1
|
Samala RV, Nurse DP, Chen X, Wei W, Crook JJ, Fada SD, Valent J. Effects of early palliative care integration on patients with newly diagnosed multiple myeloma. Support Care Cancer 2024; 32:468. [PMID: 38937310 DOI: 10.1007/s00520-024-08665-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE While numerous studies underscore the benefits of early palliative care (EPC) for patients with solid tumors, its effects on patients with multiple myeloma (MM) are not as widely known. This study aims to determine the effects of EPC integration on patients with newly diagnosed symptomatic MM and the feasibility of this approach. METHODS This prospective cohort study enrolled patients within eight weeks of diagnosis. Participants met with a palliative care team monthly for 12 months. Functional Assessment of Cancer Therapy-General (FACT-G) plus Multiple Myeloma Subscale (FACT-MM), and Hospital Anxiety and Depression Scale (HADS) were administered upon enrollment and every three months. Proportion of completed visits and assessments determined the feasibility of EPC. RESULTS Of the twenty participants enrolled from January 2020 to November 2022, median age was 65 (range 40, 77), 15 (75%) were female, 15 (75%) were white, 65% completed assessments at six months, and 60% at 12 months. The following measures significantly improved at 12 months versus baseline: FACT-G scores increased by 15.1 points (adjusted 95% CI: 2.2-28.1, adjusted p = 0.02); Functional Well-Being scores increased by 6.0 points (adjusted 95% CI: 1.1-10.9, adjusted p = 0.01); and Pain Subscale scores increased by 3.4 points (adjusted 95% CI: 0.5-6.4, adjusted p = 0.02). Depression and anxiety scores did not significantly change over time. CONCLUSION Functional well-being, pain experience and overall QOL improved in a cohort of patients with newly diagnosed MM after 12 months of EPC involvement. Although monthly visits seemed feasible, the findings suggest that further research is needed to explore the optimal timing of palliative care interventions in the MM trajectory. TRIAL REGISTRATION ClinicalTrials.gov ID NCT04248244 (Registration Date: January 30, 2020).
Collapse
Affiliation(s)
- Renato V Samala
- Department of Palliative and Supportive Care, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Ave. CA-53, Cleveland, OH, 44195, USA.
| | - Daniel P Nurse
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Xiaoying Chen
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Wei Wei
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jasmine J Crook
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sherry D Fada
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jason Valent
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
2
|
Rosiñol L, Hebraud B, Oriol A, Colin AL, Ríos Tamayo R, Hulin C, Blanchard MJ, Caillot D, Sureda A, Hernández MT, Arnulf B, Mateos MV, Macro M, San-Miguel J, Belhadj K, Lahuerta JJ, Garelik MB, Bladé J, Moreau P. Integrated analysis of randomized controlled trials evaluating bortezomib + lenalidomide + dexamethasone or bortezomib + thalidomide + dexamethasone induction in transplant-eligible newly diagnosed multiple myeloma. Front Oncol 2023; 13:1197340. [PMID: 38023148 PMCID: PMC10652744 DOI: 10.3389/fonc.2023.1197340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/09/2023] [Indexed: 12/01/2023] Open
Abstract
Objective Providing the most efficacious frontline treatment for newly diagnosed multiple myeloma (NDMM) is critical for patient outcomes. No direct comparisons have been made between bortezomib + lenalidomide + dexamethasone (VRD) and bortezomib + thalidomide + dexamethasone (VTD) induction regimens in transplant-eligible NDMM. Methods An integrated analysis was performed using patient data from four trials meeting prespecified eligibility criteria: two using VRD (PETHEMA GEM2012 and IFM 2009) and two using VTD (PETHEMA GEM2005 and IFM 2013-04). Results The primary endpoint was met, with VRD demonstrating a noninferior rate of at least very good partial response (≥ VGPR) after induction vs VTD. GEM comparison demonstrated improvement in the ≥ VGPR rate after induction for VRD vs VTD (66.3% vs 51.2%; P = .00281) that increased after transplant (74.4% vs 53.5%). Undetectable minimal residual disease rates post induction (46.7% vs 34.9%) and post transplant (62.4% vs 47.3%) support the benefit of VRD vs VTD. Treatment-emergent adverse events leading to study and/or treatment discontinuation were less frequent with VRD (3%, GEM2012; 6%, IFM 2009) vs VTD (11%, IFM 2013-04). Conclusion These results supported the benefit of VRD over VTD for induction in transplant-eligible patients with NDMM. The trials included are registered with ClinicalTrials.gov (NCT01916252, NCT01191060, NCT00461747, and NCT01971658).
Collapse
Affiliation(s)
- Laura Rosiñol
- Department of Hematology, Hospital Clínic Institut d’investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Benjamin Hebraud
- Hematology Department, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Albert Oriol
- Institut Català d’Oncologia I Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Anne-Laurène Colin
- Service de Pharmacologie Médicale et Clinique, Centre Hospitalier et Universitaire de Toulouse, Toulouse, France
| | - Rafael Ríos Tamayo
- Department of Hematology, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Cyrille Hulin
- Department of Hematology, Hôpital Haut-Lévêque, Bordeaux Pessac, France
| | | | | | - Anna Sureda
- Institut Català d’Oncologia-Hospitalet i Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | | | - Bertrand Arnulf
- Centre Hospitalier Universitaire, Hôpital St-Louis, Paris, France
| | - Maria-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Margaret Macro
- Institut d’Hématologie de Basse Normandie, Centre Hospitalier et Universitaire de Caen, Caen, France
| | - Jesús San-Miguel
- Clínica Universidad de Navarra (CUN), Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Karim Belhadj
- Lymphoid Malignancies Unit, Centre Hospitalier et Universitaire Henri Mondor, Creteil, France
| | - Juan José Lahuerta
- Clínica Universidad de Navarra (CUN), Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | | | - Joan Bladé
- Department of Hematology, Hospital Clínic Institut d’investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Philippe Moreau
- Department of Hematology, University Hospital Hôtel-Dieu, Nantes, France
| |
Collapse
|
3
|
