LY, Symer DE, Green MR, Rojas Hernandez CM, Kroll M, Afshar-Khargan V, Ndacayisaba LJ, Kuhn P, Neelapu SS, Orlowski RZ, Wang L, Manasanch EE. Single cell clonotypic and transcriptional evolution of multiple myeloma precursor disease. Cancer Cell 2023; 41:1032-1047.e4. [PMID: 37311413 PMCID: PMC10317474 DOI: 10.1016/j.ccell.2023.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/02/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023]
Abstract
Multiple myeloma remains an incurable disease, and the cellular and molecular evolution from precursor conditions, including monoclonal gammopathy of undetermined significance and smoldering multiple myeloma, is incompletely understood. Here, we combine single-cell RNA and B cell receptor sequencing from fifty-two patients with myeloma precursors in comparison with myeloma and normal donors. Our comprehensive analysis reveals early genomic drivers of malignant transformation, distinct transcriptional features, and divergent clonal expansion in hyperdiploid versus non-hyperdiploid samples. Additionally, we observe intra-patient heterogeneity with potential therapeutic implications and identify distinct patterns of evolution from myeloma precursor disease to myeloma. We also demonstrate distinctive characteristics of the microenvironment associated with specific genomic changes in myeloma cells. These findings add to our knowledge about myeloma precursor disease progression, providing valuable insights into patient risk stratification, biomarker discovery, and possible clinical applications.
Collapse
Affiliation(s)
- Minghao Dang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ruiping Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hans C Lee
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Krina K Patel
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melody R Becnel
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guangchun Han
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheeba K Thomas
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dapeng Hao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yanshuo Chu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Donna M Weber
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pei Lin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zuzana Lutter-Berka
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David A Berrios Nolasco
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mei Huang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hima Bansal
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xingzhi Song
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luz Yurany Moreno Rueda
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David E Symer
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael R Green
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristhiam M Rojas Hernandez
- Department of Internal Medicine, Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Kroll
- Department of Internal Medicine, Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vahid Afshar-Khargan
- Department of Internal Medicine, Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Peter Kuhn
- University of Southern California, Los Angeles, CA, USA
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Elisabet E Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Afrough A, Alsfeld LC, Milton DR, Delgado R, Popat UR, Nieto Y, Kebriaei P, Oran B, Saini N, Srour S, Hosing C, Cheema FH, Ahmed S, Manasanch EE, Lee HC, Kaufman GP, Patel KK, Weber DM, Orlowski RZ, Pinnix CC, Dabaja BS, Thomas SK, Champlin RE, Shpall EJ, Qazilbash MH, Bashir Q. Long-Term Outcomes of Allogeneic Hematopoietic Cell Transplantation in Patients with Newly Diagnosed Multiple Myeloma. Transplant Cell Ther 2023; 29:264.e1-264.e9. [PMID: 35605883 DOI: 10.1016/j.jtct.2022.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023]
Abstract
Despite remarkable progress in survival with the availability of novel agents, an overwhelming majority of patients with multiple myeloma (MM) have disease that relapses. Allogeneic (allo-) hematopoietic cell transplantation (HCT) is a potentially curative option for a subgroup of patients with high-risk MM. This study assessed the long-term outcome of MM patients who underwent allo-HCT while in first remission as consolidation treatment. Thirty-three patients with newly diagnosed MM who underwent allo-HCT as part of consolidation therapy between 1994 and 2016 were reviewed retrospectively. Of these patients, 70% underwent autologous HCT before allo-HCT. All patients were chemosensitive and achieved at least partial response before proceeding to allo-HCT. Most received nonmyeloablative/reduced-intensity conditioning (88%) and a matched sibling donor graft (85%). Acute graft-versus-host disease (GVHD) and chronic GVHD occurred in 30% and 61% of patients, respectively. The median duration of follow-up was 64.1 months (range, 1.4 to 199.2 months) for all patients and 164.4 months (range, 56.0 to 199.2 months) for survivors. The median progression-free survival (PFS) was 36 months (95% confidence interval (CI), 8.6 to 73.0 months). The median time from treatment to progression was 73.0 months (95% CI, 30.6 months to not reached). The median overall survival (OS) was 131.9 months (95% CI, 38.4 months to not reached). Of all patients, 39% were alive for more than 10 years, with 46% (n = 6) without progression or relapse. The cumulative incidence of relapse was 18% at 1 year, 39% at 5 years, and 46% at 10 years post-allo-HCT. The cumulative incidence of nonrelapse mortality was 3% at 100 days, 18% at 1 year, 21% at 3 years, and 24% at 5 year post-allo-HCT. On multivariable analysis, high-risk cytogenetics were associated with a shorter PFS (hazard ratio [HR], 2.7; 95% CI, 1.01 to 7.21; P = .047) and OS (HR, 4.91; 95% CI, 1.48 to 16.27; P = .009). Achieving complete remission after allo-HCT also was associated with longer PFS (HR, 0.24; 95% CI, 0.09 to 0.64; P = .004) and OS (HR, .23; 95% CI, .07 to .72; P = .012). Allo-HCT may confer a survival advantage in a selected population of MM patients when performed early in the disease course; additional data on identifying the patients who will benefit the most are needed.
Collapse
Affiliation(s)
- Aimaz Afrough
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Leonard C Alsfeld
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Denái R Milton
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ruby Delgado
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Uday R Popat
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yago Nieto
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Betul Oran
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeraj Saini
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samer Srour
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chitra Hosing
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Faisal H Cheema
- The University of Houston College of Medicine, Houston, Texas
| | - Sairah Ahmed
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elisabet E Manasanch
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hans C Lee
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory P Kaufman
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Krina K Patel
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Donna M Weber
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chelsea C Pinnix
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bouthaina S Dabaja
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sheeba K Thomas
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qaiser Bashir
- Department of Stem Cell Transplantation & Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
5
|
Gordan LN, Marks SM, Xue M, Nagovski N, Lambert JH, Smith RE. Daratumumab utilization and cost analysis among patients with multiple myeloma in a US community oncology setting. Future Oncol 2021; 18:301-309. [PMID: 34709061 DOI: 10.2217/fon-2021-1072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: The introduction of daratumumab into the treatment of multiple myeloma has improved outcomes in patients; however, community oncologists often dose more frequently than the US FDA-approved label. Materials and methods: Integra analyzed its database to elucidate daratumumab treatment patterns and the impact of increased utilization on the cost of care for multiple myeloma. Results: Following week 24, 671 (65%) of 1037 patients remained on daratumumab-containing regimens, with 330 patients continuing more frequent treatments than the expected once-every-4-weeks dosing described in the standard dosing schedule. Patients received an average of 14% more daratumumab doses than the FDA-approved label indicates, increasing the 1-year daratumumab costs by an estimated US$31,353. Conclusion: Daratumumab is utilized more frequently than the FDA-recommended dosing, leading to higher multiple myeloma treatment costs.
Collapse
Affiliation(s)
| | - Stanley M Marks
- University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Mei Xue
- Integra PrecisionQ, West Palm Beach, FL, USA
| | | | | | | |
Collapse
|
6
|
Long-term outcomes after autologous stem cell transplantation for multiple myeloma. Blood Adv 2020; 4:422-431. [PMID: 31990333 DOI: 10.1182/bloodadvances.2019000524] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/01/2019] [Indexed: 01/07/2023] Open
Abstract
As multiple myeloma (MM) treatments evolve, frequent updates are required to monitor the long-term effect of changes in approach. Traditionally, MM is considered an incurable disease, with most patients eventually relapsing. However, improvements in treatments has raised the possibility that MM might be functionally curable. To examine improvements in long-term survival, we followed 4329 patients with newly diagnosed MM treated with autologous stem cell transplantation (ASCT) at the University of Arkansas for Medical Sciences from 1989 through 2018. Overall survival (OS) and progression-free survival (PFS) were evaluated using Kaplan-Meier analysis, Cox proportional hazards models, relative survival analysis, and cure modeling among different time periods, risk groups, and demographic traits. Steady improvements in OS were found, with patients treated in 2014 or later having superior OS (hazard ratio, 0.35; 95% confidence interval [CI], 0.27-0.45) and reduced excess risk for MM death (relative excess risk, 0.30; 95% CI, 0.22-0.41) compared with patients treated in 1997 or earlier. Patients treated during intervening time periods often had intermediate survival, but trends in OS, PFS, and landmarked analyses were inconsistent. Cure models support the potential for cure, ranging from 6.3% to 31.3%, depending on the year of treatment, with 10.0% to 18.6% of patients achieving their normal life expectancy across multiple periods. There was some evidence of reductions in early mortality within 3 years of diagnosis, longer complete response (CR) duration, and reductions in relapse after achieving CR. However, results differed depending on age, risk group, and cytogenetic characteristics.