Suzuki K, Shimazu Y, Minakata D, Ikeda T, Takahashi H, Tsukada N, Kanda Y, Doki N, Nishiwaki K, Miwa A, Sawa M, Kataoka K, Hiramoto N, Ota S, Itagaki M, Ichinohe T, Atsuta Y, Yano S, Kawamura K. Efficacy of Autologous Stem Cell Transplantation for Myeloma Patients with Suboptimal Response: A Multicenter Retrospective Analysis. Transplant Cell Ther 2023; 29:688.e1-688.e13. [PMID: 37574125 DOI: 10.1016/j.jtct.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/20/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
Autologous stem cell transplantation (ASCT) is the standard of care for myeloma patients who achieve partial response (PR) or better after induction therapy. However, its clinical significance in patients with suboptimal response (SR) before ASCT, including stable disease (SD) and progressive disease (PD), has not been established. Additionally, functional high-risk, including SR and early PD within 12 months, was a poor prognostic factor up to now. This study aimed to evaluate the efficacy of ASCT in myeloma patients with SR in the novel agent era. This multicenter retrospective study was conducted using the Transplant Registry Unified Management Program database of the Japanese Society of Transplantation and Cellular Therapy and included 3898 transplantation-eligible patients with newly diagnosed multiple myeloma who underwent ASCT between 2007 and 2020 and were followed up until 2021. The SR rate was 4.7%, including 1.7% with PD. In survival time analysis for overall cases, a significant difference in PFS between the very good partial response (VGPR) and PR groups was observed, whereas there was no significant difference in overall survival (OS) between the VGPR and PR groups. Additionally, there was no significant difference in OS or PFS between the PR and SD groups. Therefore, we focused on the PR, SD, and PD groups, as the purpose of this retrospective study was to investigate the clinical significance of ASCT in patients with SR compared with those with PR. The median patient age was 60 years (range, 30 to 77 years). In total, 1605 (97.4%) patients received bortezomib, 561 (38.2%) received an immunomodulatory drug (ImiD), and 512 (34.9%) received both bortezomib and an ImiD. A total of 558 patients (38.0%) received reinduction therapy. There were 229 patients (37.7%) with high-risk cytogenetics (HRCA). With a median follow-up of 31.7 months, there was a significant difference in 30-month OS rates among the PR, SD, and PD groups (86.3%, 78.5%, and 39.4%, respectively; P <.001). OS was significantly shorter in the SD group compared to the PR group among the patients with HRCA (P < .001) and patients treated with reinduction therapy (P = .013). In the PD group, the 30-month OS and PFS rates were 39.4% and 17.9%, respectively. Finally, early PD within 12 months after ASCT was predictive of short OS, whereas OS without early PD even in the PD group was similar to that in the SD and PR groups. In conclusion, OS in the SR group was not always short, but SR in the HRCA and the reinduction therapy groups was predictive of short OS, so that therapeutic alternatives to ASCT are needed. OS in the PD group was significantly short, but ASCT improved clinical outcomes when early PD did not occur even in the PD group.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology/Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan; Division of Clinical Oncology/Hematology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Chiba, Japan.
| | - Yutaka Shimazu
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Minakata
- Division of Hematology, Jichi Medical University, Tochigi, Japan
| | - Takashi Ikeda
- Division of Hematology and Stem Cell Transplantation, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroyuki Takahashi
- Department of Hematology and Medical Oncology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Nobuhiro Tsukada
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Jichi Medical University, Tochigi, Japan; Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kaichi Nishiwaki
- Division of Clinical Oncology/Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan; Division of Clinical Oncology/Hematology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Chiba, Japan
| | - Akiyoshi Miwa
- Department of Hematology, Tokyo-Kita Medical Center, Tokyo, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Aichi, Japan
| | - Keisuke Kataoka
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiro Hiramoto
- Department of Hematology, Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Hokkaido, Japan
| | - Mitsuhiro Itagaki
- Department of Hematology, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Hiroshima, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Aichi, Japan; Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Aichi, Japan
| | - Shingo Yano
- Division of Clinical Oncology/Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Koji Kawamura
- Division of Clinical Laboratory Medicine, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Tottori, Japan
| |
Collapse
|
4
|
Richardson PG, Facon T, Venner CP, Bahlis NJ, Offner F, White D, Karlin L, Benboubker L, Voog E, Yoon S, Suzuki K, Shibayama H, Zhang X, Villarreal M, Twumasi‐Ankrah P, Labotka R, Rifkin RM, Lonial S, Kumar SK, Rajkumar SV, Moreau P. Late versus early response and depth of response are associated with improved outcomes in patients with newly diagnosed multiple myeloma enrolled in the TOURMALINE-MM2 trial. EJHAEM 2023; 4:995-1005. [PMID: 38024593 PMCID: PMC10660432 DOI: 10.1002/jha2.759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 12/01/2023]
Abstract
Deeper responses are associated with longer survival in multiple myeloma (MM); however, limited data exist on the impact of response kinetics on outcomes. We investigated progression-free survival (PFS) and duration of response (DOR) by response depth and in early (best confirmed response 0-4 months; n = 424) versus late responders (best confirmed response >4 months; n = 281). Newly diagnosed patients enrolled in TOURMALINE-MM2 receiving ixazomib-lenalidomide-dexamethasone (IRd) (n = 351) or placebo-Rd (n = 354) were evaluated post hoc. Deeper responses were associated with longer PFS (complete response [CR] not reached [NR], very good partial response [VGPR] 37.2 months, partial response [PR] 16.4 months) and DOR (CR NR, VGPR 42.6 months, PR 15.4 months). Among patients with a PFS (n = 511) or DOR (n = 484) of ≥6 months who achieved ≥PR, median PFS was prolonged among late versus early responders receiving IRd (59.7 vs. 17.9 months) or placebo-Rd (56.6 vs. 12.4 months), as was median DOR (IRd, NR vs. 20.9 months; placebo-Rd, 58.2 vs. 11.7 months). While the treatment paradigm for newly diagnosed MM is treatment to progression, our findings suggest slowness of response to a proteasome inhibitor-immunomodulatory drug-steroid combination is not a negative predictor of outcome.
Collapse
Affiliation(s)
- Paul G. Richardson
- Harvard Medical SchoolJerome Lipper Multiple Myeloma Center, Dana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Thierry Facon
- Centre Hospitalier Universitaire (CHU) LilleService des Maladies du Sang, University of LilleLilleFrance
| | - Christopher P. Venner
- Cross Cancer InstituteUniversity of AlbertaEdmontonAlbertaCanada
- BC Cancer Vancouver CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Nizar J. Bahlis
- Arnie Charbonneau Cancer InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | | | - Darrell White
- QEII Health Sciences Center and Dalhousie UniversityHalifaxNova ScotiaCanada
| | | | | | | | - Sung‐Soo Yoon
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
| | | | | | - Xiaoquan Zhang
- Takeda Development Center Americas, Inc. (TDCA)LexingtonMassachusettsUSA
| | - Miguel Villarreal
- Takeda Development Center Americas, Inc. (TDCA)LexingtonMassachusettsUSA
| | | | - Richard Labotka
- Takeda Development Center Americas, Inc. (TDCA)LexingtonMassachusettsUSA
| | - Robert M. Rifkin
- US Oncology Research – Rocky Mountain Cancer CentersDenverColoradoUSA
| | - Sagar Lonial
- Department of Hematology and Medical OncologyWinship Cancer InstituteEmory University School of MedicineAtlantaGeorgiaUSA
| | | | | | | |
Collapse
|
5
|
Jew S, Bujarski S, Regidor B, Emamy-Sadr M, Swift R, Eades B, Kim S, Eshaghian S, Berenson JR. Clinical Outcomes and Serum B-Cell Maturation Antigen Levels in a Real-World Unselected Population of Newly Diagnosed Multiple Myeloma Patients. Target Oncol 2023; 18:735-747. [PMID: 37682503 DOI: 10.1007/s11523-023-00990-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Progression-free survival (PFS) and overall survival (OS) of newly diagnosed multiple myeloma (MM) patients have been widely published in the clinical trials setting, but data published from real-world settings are limited. OBJECTIVE We determined the survival and factors that predict outcomes among 161 unselected, newly diagnosed MM patients whose frontline therapy was started at a single clinic specializing in the treatment of this B-cell malignancy. PATIENTS AND METHODS None of these patients underwent an autologous stem cell transplantation as part of their initial therapy and the population had a high proportion (35%) of cytogenetic high-risk patients. RESULTS With a median follow-up of 42.7 months, the cohort had a median PFS of 22.8 months and a median OS of 136.2 months. The 1-, 3-, and 5-year survival rates were 97.5%, 85.3%, and 76.2%, respectively. These results are considerably better than those reported from patients enrolled in clinical trials and those from countries with national registries. Age <65 years predicted for a longer OS (p = 0.0004). Baseline serum B-cell maturation antigen (sBCMA) levels were also assessed and showed median and mean levels of 320.3 ng/mL and 551.1 ng/mL, respectively. Furthermore, patients with baseline sBCMA levels in the lowest quartile (≤136.2 ng/mL) showed a longer PFS (p = 0.0262). CONCLUSION These results provide clinicians with a real-world understanding of the survival of unselected, newly diagnosed patients initiating therapy in a clinic specializing in the care of MM patients.
Collapse
Affiliation(s)
- Scott Jew
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA
- Berenson Cancer Center, West Hollywood, CA, USA
| | - Sean Bujarski
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA
- Berenson Cancer Center, West Hollywood, CA, USA
| | | | | | | | | | | | | | - James R Berenson
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA.
- Berenson Cancer Center, West Hollywood, CA, USA.
- ONCOtherapeutics, West Hollywood, CA, USA.
- ONCOtracker, West Hollywood, CA, USA.
| |
Collapse
|
6
|
Samur MK, Roncador M, Aktas Samur A, Fulciniti M, Bazarbachi AH, Szalat R, Shammas MA, Sperling AS, Richardson PG, Magrangeas F, Minvielle S, Perrot A, Corre J, Moreau P, Thakurta A, Parmigiani G, Anderson KC, Avet-Loiseau H, Munshi NC. High-dose melphalan treatment significantly increases mutational burden at relapse in multiple myeloma. Blood 2023; 141:1724-1736. [PMID: 36603186 PMCID: PMC10273091 DOI: 10.1182/blood.2022017094] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
High-dose melphalan (HDM) improves progression-free survival in multiple myeloma (MM), yet melphalan is a DNA-damaging alkylating agent; therefore, we assessed its mutational effect on surviving myeloma cells by analyzing paired MM samples collected at diagnosis and relapse in the IFM 2009 study. We performed deep whole-genome sequencing on samples from 68 patients, 43 of whom were treated with RVD (lenalidomide, bortezomib, and dexamethasone) and 25 with RVD + HDM. Although the number of mutations was similar at diagnosis in both groups (7137 vs 7230; P = .67), the HDM group had significantly more mutations at relapse (9242 vs 13 383, P = .005). No change in the frequency of copy number alterations or structural variants was observed. The newly acquired mutations were typically associated with DNA damage and double-stranded breaks and were predominantly on the transcribed strand. A machine learning model, using this unique pattern, predicted patients who would receive HDM with high sensitivity, specificity, and positive prediction value. Clonal evolution analysis showed that all patients treated with HDM had clonal selection, whereas a static progression was observed with RVD. A significantly higher percentage of mutations were subclonal in the HDM cohort. Intriguingly, patients treated with HDM who achieved complete remission (CR) had significantly more mutations at relapse yet had similar survival rates as those treated with RVD who achieved CR. This similarity could have been due to HDM relapse samples having significantly more neoantigens. Overall, our study identifies increased genomic changes associated with HDM and provides rationale to further understand clonal complexity.
Collapse
Affiliation(s)
- Mehmet Kemal Samur
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | | | - Anil Aktas Samur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Abdul Hamid Bazarbachi
- Department of Internal Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, New York, NY
| | - Raphael Szalat
- Department of Hematology and Medical Oncology, Boston University Medical Center, Boston, MA
| | - Masood A. Shammas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Adam S. Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Paul G. Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Florence Magrangeas
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | - Stephane Minvielle
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | - Aurore Perrot
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Jill Corre
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Philippe Moreau
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | | | - Giovanni Parmigiani
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | - Kenneth C. Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Hervé Avet-Loiseau
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Nikhil C. Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
- VA Boston Healthcare System, Boston, MA
| |
Collapse
|
7
|
Fonseca R, Arribas M, Wiedmeier-Nutor JE, Kusne YN, González Vélez M, Kosiorek HE, Butterfield RDJ, Kirsch IR, Mikhael JR, Stewart AK, Reeder C, Larsen J, Bergsagel PL, Fonseca R. Integrated analysis of next generation sequencing minimal residual disease (MRD) and PET scan in transplant eligible myeloma patients. Blood Cancer J 2023; 13:32. [PMID: 36878906 PMCID: PMC9988896 DOI: 10.1038/s41408-023-00794-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 03/08/2023] Open
Abstract
Minimal residual disease (MRD) assays allow response assessment in patients with multiple myeloma (MM), and negativity is associated with improved survival outcomes. The role of highly sensitive next generation sequencing (NGS) MRD in combination with functional imaging remains to be validated. We performed a retrospective analysis on MM patients who underwent frontline autologous stem cell transplant (ASCT). Patients were evaluated at day 100 post-ASCT with NGS-MRD and positron emission tomography (PET-CT). Patients with ≥ 2 MRD measurements were included in a secondary analysis for sequential measurements. 186 patients were included. At day 100, 45 (24.2%) patients achieved MRD negativity at a sensitivity threshold of 10-6. MRD negativity was the most predictive factor for longer time to next treatment (TTNT). Negativity rates did not differ according to MM subtype, R-ISS Stage nor cytogenetic risk. PET-CT and MRD had poor agreement, with high rates of PET-CT negativity in MRD-positive patients. Patients with sustained MRD negativity had longer TTNT, regardless of baseline risk characteristics. Our results show that the ability to measure deeper and sustainable responses distinguishes patients with better outcomes. Achieving MRD negativity was the strongest prognostic marker and could help guide therapy-related decisions and serve as a response marker for clinical trials.