Collapse
|
7
|
Bortezomib, lenalidomide, and dexamethasone with panobinostat for front-line treatment of patients with multiple myeloma who are eligible for transplantation: a phase 1 trial. LANCET HAEMATOLOGY 2018; 5:e628-e640. [DOI: 10.1016/s2352-3026(18)30174-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 11/24/2022]
|
8
|
Wang M, Law ME, Castellano RK, Law BK. The unfolded protein response as a target for anticancer therapeutics. Crit Rev Oncol Hematol 2018; 127:66-79. [DOI: 10.1016/j.critrevonc.2018.05.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/22/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022] Open
|
9
|
Danhof S, Hudecek M, Smith EL. CARs and other T cell therapies for MM: The clinical experience. Best Pract Res Clin Haematol 2018; 31:147-157. [PMID: 29909915 DOI: 10.1016/j.beha.2018.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/12/2018] [Accepted: 03/02/2018] [Indexed: 12/15/2022]
Abstract
Harnessing the endogenous immune system to eliminate malignant cells has long been an intriguing approach. After considerable success in the treatment of B-cell acute lymphoblastic leukemia, chimeric antigen receptor (CAR)-modified T cells have entered early clinical evaluation in the field of multiple myeloma (MM). The choice of suitable non-CD19 target antigens is challenging and a variety of myeloma-associated surface molecules have been under preclinical investigation. Most recent clinical protocols have focused on targeting B-cell maturation antigen (BCMA), and early results are promising. The trials differ in receptor constructs, patient selection, dosing strategies and conditioning chemotherapy and will thus pave the way to eventually define the optimal parameters. Other sources for autologous T-cell therapy of MM include affinity-enhanced T-cell receptor-modified cells and marrow infiltrating lymphocytes. In summary, adoptive T-cell transfer for the treatment of MM is still in its infancy, but if early response rates indicate durability, will be a paradigm changing therapeutic modality for the treatment of MM.
Collapse
Affiliation(s)
- Sophia Danhof
- Department of Hematology and Medical Oncology, University Hospital Wuerzburg, Versbacherstrasse 5, 97078 Wuerzburg, Germany.
| | - Michael Hudecek
- Department of Hematology and Medical Oncology, University Hospital Wuerzburg, Versbacherstrasse 5, 97078 Wuerzburg, Germany.
| | - Eric L Smith
- Myeloma Service, Cellular Therapeutics Center, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, NY 10065, New York, USA.
| |
Collapse
|
10
|
Ravi P, Kumar SK, Cerhan JR, Maurer MJ, Dingli D, Ansell SM, Rajkumar SV. Defining cure in multiple myeloma: a comparative study of outcomes of young individuals with myeloma and curable hematologic malignancies. Blood Cancer J 2018. [PMID: 29531285 PMCID: PMC5849889 DOI: 10.1038/s41408-018-0065-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Advances in therapy in recent years have led investigators to challenge the dogma that multiple myeloma (MM) is incurable. We assessed overall (OS) and progression-free survival (PFS) of young patients ( ≤ 50 years) with MM and compared outcomes with follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), and Hodgkin lymphoma (HL). All patients ≤ 50 years with newly diagnosed MM (n = 212), FL (n = 168), DLBCL (n = 195), and HL (n = 233) between 1 January 2005 and 31 December 2015 were included. Observed vs. expected survival was summarized by standardized mortality ratios (SMR). Compared to the background US population, excess mortality risk was seen at diagnosis in all four cancers, SMR 19.5 (15.2–24.5) in MM, 4.2 (2.3–7.2) in FL, 13.0 (9.2–18.4) in DLBCL, and 5.2 (2.6–9.3) in HL. We reasoned that cure would most likely occur in the first 3 years after diagnosis and be reflected by an overall survival probability similar to the background population. From the 36-month landmark, excess mortality risk was seen in MM (SMR 20.7 [14.7–28.3]) and FL (SMR 3.8 [1.5–7.8]), but not with DLBCL (SMR 3.1 [0.8–8.0]) or HL (SMR 0.9 [0.0–5.1]). MM patients have 20-fold excess mortality risk compared to the background population at diagnosis and at 3 years after diagnosis, suggesting that MM remains an incurable cancer.
Collapse
Affiliation(s)
- Praful Ravi
- Department of Internal Medicine,, Mayo Clinic, Rochester, MN, USA
| | - Shaji K Kumar
- Division of Hematology,, Mayo Clinic, Rochester, MN, USA
| | - James R Cerhan
- Division of Epidemiology, Department of Health Sciences Research,, Mayo Clinic, Rochester, MN, USA
| | | | - David Dingli
- Division of Hematology,, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
11
|
Ghosh A, Mailankody S, Giralt SA, Landgren CO, Smith EL, Brentjens RJ. CAR T cell therapy for multiple myeloma: where are we now and where are we headed? Leuk Lymphoma 2017; 59:2056-2067. [PMID: 29105517 DOI: 10.1080/10428194.2017.1393668] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
While recent progress has been made in the management of multiple myeloma, it remains a highly fatal malignancy especially among patients with relapsed-refractory disease. Immunotherapy with adoptive T cells targeting myeloma-associated antigens are at various stages of development and have brought about a new hope for cure. This is a review on the emerging field of adoptively transferred engineered T cell based approaches, with an in-depth focus on chimeric antigen receptors (CAR) targeting multiple myeloma. The recent results from CAR T cells targeting B cell maturation antigen are encouraging but eventual resistance to the CAR T cell therapies remain problematic. With newer approaches in therapies for multiple myeloma, the role of transplantation is evolved to form a platform for T cell therapies.