Collapse
Affiliation(s)
- Rodrigo Fonseca
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Mariano Arribas
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Yael N Kusne
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Heidi E Kosiorek
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Ilan R Kirsch
- Translational Medicine, Adaptive Biotechnologies, Seattle, WA, USA
| | - Joseph R Mikhael
- Translational Genomics Research Institute, City of Hope Cancer Center, Phoenix, AZ, USA
| | - A Keith Stewart
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Craig Reeder
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Jeremy Larsen
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - P Leif Bergsagel
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Rafael Fonseca
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA.
| |
Collapse
|
8
|
Hu SL, Liu M, Zhang JY. Comparing the efficacy of different dexamethasone regimens for maintenance treatment of multiple myeloma in standard-risk patients non-eligible for transplantation. World J Clin Cases 2022; 10:11712-11725. [PMID: 36405288 PMCID: PMC9669873 DOI: 10.12998/wjcc.v10.i32.11712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/13/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a plasma cell malignancy, while MM outcomes have significantly improved due to novel agents and combinations, MM remains an incurable disease. The key goal of treatment in MM is to achieve a maximal response and the subsequent consolidation of response after initial therapy. Many studies analyzed an improved progression-free survival (PFS) following lenalidomide alone maintenance versus placebo or observation after autologous stem cell transplant (ASCT) in patients with NDMM. In the SWOG S0777 clinical trial, patients newly diagnosed with MM (NDMM) without ASCT received lenalidomide plus low-dose dexamethasone (DXM) maintenance until progressive disease, where PFS and overall survival (OS) were significantly improved. In the present study, we assessed the efficacy and toxicity of the different doses of DXM combined with lenalidomide for maintenance treatment of NDMM for transplant noneligible patients in the standard-risk group.
AIM To investigate the efficacy and adverse effects of different administration modes of DXM combined with lenalidomide for maintenance treatment of MM in standard-risk patients ineligible for transplantation.
METHODS A total of 96 MM patients were enrolled in this study, among whom 48 patients received maintenance treatment that consisted of oral administration of 25 milligrams (mg) of lenalidomide from days 1-21 and 40 mg of DXM on days 1, 8, 15, and 22 (DXM 40 mg group), repeated every 4 wk. Another group was treated with oral administration of 25 mg of lenalidomide from days 1-21 and 20 mg of DXM on days 1-2, 8-9, 15-16, and 22-23 (DXM 20 mg group), which was also repeated every 4 wk.
RESULTS The median PFS was 37.25 mo in the DXM 40.00 mg group and 38.17 mo in the DXM 20 mg group (P = 0.171). The median OS was 50.78 mo in the DXM 40 mg group and 51.69 mo in the DXM 20 mg group (P = 0.171). Fourteen patients in the DXM 40 mg group and 6 patients in the DXM 20 mg group suffered from adverse gastrointestinal reactions after the oral administration of the DXM tablet (P = 0.044). Ten patients suffered from abnormal glucose tolerance (GTA), impaired fasting glucose (IFG), or diabetes mellitus in the DXM 40 mg group during our observation time compared to 19 patients with GTA, IFG, or DM in the DXM 20 mg group (P = 0.033). Abnormal β-crosslaps or higher were found in 5 patients in the DXM 40 mg group and 12 patients in the DXM 20 mg group (P = 0.049). Insomnia or an increase in insomnia compared to the previous condition was evident in 2 patients in the DXM 40 mg group after maintenance treatment for more than 6 mo compared to 11 patients in the DXM 20 mg group (P = 0.017).
CONCLUSION The DXM 40 mg group exhibited efficacy similar to that of the DXM 20 mg group. However, the DXM 40 mg group had significantly decreased toxicity compared with the DXM 20 mg group in the long term.
Collapse
Affiliation(s)
- Sai-Ling Hu
- Department of Cardiology, Lishui Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Min Liu
- Department of Hematology, Lishui Municipal Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Jun-Yu Zhang
- Department of Hematology, Lishui Municipal Central Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
9
|
Radiomics Models Based on Magnetic Resonance Imaging for Prediction of the Response to Bortezomib-Based Therapy in Patients with Multiple Myeloma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6911246. [PMID: 36105939 PMCID: PMC9467708 DOI: 10.1155/2022/6911246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/04/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022]
Abstract
Purpose. To identify significant radiomics features based on MRI and establish effective models for predicting the response to bortezomib-based regimens. Materials and Methods. In total, 95 MM patients treated with bortezomib-based therapy were enrolled, including 77 with bortezomib, cyclophosphamide, and dexamethasone (BCD) and 18 with bortezomib, lenalidomide, and dexamethasone (VRD). Based on T1-weighted imaging (T1WI) and T2-weighted imaging with fat suppression (T2WI-fs), radiomics features were extracted and then selected. The random forest (RF),
-nearest neighbor, support vector machine, logistic regression, decision tree, and Bayes models were built using the selected features. The predictive power of six models for response to BCD and VRD regimens were evaluated. The correlation between the selected features and progression-free survival (PFS) was also analyzed. Results. Four wavelet features were correlated with BCD treatment response. The six models all showed predictive power for BCD regimen (AUC: 0.84-0.896 in the training set, 0.801-0.885 in the validation set), and RF performed relatively better than others. Nevertheless, all the BCD-based models were incapable of predicting the VRD treatment response. The wavelet-HLH_firstorder_kurtosis was also associated with PFS (log-rank
). Conclusion. The four wavelet features were valuable biomarkers for predicting the response to BCD regimen. The six models based on these features showed predictive power, and RF was the best. One wavelet feature was also a survival-related biomarker. MRI-based radiomics had the potential to guide clinicians in MM management.
Collapse
|
10
|
Steps towards a Multiple Myeloma Cure? J Pers Med 2022; 12:jpm12091451. [PMID: 36143236 PMCID: PMC9504254 DOI: 10.3390/jpm12091451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple myeloma survival has increased in last 20 years because of new treatments, better clinical management due to novel diagnostic tools such as imaging, and better understanding of the disease, biologically and genetically. Novel drugs have been introduced that act with different therapeutic mechanisms, but so have novel therapeutic strategies such as consolidation and maintenance after autologous stem cell transplant. Imaging (such as PET-CT and MRI) has been applied at diagnosis and after therapy for minimal residual disease monitoring. Multiparametric flow and molecular NGS may detect, with high-sensitivity, residual monoclonal plasma cells in the bone marrow. With this novel therapeutic and biological approach, a considerable fraction of multiple myeloma patients can achieve durable remission or even MGUS-like regression, which can ultimately lead to disease disappearance. The big dogma, “Myeloma is an incurable disease”, is hopefully fading.