Collapse
Affiliation(s)
- Arnab Ghosh
- a Hematology/Oncology/BMT Fellowship Program, Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Sham Mailankody
- b Myeloma Service, Division of Hematologic Oncology, Department of Medicine , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Sergio A Giralt
- c Adult BMT Service, Memorial Sloan Kettering Cancer Center , New York , NY , USA.,d Cellular Therapeutics Center, Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - C Ola Landgren
- b Myeloma Service, Division of Hematologic Oncology, Department of Medicine , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Eric L Smith
- b Myeloma Service, Division of Hematologic Oncology, Department of Medicine , Memorial Sloan Kettering Cancer Center , New York , NY , USA.,d Cellular Therapeutics Center, Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Renier J Brentjens
- d Cellular Therapeutics Center, Memorial Sloan Kettering Cancer Center , New York , NY , USA.,e Leukemia Service, Department of Medicine , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| |
Collapse
|
12
|
Ríos-Tamayo R, Martín-García A, Alarcón-Payer C, Sánchez-Rodríguez D, de la Guardia AMDVD, García Collado CG, Jiménez Morales A, Jurado Chacón M, Cabeza Barrera J. Pomalidomide in the treatment of multiple myeloma: design, development and place in therapy. Drug Des Devel Ther 2017; 11:2399-2408. [PMID: 28860711 PMCID: PMC5574598 DOI: 10.2147/dddt.s115456] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Multiple myeloma is a very heterogeneous disease with variable survival. Despite recent progress and the widespread use of new agents, patients with relapsed and refractory disease have a poor outcome. Immunomodulatory drugs play a key role in both the front-line and the relapsed/refractory setting. The combination of pomalidomide (POM) and dexamethasone is safe and effective in relapsed and refractory patients, even in those with high-risk cytogenetic features. Furthermore, it can be used in most patients without the need to adjust according to the degree of renal failure. In order to further improve the results, POM-based triplet therapies are currently used. This article highlights the most relevant issues of POM and POM-based combinations in the relapsed/refractory multiple myeloma setting, from a pharmacological and clinical point of view.
Collapse
Affiliation(s)
- Rafael Ríos-Tamayo
- Monoclonal Gammopathies Unit
- Department of Hematology, University Hospital Virgen de las Nieves, Granada, Spain
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| | - Agustín Martín-García
- Department of Pharmacy
- Clinical Trials Unit, University Hospital Virgen de las Nieves, Granada, Spain
| | | | | | | | | | | | - Manuel Jurado Chacón
- Monoclonal Gammopathies Unit
- Department of Hematology, University Hospital Virgen de las Nieves, Granada, Spain
- Genomic Oncology Area, GENYO, Center for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| | - José Cabeza Barrera
- Instituto de Investigación Biosanitaria de Granada (Ibs.GRANADA), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- Department of Pharmacy
| |
Collapse
|
13
|
Lee HC, Wang H, Baladandayuthapani V, Lin H, He J, Jones RJ, Kuiatse I, Gu D, Wang Z, Ma W, Lim J, O'Brien S, Keats J, Yang J, Davis RE, Orlowski RZ. RNA Polymerase I Inhibition with CX-5461 as a Novel Therapeutic Strategy to Target MYC in Multiple Myeloma. Br J Haematol 2017; 177:80-94. [PMID: 28369725 DOI: 10.1111/bjh.14525] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Dysregulation of MYC is frequently implicated in both early and late myeloma progression events, yet its therapeutic targeting has remained a challenge. Among key MYC downstream targets is ribosomal biogenesis, enabling increases in protein translational capacity necessary to support the growth and self-renewal programmes of malignant cells. We therefore explored the selective targeting of ribosomal biogenesis with the small molecule RNA polymerase (pol) I inhibitor CX-5461 in myeloma. CX-5461 induced significant growth inhibition in wild-type (WT) and mutant TP53 myeloma cell lines and primary samples, in association with increases in downstream markers of apoptosis. Moreover, Pol I inhibition overcame adhesion-mediated drug resistance and resistance to conventional and novel agents. To probe the TP53-independent mechanisms of CX-5461, gene expression profiling was performed on isogenic TP53 WT and knockout cell lines and revealed reduction of MYC downstream targets. Mechanistic studies confirmed that CX-5461 rapidly suppressed both MYC protein and MYC mRNA levels. The latter was associated with an increased binding of the RNA-induced silencing complex (RISC) subunits TARBP2 and AGO2, the ribosomal protein RPL5, and MYC mRNA, resulting in increased MYC transcript degradation. Collectively, these studies provide a rationale for the clinical translation of CX-5461 as a novel therapeutic approach to target MYC in myeloma.