Collapse
|
11
|
Jakubowiak AJ, Kumar S, Medhekar R, Pei H, Lefebvre P, Kaila S, He J, Lafeuille MH, Cortoos A, Londhe A, Mavros P, Lin TS, Usmani SZ. Daratumumab Improves Depth of Response and Progression-free Survival in Transplant-ineligible, High-risk, Newly Diagnosed Multiple Myeloma. Oncologist 2022; 27:e589-e596. [PMID: 35462406 PMCID: PMC9256027 DOI: 10.1093/oncolo/oyac067] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/04/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Patients with high-risk, newly diagnosed multiple myeloma (HR-NDMM) who are ineligible for autologous stem cell transplant (ASCT) have limited first-line treatment options. Recent meta-analyses evaluating the impact of incorporating daratumumab in the backbone regimen on progression-free survival (PFS) have found mixed results in these patients. MATERIALS AND METHODS A pooled analysis of patient-level data for ASCT-ineligible patients with HR-NDMM [ie, del(17p), t(4;14), t(14;16)] from the MAIA and ALCYONE trials; stratified by study identifier and adjusting for cytogenetic abnormality subtype, baseline performance status, International Staging System stage, myeloma type, and renal impairment; was conducted. Impact of daratumumab on PFS and rates of complete response or better (≥CR), minimal residual disease (MRD)-negative CR, very good partial response or better (≥VGPR), and overall response (ORR) was compared to control. RESULTS Among 101 patients in the daratumumab and 89 patients in the control cohort, median follow-up was 43.7 months. Daratumumab reduced the risk of progression or death by 41% (adjusted hazard ratio for PFS [95% confidence interval (CI)] = 0.59 [0.41-0.85]) versus control. At 36 months, the estimated proportion of patients who did not progress and were still alive was 41.3% in the daratumumab and 19.9% in the control cohort. Rates of ≥CR (41.6% vs. 22.5%), MRD-negative CR (24.8% vs. 5.6%), ≥VGPR (75.2% vs. 46.1%), and ORR (92.1% vs. 74.2%) were higher for daratumumab versus control. CONCLUSION These findings demonstrate that incorporation of daratumumab in frontline treatment regimens reduced the risk of progression or death and improved response rates among ASCT-ineligible HR-NDMM patients.
Collapse
Affiliation(s)
| | - Shaji Kumar
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Huiling Pei
- Janssen Research & Development, Raritan, NJ, USA
| | | | | | - Jianming He
- Janssen Global Services, LLC, Raritan, NJ, USA
| | | | | | - Anil Londhe
- Janssen Research & Development, Raritan, NJ, USA
| | | | - Thomas S Lin
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
| | - Saad Z Usmani
- Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| |
Collapse
|
12
|
Impact of Induction with VCD vs. VRD on the Outcome of Patients with Multiple Myeloma After an Autologous Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2022; 28:307.e1-307.e8. [PMID: 35331973 DOI: 10.1016/j.jtct.2022.03.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/01/2022] [Accepted: 03/17/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND nduction therapy with a triplet regimen, followed by high-dose therapy and autologous hematopoietic stem cell transplantation (auto-HCT) is the standard of care for newly diagnosed, transplant-eligible patients with multiple myeloma (MM). Bortezomib-dexamethasone with cyclophosphamide (VCD) or lenalidomide (VRD) are the most used induction regimens. However, previous studies comparing VCD and VRD showed disparate results. OBJECTIVES The goal of this retrospective study was to compare the "real world" results of VCD and VRD, in transplant-eligible MM patients outside of a clinical trial. METHODS/RESULTS We identified 322 patients who received VRD or VCD induction before auto-HCT at our institution. All patients received melphalan conditioning and single-agent lenalidomide maintenance therapy. Overall, 114 patients received VCD and 208 received VRD. The median age at auto-HCT was 61.9 years (range, 33.9-79.6), with 35.4% (114/322) of the cohort being 65 years or older. The overall response rate was 99.7% post-auto-HCT, with a significantly lower complete remission rate as the final response in the VCD compared to the VRD group (34% vs. 53%, p=0.001). However, there was no significant difference between the best response rate of VGPR or better in the VCD compared to the VRD group (92% vs. 85%, p=0.078). The median duration of ≥VGPR was 50.0 months (95%CI: 42.0-69.1) for both cohorts and there was no difference between VCD and VRD (p=0.769; HR, 0.95; 95% CI, 0.69-1.31). Median follow-up of survivors was 73 months. There was no difference in the relapse rate between VCD and VRD (p=0.749). Median progression-free survival (PFS) was 48.7 months in the VCD and 44.6 months in the VRD group (p=0.858). Median overall survival (OS) was 103.8 months with VCD and 101.7 months with VRD (p=0.891). At 5 years, the PFS and OS were 38.1% and 76.9% for the VCD group, respectively, and 40.7% and 74.6% for the VRD group, respectively. On multivariate analysis for OS in the entire cohort, R-ISS I and post-auto-HCT best response of stringent complete response (sCR)/CR emerged as significant predictors of superior OS. There was no impact of the type of induction regimen on the OS in the multivariate analysis. CONCLUSIONS Induction therapy with VCD compared to VRD was associated with a lower CR rate, but there was no difference in PFS or OS between the two regimens.
Collapse
|
13
|
Wålinder G, Genell A, Juliusson G, Svensson R, Santamaria AI, Crafoord J, Carlson K, Knut‐Bojanowska D, Veskovski L, Lauri B, Lund J, Turesson I, Hansson M, Blimark CH, Nahi H. Regional differences in treatment and outcome for myeloma patients in Sweden: A population based Swedish myeloma register study. Cancer Rep (Hoboken) 2022; 5:e1614. [PMID: 35243814 PMCID: PMC9675390 DOI: 10.1002/cnr2.1614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 01/11/2022] [Accepted: 02/16/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND We wanted to evaluate if health care for multiple myeloma (MM) patients is equal in different regions of Sweden. AIM To study differences in survival for MM depending on health care region and early use of modern treatment. METHODS AND RESULTS Data from the Swedish Myeloma Register from patients diagnosed between 2008 and 2017 was used. Cohorts were defined by the six healthcare regions (labeled A-F) in Sweden and modern initial treatment was defined as including certain drug combinations. To adjust for time to treatment bias, survival analyses were performed also for patients alive 6 months after diagnosis. In all treated MM patients (n = 5326), we observed a superior overall survival (OS) for region A compared to all other regions (p < .01 for all respectively). After adjusting for time to treatment there was also a superior survival in the region with highest use of modern initial treatment (region A) compared to the regions defined in the study as having intermediate and low use (p < .01 for both). In patients receiving autologous stem cell transplantation (ASCT) a superior survival was observed for region A compared to all regions besides region B. Similar results were seen when adjusting for a time to treatment bias. In patients not receiving ASCT, 75 years or older and adjusted for time to treatment bias, a difference was noted only between region A and E (log rank p = .04, HR 1.2, CI 1.00-1.44, p = .06). In multivariate analyses including age, international staging system stage and time period of diagnosis, differences in survival remained for patients receiving ASCT between region A versus C, D, E and F (p = .01, p < .01, p < .01, p = .03). CONCLUSION We observed a superior survival in region A for patients receiving ASCT. Explanations may be higher usage of modern initial treatment or regional residual confounding. For patients not receiving ASCT, 75 years or older, differences in survival could be adjusted for.
Collapse
Affiliation(s)
- Göran Wålinder
- Department of Medicine HuddingeKarolinska InstituteSolnaSweden,Department of HematologyKarolinska University Hospital HuddingeHuddingeSweden
| | | | | | - Ronald Svensson
- Department of HematologyLinköping University HospitalLinköpingSweden
| | | | - Jacob Crafoord
- Department of HematologyÖrebro University HospitalÖrebroSweden
| | - Kristina Carlson
- Department of HematologyUppsala University HospitalUppsalaSweden
| | | | | | | | - Johan Lund
- Department of Medicine HuddingeKarolinska InstituteSolnaSweden,Department of HematologyKarolinska University Hospital HuddingeHuddingeSweden
| | | | - Markus Hansson
- Department of HematologySahlgrenska University HospitalGothenburgSweden
| | | | - Hareth Nahi
- Department of Medicine HuddingeKarolinska InstituteSolnaSweden,Department of HematologyKarolinska University Hospital HuddingeHuddingeSweden
| |
Collapse
|
14
|
Ćojbašić I, Vučić M, Tijanić I, Ćojbašić Ž. Impact of quality of response on survival outcomes among multiple myeloma patients treated with novel agents - a retrospective analysis. SAO PAULO MED J 2022; 140:222-228. [PMID: 35195237 PMCID: PMC9610252 DOI: 10.1590/1516-3180.2021.0174.r2.22062021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/22/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND In this era of target therapies, novel data on the correlation between response endpoints and survival outcomes in multiple myeloma have arisen. OBJECTIVE To determine the impact of quality of response on clinical outcomes, using first-line treatment, and identify risk factors influencing progression-free survival (PFS) and overall survival (OS) among myeloma patients. DESIGN AND SETTING Retrospective analysis on myeloma patients who were treated at the Clinic of Hematology and Clinical Immunology, University Clinical Centre, Niš, Serbia, over a four-year period. METHODS A total of 108 newly diagnosed patients who received first-line therapy consisting of conventional chemotherapy or novel agent-based regimens were included in this analysis. RESULTS The quality of response to first-line therapy for the whole cohort was classified as follows: complete response (CR) in 19%; very good partial response (VGPR) in 23%; partial response (PR) in 38%; and less than PR for the remaining patients. After a median follow-up of 25.4 months, the three-year PFS and OS for the entire study population were 47% and 70%, respectively. Achievement of CR was the main factor associated with significantly prolonged PFS and OS, in comparison with patients who reached VGPR and PR. Likewise, addition of the new drugs bortezomib and thalidomide to standard chemotherapy led to considerably extended PFS and OS, compared with conventional therapy alone. CONCLUSIONS This analysis demonstrated that the quality of response after application of first-line treatment using novel agent-based regimens among multiple myeloma patients was a prognostic factor for PFS and OS, which are the most clinically relevant outcomes.
Collapse
Affiliation(s)
- Irena Ćojbašić
- MD, PhD. Assistant Professor, Department of Internal Medicine, Faculty of Medicine, University of Niš, Niš, Serbia; and Hematologist, Clinic of Hematology and Clinical Immunology, University Clinical Centre Niš, Niš, Serbia.
| | - Miodrag Vučić
- MD, PhD. Associate Professor, Department of Internal Medicine, Faculty of Medicine, University of Niš, Niš, Serbia; and Hematologist, Clinic of Hematology and Clinical Immunology, University Clinical Centre Niš, Niš, Serbia.
| | - Ivan Tijanić
- MD, PhD. Assistant Professor, Department of Internal Medicine, Faculty of Medicine, University of Niš, Niš, Serbia; and Hematologist, Clinic of Hematology and Clinical Immunology, University Clinical Centre Niš, Niš, Serbia.
| | - Žarko Ćojbašić
- PhD. Full Professor, Department of Mechatronics and Control, Faculty of Mechanical Engineering, University of Niš, Niš, Serbia.
| |
Collapse
|
15
|
Wang J, Arroyo-Suarez R, Dasari S, Sekaran K, Tse W. Early versus late response to daratumumab-based triplet therapies in patients with multiple myeloma: a pooled analysis of trials POLLUX, CASTOR and MAIA. Leuk Lymphoma 2022; 63:1669-1677. [PMID: 35142582 DOI: 10.1080/10428194.2022.2038376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In multiple myeloma (MM), it is unclear whether early and late responders to daratumumab have similar outcomes. We pooled individual-level data from phase 3 trials and divided them into early and late response groups based on median time to response. Altogether 670 and 213 patients achieved very good partial response (VGPR) or better and minimal residual disease (MRD) negativity, respectively. Among VGPR or better, there was no significant difference of modified progression-free survival (mPFS, hazard ratio [HR] 1.00, 95% confidence interval [CI] 0.69-1.44) or duration of response (DOR) (HR 1.02, 95%CI 0.68-1.53). Among relapsed/refractory MM (RRMM) achieving MRD negativity, late responders had significantly longer mPFS (p = 0.038) and DOR (p = 0.043). These results support that for patients who failed to achieve an early response to daratumumab, therapies should be continued with the goal of achieving ongoing and stepwise improvement of response.