Collapse
Affiliation(s)
- Hans C Lee
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hua Wang
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Heather Lin
- The Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jin He
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard J Jones
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Isere Kuiatse
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dongmin Gu
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhiqiang Wang
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wencai Ma
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Lim
- Senhwa Biosciences, Inc., San Diego, CA, USA
| | | | - Jonathan Keats
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Jing Yang
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Davis
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Z Orlowski
- The Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
Krishnan SR, Jaiswal R, Brown RD, Luk F, Bebawy M. Multiple myeloma and persistence of drug resistance in the age of novel drugs (Review). Int J Oncol 2016; 49:33-50. [PMID: 27175906 DOI: 10.3892/ijo.2016.3516] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/17/2015] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is a mature B cell neoplasm that results in multi-organ failure. The median age of onset, diverse clinical manifestations, heterogeneous survival rate, clonal evolution, intrinsic and acquired drug resistance have impact on the therapeutic management of the disease. Specifically, the emergence of multidrug resistance (MDR) during the course of treatment contributes significantly to treatment failure. The introduction of the immunomodulatory agents and proteasome inhibitors has seen an increase in overall patient survival, however, for the majority of patients, relapse remains inevitable with evidence that these agents, like the conventional chemotherapeutics are also subject to the development of MDR. Clinical management of patients with MM is currently compromised by lack of a suitable procedure to monitor the development of clinical drug resistance in individual patients. The current MM prognostic measures fail to pick the clonotypic tumor cells overexpressing drug efflux pumps, and invasive biopsy is insufficient in detecting sporadic tumors in the skeletal system. This review summarizes the challenges associated with treating the complex disease spectrum of myeloma, with an emphasis on the role of deleterious multidrug resistant clones orchestrating relapse.
Collapse
Affiliation(s)
- Sabna Rajeev Krishnan
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ritu Jaiswal
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ross D Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Frederick Luk
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Mary Bebawy
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| |
Collapse
|
15
|
A multicenter clinical study to determine the feasible initial dose of lenalidomide for maintenance therapy in patients with multiple myeloma following autologous peripheral blood stem-cell transplantation. Mol Clin Oncol 2016; 4:965-970. [PMID: 27313858 DOI: 10.3892/mco.2016.833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/13/2016] [Indexed: 11/05/2022] Open
Abstract
Maintenance therapy with lenalidomide (LEN) for patients with multiple myeloma (MM) following autologous peripheral blood stem cell transplantation (auto-PBSCT) may be a promising option for preventing relapse or disease progression. However, the recommended dose of LEN has yet to be firmly established. We herein report the results of a multicenter clinical study for determining the feasible initial dose (FID) of LEN. In this trial, a total of 11 patients who achieved a very good partial response or complete response following auto-PBSCT were enrolled from five transplant centers in Japan. Three dose levels of LEN (level 0, 5 mg; level 1, 10 mg; and level 2,:15 mg) were tested in this study. FID was defined as the maximum estimated dose at which 70% of the patients could receive maintenance therapy for 12 weeks without any serious adverse events or disease progression. Using a continual reassessment method, 6 patients were assigned to level 0 and the remaining 5 patients were assigned to level 1. All 6 patients (100%) at level 0, but only 2 patients (40%) at level 1, completed 12 weeks of administration with their assigned dose of LEN. The results of our study demonstrated that, although 5 mg of LEN was acceptable in terms of safety, 7.5 mg of LEN may also be an acceptable FID.
Collapse
|
16
|
Kim H, Bhattacharya A, Qi L. Endoplasmic reticulum quality control in cancer: Friend or foe. Semin Cancer Biol 2015; 33:25-33. [PMID: 25794824 DOI: 10.1016/j.semcancer.2015.02.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/25/2015] [Indexed: 12/22/2022]
Abstract
Quality control systems in the endoplasmic reticulum (ER) mediated by unfolded protein response (UPR) and endoplasmic reticulum associated degradation (ERAD) ensure cellular function and organismal survival. Recent studies have suggested that ER quality-control systems in cancer cells may serve as a double-edged sword that aids progression as well as prevention of tumor growth in a context-dependent manner. Here we review recent advances in our understanding of the complex relationship between ER proteostasis and cancer pathology, with a focus on the two most conserved ER quality-control mechanisms--the IRE1α-XBP1 pathway of the UPR and SEL1L-HRD1 complex of the ERAD.