Collapse
Affiliation(s)
- Jiasheng Wang
- Department of Hematology and Oncology, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Raul Arroyo-Suarez
- Department of Internal Medicine, MetroHealth Medical Center, Cleveland, OH, USA
| | - Srilatha Dasari
- Department of Internal Medicine, MetroHealth Medical Center, Cleveland, OH, USA
| | - Kanithra Sekaran
- Department of Internal Medicine, MetroHealth Medical Center, Cleveland, OH, USA
| | - William Tse
- Department of Hematology and Oncology, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
16
|
Cowan AJ, Green DJ, Kwok M, Lee S, Coffey DG, Holmberg LA, Tuazon S, Gopal AK, Libby EN. Diagnosis and Management of Multiple Myeloma: A Review. JAMA 2022; 327:464-477. [PMID: 35103762 DOI: 10.1001/jama.2022.0003] [Citation(s) in RCA: 399] [Impact Index Per Article: 199.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Multiple myeloma is a hematologic malignancy characterized by presence of abnormal clonal plasma cells in the bone marrow, with potential for uncontrolled growth causing destructive bone lesions, kidney injury, anemia, and hypercalcemia. Multiple myeloma is diagnosed in an estimated 34 920 people in the US and in approximately 588 161 people worldwide each year. OBSERVATIONS Among patients with multiple myeloma, approximately 73% have anemia, 79% have osteolytic bone disease, and 19% have acute kidney injury at the time of presentation. Evaluation of patients with possible multiple myeloma includes measurement of hemoglobin, serum creatinine, serum calcium, and serum free light chain levels; serum protein electrophoresis with immunofixation; 24-hour urine protein electrophoresis; and full-body skeletal imaging with computed tomography, positron emission tomography, or magnetic resonance imaging. The Revised International Staging System combines data from the serum biomarkers β2 microglobulin, albumin, and lactate dehydrogenase in conjunction with malignant plasma cell genomic features found on fluorescence in situ hybridization-t(4;14), del(17p), and t(14;16)-to assess estimated progression-free survival and overall survival. At diagnosis, 28% of patients are classified as having Revised International Staging stage I multiple myeloma, and these patients have a median 5-year survival of 82%. Among all patients with multiple myeloma, standard first-line (induction) therapy consists of a combination of an injectable proteasome inhibitor (ie, bortezomib), an oral immunomodulatory agent (ie, lenalidomide), and dexamethasone and is associated with median progression-free survival of 41 months, compared with historical reports of 8.5 months without therapy. This induction therapy combined with autologous hematopoietic stem cell transplantation followed by maintenance lenalidomide is standard of care for eligible patients. CONCLUSIONS AND RELEVANCE Approximately 34 920 people in the US and 155 688 people worldwide are diagnosed with multiple myeloma each year. Induction therapy with an injectable proteasome inhibitor, an oral immunomodulatory agent and dexamethasone followed by treatment with autologous hematopoietic stem cell transplantation, and maintenance therapy with lenalidomide are among the treatments considered standard care for eligible patients.
Collapse
Affiliation(s)
- Andrew J Cowan
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Seattle Cancer Care Alliance, Seattle, Washington
| | - Damian J Green
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Seattle Cancer Care Alliance, Seattle, Washington
| | - Mary Kwok
- Seattle Cancer Care Alliance, Seattle, Washington
- Division of Hematology, Department of Medicine, University of Washington, Seattle
| | - Sarah Lee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Seattle Cancer Care Alliance, Seattle, Washington
| | - David G Coffey
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, Florida
| | - Leona A Holmberg
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Seattle Cancer Care Alliance, Seattle, Washington
| | - Sherilyn Tuazon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Now with Bristol Myers Squibb, Seattle, Washington
| | - Ajay K Gopal
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Seattle Cancer Care Alliance, Seattle, Washington
| | - Edward N Libby
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Seattle Cancer Care Alliance, Seattle, Washington
| |
Collapse
|
17
|
ALCAM regulates multiple myeloma chemoresistant side population. Cell Death Dis 2022; 13:136. [PMID: 35145058 PMCID: PMC8831486 DOI: 10.1038/s41419-022-04556-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022]
Abstract
Drug-resistance is a major problem preventing a cure in patients with multiple myeloma (MM). Previously, we demonstrated that activated-leukocyte-cell-adhesion-molecule (ALCAM) is a prognostic factor in MM and inhibits EGF/EGFR-initiated MM clonogenicity. In this study, we further showed that the ALCAM-EGF/EGFR axis regulated the MM side population (SP)-mediated drug-resistance. ALCAM-knockdown MM cells displayed an enhanced ratio of SP cells in the presence of bone marrow stromal cells (BMSCs) or with the supplement of recombinant EGF. SP MM cells were resistant to chemotherapeutics melphalan or bortezomib. Drug treatment stimulated SP-genesis. Mechanistically, EGFR, primed with EGF, activated the hedgehog pathway and promoted the SP ratio; meanwhile, ALCAM inhibited EGFR downstream pro-MM cell signaling. Further, SP MM cells exhibited an increased number of mitochondria compared to the main population. Interference of the mitochondria function strongly inhibited SP-genesis. Animal studies showed that combination therapy with both an anti-MM agent and EGFR inhibitor gefitinib achieved prolonged MM-bearing mice survival. Hence, our work identifies ALCAM as a novel negative regulator of MM drug-resistance, and EGFR inhibitors may be used to improve MM therapeutic efficacy.
Collapse
|
18
|
Gogia A, Ganguly S. Triplet therapy, transplantation, and maintenance until progression in multiple myeloma. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_241_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
19
|
Lebel E, Li X, Paul H, Masih-Khan E, Bhella S, Chen C, Prica A, Reece D, Tiedemann R, Trudel S, Kukreti V. Kinetics of response to first- and second-line therapies in multiple myeloma: Assessment by both M-spikes and light chains. Eur J Haematol 2021; 108:204-211. [PMID: 34767270 DOI: 10.1111/ejh.13726] [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: 07/28/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The prognostic value of kinetics of response to multiple myeloma (MM) therapy is controversial. We aimed to expand the knowledge on this topic by reviewing the kinetics of response to both first- and second-line MM therapy, utilizing a homogeneously treated cohort and analyzing separately both M-spike and light chain (LC) responses for each patient. METHODS We reviewed all patients who received first-line cyclophosphamide, bortezomib and dexamethasone induction followed by autologous transplant with melphalan and lenalidomide maintenance in our center between 2007 and 2019. RESULTS Analyzing 360 patients, we observed no correlation between response kinetics to first- versus second-line therapy at the individual patient level. Time to best response to first-line therapy was not a predictor of outcome; however, longer time to best response was highly predictive of a favorable outcome in the second-line setting, independent of other factors. Patients with IgA-MM cleared their M-spike faster than IgG-MM, probably reflecting different half-lives of these isotypes rather than disease biology, as the clearance of LC in both subtypes was similar. CONCLUSIONS Analyzing both M-spike and LC responses in a homogenously treated cohort, we identified important insights regarding the prognostic value of kinetic patterns. Prospective analysis may shed more light on unsolved questions.