Collapse
Affiliation(s)
- Hana Kim
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, United States
| | - Asmita Bhattacharya
- Graduate Program in Genetics Genomics and Development, Cornell University, Ithaca, NY 14853, United States
| | - Ling Qi
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, United States; Graduate Program in Genetics Genomics and Development, Cornell University, Ithaca, NY 14853, United States.
| |
Collapse
|
17
|
Gul Z, Bashir Q, Cremer M, Yusuf SW, Gunaydin H, Arora S, Slone S, Nieto Y, Sherwani N, Parmar S, Shah N, Dinh YT, Hosing CM, Popat UR, Kebriaei P, Shpall EJ, Giralt SA, Champlin RE, Qazilbash MH. Short-term cardiac toxicity of autologous hematopoietic stem cell transplant for multiple myeloma. Leuk Lymphoma 2015; 56:533-5. [PMID: 24882259 DOI: 10.3109/10428194.2014.926346] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Zartash Gul
- Department of Hematology/BMT, Markey Cancer Center, University of Kentucky Medical Center , Lexington, KY , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Does the dogma that multiple myeloma is incurable still hold?. The genomic chaos and resulting resistance to apoptosis of myeloma, long considered an obstacle to cure, formed the basis of Total Therapy (TT) program. The TT approach uses all myeloma-active drugs upfront to target drug-resistant subclones during initial treatment to prevent later relapse. Long-term follow-up of 1202 patients (TT1: n = 231, median follow-up: 21 years; TT2: 668, median follow-up: 12 years; TT3a: n = 303, median follow-up: 9 years) permitted investigation of whether progression-free survival (PFS) and complete response (CR) duration were consistent with curability, ie observation of plateaus in Kaplan-Meier plots for PFS and CR duration. In the subset of 627 patients with plasma cell gene expression profiling data, cure plateaus were apparent at 5 years in the 14% with high-risk myeloma compared with 10 years in the remainder with low-risk disease. A parametric model based on PFS and CR duration supported an increase in curability: 10-year PFS and CR estimates increased from 8.8%/17.9% in TT1 to 15.5%/28.2% in TT2's control arm to 25.1%/35.6% in TT2's thalidomide arm and to 32.9%/48.8% in TT3a. Toward developing novel therapies, we recommend a concerted focus on patients with high-risk myeloma whose outcome has not been advanced.
Collapse
|
19
|
Turner JG, Dawson J, Cubitt CL, Baz R, Sullivan DM. Inhibition of CRM1-dependent nuclear export sensitizes malignant cells to cytotoxic and targeted agents. Semin Cancer Biol 2014; 27:62-73. [PMID: 24631834 PMCID: PMC4108511 DOI: 10.1016/j.semcancer.2014.03.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 02/25/2014] [Accepted: 03/01/2014] [Indexed: 10/25/2022]
Abstract
Nuclear-cytoplasmic trafficking of proteins is a significant factor in the development of cancer and drug resistance. Subcellular localization of exported proteins linked to cancer development include those involved in cell growth and proliferation, apoptosis, cell cycle regulation, transformation, angiogenesis, cell adhesion, invasion, and metastasis. Here, we examined the basic mechanisms involved in the export of proteins from the nucleus to the cytoplasm. All proteins over 40kDa use the nuclear pore complex to gain entry or exit from the nucleus, with the primary nuclear export molecule involved in these processes being chromosome region maintenance 1 (CRM1, exportin 1 or XPO1). Proteins exported from the nucleus must possess a hydrophobic nuclear export signal (NES) peptide that binds to a hydrophobic groove containing an active-site Cys528 in the CRM1 protein. CRM1 inhibitors function largely by covalent modification of the active site Cys528 and prevent binding to the cargo protein NES. In the absence of a CRM1 inhibitor, CRM1 binds cooperatively to the NES of the cargo protein and RanGTP, forming a trimer that is actively transported out of the nucleus by facilitated diffusion. Nuclear export can be blocked by CRM1 inhibitors, NES peptide inhibitors or by preventing post-translational modification of cargo proteins. Clinical trials using the classic CRM1 inhibitor leptomycin B proved too toxic for patients; however, a new generation of less toxic small molecule inhibitors is being used in clinical trials in patients with both hematological malignancies and solid tumors. Additional trials are being initiated using small-molecule CRM1 inhibitors in combination with chemotherapeutics such as pegylated liposomal doxorubicin. In this review, we present evidence that combining the new CRM1 inhibitors with other classes of therapeutics may prove effective in the treatment of cancer. Potential combinatorial therapies discussed include the use of CRM1 inhibitors and the addition of alkylating agents (melphalan), anthracyclines (doxorubicin and daunomycin), BRAF inhibitors, platinum drugs (cisplatin and oxaliplatin), proteosome inhibitors (bortezomib and carfilzomib), or tyrosine-kinase inhibitors (imatinib). Also, the sequence of treatment may be important for combination therapy. We found that the most effective treatment regimen involved first priming the cancer cells with the CRM1 inhibitor followed by doxorubicin, bortezomib, carfilzomib, or melphalan. This order sensitized both de novo and acquired drug-resistant cancer cell lines.
Collapse
Affiliation(s)
- Joel G Turner
- Department of Blood and Marrow Transplantation and Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jana Dawson
- Department of Blood and Marrow Transplantation and Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Christopher L Cubitt
- Translational Research Core Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Rachid Baz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Daniel M Sullivan
- Department of Blood and Marrow Transplantation and Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
20
|
Kapoor P, Kumar SK, Dispenzieri A, Lacy MQ, Buadi F, Dingli D, Russell SJ, Hayman SR, Witzig TE, Lust JA, Leung N, Lin Y, Zeldenrust SR, McCurdy A, Greipp PR, Kyle RA, Rajkumar SV, Gertz MA. Importance of achieving stringent complete response after autologous stem-cell transplantation in multiple myeloma. J Clin Oncol 2013; 31:4529-35. [PMID: 24248686 DOI: 10.1200/jco.2013.49.0086] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To study the impact of achieving stringent complete response (sCR), an increasingly attainable goal, after autologous stem-cell transplantation (ASCT) in patients with multiple myeloma (MM). PATIENTS AND METHODS Maximal response rates were determined in 445 consecutive patients who underwent ASCT within 12 months of diagnosis of MM. The patients achieving varying degrees of complete response (CR) are the focus of our study. RESULTS One hundred and nine patients (25%) achieved sCR after ASCT. The median overall survival (OS) rate from the time of transplantation for patients attaining sCR was not reached (NR), in contrast to those patients achieving conventional complete response (CR; n = 37; OS, 81 months) or near CR (nCR; n = 91; OS, 60 months; P < .001). Five-year OS rates were 80%, 53%, and 47% for sCR, CR, and nCR, respectively. The median time to progression (TTP) from ASCT of patients achieving sCR was significantly longer (50 months) than TTP of patients achieving CR or nCR (20 months and 19 months, respectively). On multivariable analysis, post-ASCT response of sCR was an independent prognostic factor for survival (hazard ratio, 0.44; 95% CI, 0.25 to 0.80; versus CR; P = .008), in addition to proliferation rate, pre-ASCT cytogenetics, and performance status. OS rates of patients attaining sCR continued to remain superior at 2-year landmark (median, NR v 70 months for conventional CR group; P = .007). CONCLUSION Improved long-term outcome is seen after ASCT with achievement of sCR when compared with lesser degrees of responses. Myeloma trials reporting the response rates should identify patients achieving sCR and CR separately, owing to markedly disparate outcomes of the two categories.
Collapse
|
21
|
Abstract
The incorporation of novel agents such as bortezomib and lenalidomide into initial therapy for multiple myeloma has improved the response rate of induction regimens. Also, these drugs are being increasingly used in the peri-transplant setting for transplant-eligible patients, and as part of consolidation and/or maintenance after front-line treatment, including in transplant-ineligible patients. Together, these and other strategies have contributed to a prolongation of progression-free survival (PFS) and overall survival (OS) in myeloma patients, and an increasing proportion are able to sustain a remission for many years. Despite these improvements, however, the vast majority of patients continue to suffer relapses, which suggests a prominent role for either primary, innate drug resistance, or secondary, acquired drug resistance. As a result, there remains a strong need to develop new proteasome inhibitors and immunomodulatory agents, as well as new drug classes, which would be effective in the relapsed and/or refractory setting, and overcome drug resistance. This review will focus on novel drugs that have reached phase III trials, including carfilzomib and pomalidomide, which have recently garnered regulatory approvals. In addition, agents that are in phase II or III, potentially registration-enabling trials will be described as well, to provide an overview of the possible landscape in the relapsed and/or refractory arena over the next 5 years.
Collapse
Affiliation(s)
- Robert Z Orlowski
- Department of Lymphoma/Myeloma, and Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
22
|
Leung-Hagesteijn C, Erdmann N, Cheung G, Keats JJ, Stewart AK, Reece D, Chung KC, Tiedemann RE. Xbp1s-negative tumor B cells and pre-plasmablasts mediate therapeutic proteasome inhibitor resistance in multiple myeloma. Cancer Cell 2013; 24:289-304. [PMID: 24029229 PMCID: PMC4118579 DOI: 10.1016/j.ccr.2013.08.009] [Citation(s) in RCA: 269] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 06/13/2013] [Accepted: 08/13/2013] [Indexed: 12/11/2022]
Abstract
Proteasome inhibitor (PI) resistance mechanisms in multiple myeloma (MM) remain controversial. We report the existence of a progenitor organization in primary MM that recapitulates maturation stages between B cells and plasma cells and that contributes to clinical PI resistance. Xbp1s(-) tumor B cells and pre-plasmablasts survive therapeutic PI, preventing cure, while maturation arrest of MM before the plasmablast stage enables progressive disease on PI treatment. Mechanistically, suppression of Xbp1s in MM is shown to induce bortezomib resistance via de-commitment to plasma cell maturation and immunoglobulin production, diminishing endoplasmic reticulum (ER) front-loading and cytotoxic susceptibility to PI-induced inhibition of ER-associated degradation. These results reveal the tumor progenitor structure in MM and highlight its role in therapeutic failure.
Collapse
Affiliation(s)
| | | | - Grace Cheung
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - A Keith Stewart
- Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Donna Reece
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | | | - Rodger E Tiedemann
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
- Contact Information: Dr. Rodger E. Tiedemann, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada, Tel: 416-581-8451, Fax: 416-946-6546,
| |
Collapse
|
23
|
Azevedo LM, Lansdell TA, Ludwig JR, Mosey RA, Woloch DK, Cogan DP, Patten GP, Kuszpit MR, Fisk JS, Tepe JJ. Inhibition of the human proteasome by imidazoline scaffolds. J Med Chem 2013; 56:5974-8. [PMID: 23789888 DOI: 10.1021/jm400235r] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The proteasome has emerged as the primary target for the treatment of multiple myeloma. Unfortunately, nearly all patients develop resistance to competitive-type proteasome inhibitors such as bortezomib. Herein, we describe the optimization of noncompetitive proteasome inhibitors to yield derivatives that exhibit nanomolar potency (compound 49, IC50 130 nM) toward proteasome inhibition and overcome bortezomib resistance. These studies illustrate the feasibility of the development of noncompetitive proteasome inhibitors as additives and/or alternatives to competitive proteasome inhibitors.
Collapse
Affiliation(s)
- Lauren M Azevedo
- Department of Chemistry, Michigan State University , East Lansing, Michigan 48824, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lansdell TA, Hurchla MA, Xiang J, Hovde S, Weilbaecher KN, Henry RW, Tepe JJ. Noncompetitive modulation of the proteasome by imidazoline scaffolds overcomes bortezomib resistance and delays MM tumor growth in vivo. ACS Chem Biol 2013. [PMID: 23198928 DOI: 10.1021/cb300568r] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Multiple myeloma (MM) is a malignant disorder of differentiated B-cells for which standard care involves the inhibition of the proteasome. All clinically used proteasome inhibitors, including the chemotherapeutic drug bortezomib, target the catalytic active sites of the proteasome and inhibit protein proteolysis by competing with substrate binding. However, nearly all (~97%) patients become intolerant or resistant to treatments within a few years, after which the average survival time is less than 1 year. We describe herein the inhibition of the human proteasome via a noncompetitive mechanism by the imidazoline scaffold, TCH-13. Consistent with a mechanism distinct from that of competitive inhibitors, TCH-013 acts additively with and overcomes resistance to bortezomib. Importantly, TCH-013 induces apoptosis in a panel of myeloma and leukemia cell lines, but in contrast, normal lymphocytes, primary bone marrow stromal cells (hBMSC), and macrophages are resistant to its cytotoxic effects. TCH-013 was equally effective in blocking MM cell growth in co-cultures of MM cells with hBMSC isolated from CD138 negative bone marrow (BM) samples of MM patients. The cellular activity translated well in vivo where TCH-013 delayed tumor growth in an MM xenograft model to a similar extent as bortezomib.
Collapse
Affiliation(s)
| | - Michelle A. Hurchla
- Department of Medicine, Division
of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jingyu Xiang
- Department of Medicine, Division
of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States
| | | | - Katherine N. Weilbaecher
- Department of Medicine, Division
of Oncology, Washington University School of Medicine, St. Louis, Missouri, United States
| | | | | |
Collapse
|