Collapse
Affiliation(s)
- Eyal Lebel
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Xuan Li
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Harminder Paul
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Esther Masih-Khan
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Sita Bhella
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Christine Chen
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Anca Prica
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Donna Reece
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Rodger Tiedemann
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Suzanne Trudel
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Vishal Kukreti
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Ho M, Zanwar S, Kapoor P, Gertz M, Lacy M, Dispenzieri A, Hayman S, Dingli D, Baudi F, Muchtar E, Leung N, Kourelis T, Warsame R, Fonder A, Hwa L, Hobbs M, Kyle R, Rajkumar SV, Kumar S. The Effect of Duration of Lenalidomide Maintenance and Outcomes of Different Salvage Regimens in Patients with Multiple Myeloma (MM). Blood Cancer J 2021; 11:158. [PMID: 34552051 PMCID: PMC8458275 DOI: 10.1038/s41408-021-00548-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/20/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022] Open
Abstract
The optimal duration of lenalidomide maintenance post-autologous stem cell transplant (ASCT) in Multiple Myeloma (MM), and choice of therapy at relapse post-maintenance, need further evaluation. This retrospective study assessed outcomes of patients with MM (n = 213) seen at Mayo Clinic, Rochester between 1/1/2005-12/31/2016 who received lenalidomide maintenance post-ASCT. The median PFS was 4 (95% CI: 3.4, 4.5) years from diagnosis of MM; median OS was not reached (5-year OS: 77%). Excluding patients who stopped lenalidomide maintenance within 3 years due to progression on maintenance, ≥3 years of maintenance had a superior 5-year OS of 100% vs. 85% in <3 years (p = 0.011). Median PFS was 7.2 (95% CI: 6, 8.5) years in ≥3 years vs. 4.4 (95% CI: 4.3, 4.5) years in <3 years (p < 0.0001). Lenalidomide refractoriness at first relapse was associated with inferior PFS2 [8.1 (95% CI: 6.4, 9.9) months vs. 19.9 (95% CI: 9.7, 30.2; p = 0.002) months in nonrefractory patients]. At first relapse post-maintenance, median PFS2 was superior with daratumumab-based regimens [18.4 (95% CI: 10.9, 25.9) months] versus regimens without daratumumab [8.9 (95% CI: 5.5, 12.3) months; p = 0.006]. Daratumumab + immunomodulatory drugs had superior median PFS2 compared to daratumumab + bortezomib [NR vs 1 yr (95% CI: 0.5, 1.5); p = 0.004].
Collapse
Affiliation(s)
- Matthew Ho
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Saurabh Zanwar
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Prashant Kapoor
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Morie Gertz
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Martha Lacy
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Angela Dispenzieri
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Suzanne Hayman
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - David Dingli
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Francis Baudi
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Eli Muchtar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nelson Leung
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Taxiarchis Kourelis
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rahma Warsame
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amie Fonder
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lisa Hwa
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Miriam Hobbs
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Robert Kyle
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - S Vincent Rajkumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shaji Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
21
|
EPIDEMIOLOGICAL LANDSCAPE OF YOUNG MULTIPLE MYELOMA PATIENTS DIAGNOSED EARLIER THAN 40 YEARS: THE FRENCH EXPERIENCE. Blood 2021; 138:2686-2695. [PMID: 34479366 DOI: 10.1182/blood.2021011285] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 07/27/2021] [Indexed: 11/20/2022] Open
Abstract
Multiple Myeloma (MM) is rare in young patients - especially before 40 years at diagnosis, representing less than 2% of all patients with MM. Little is known about the disease characteristics and prognosis of these patients. In this study we examined 214 patients diagnosed with MM ≤ 40 years old over 15 years, in the era of modern treatments. Among them, 189 patients had symptomatic MM. Disease characteristics were similar to older patients: 35% had anemia, 17% had renal impairment, and 13% hypercalcemia. The staging was ISS-1 in 52.4%, ISS-2 in 27.5% and ISS-3 in 20.1%. Overall, 18% of patients had high risk cytogenetics (del 17p and/or t(4;14)). Ninety percent of patients received intensive chemotherapy followed by autologous stem cell transplant, and 25% of patients had allogeneic stem cell transplantation predominantly at time of relapse. The median follow-up was 76 months, the estimated median overall survival was 14.5 years and the median PFS was 41 months. In multivariate analysis, bone lesions (HR=3.95; p=0.01), high ISS score (HR=2.14; p=0.03) and high-risk cytogenetics (HR=4.54; p<0.0001) were significant risk factors for poor outcomes. Among predefined time-dependent covariables, onset of progression (HR=13.2; p<0.0001) significantly shortened OS. At 5 years, Relative Survival compared to same age and sex matched individuals was 83.5%, and estimated Standardized Mortality Ratio was 69.9 (95%CI 52.7-91.1), confirming that MM dramatically shortens the survival of young patients despite an extended survival after diagnosis.
Collapse
|
22
|
Zanwar S, Kumar S. Disease heterogeneity, prognostication and the role of targeted therapy in multiple myeloma. Leuk Lymphoma 2021; 62:3087-3097. [PMID: 34304677 DOI: 10.1080/10428194.2021.1957875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma cell malignancy with a heterogeneous disease course. Insights into the genetics of the disease have identified certain high-risk cytogenetic features that are associated with adverse outcomes. While the advances in therapy have translated into dramatic improvements in the outcome of patients with MM, those with high-risk genetic features continue to perform poorly. This has resulted in a need for clinical trials focusing on the high-risk subgroup of MM as they search for additional biomarkers and therapeutic targets continue. In this review, we discuss the currently existing data on prognostic and predictive biomarkers in MM and speculate the role of treatment stratification based on the genetic features of the disease.
Collapse
Affiliation(s)
- Saurabh Zanwar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shaji Kumar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
23
|
Weisel K, Mateos MV, Gay F, Delforge M, Cook G, Szabo Z, Desgraz R, DeCosta L, Moreau P. Efficacy and safety profile of deep responders to carfilzomib-based therapy: a subgroup analysis from ASPIRE and ENDEAVOR. Leukemia 2021; 35:1732-1744. [PMID: 33067574 PMCID: PMC8179852 DOI: 10.1038/s41375-020-01049-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/30/2020] [Accepted: 09/29/2020] [Indexed: 11/13/2022]
Abstract
To understand the profile of best responders (complete response or better [≥CR]) to carfilzomib, we described the characteristics, progression-free survival (PFS), overall survival (OS) data, and the safety of patients who achieved ≥CR to carfilzomib-based treatment in ASPIRE and ENDEAVOR. In post hoc analyses from ASPIRE and ENDEAVOR, median PFS and OS were longer for ≥CR patients versus those who achieved a very good partial response or partial response (VGPR/PR). In the carfilzomib arm of ASPIRE, median PFS was 50.4 months for ≥CR versus 22.1 months for VGPR/PR; median OS was 67.0 versus 44.2 months, respectively. In the carfilzomib arm of ENDEAVOR, median PFS was 34.0 for ≥CR versus 20.4 months for VGPR/PR; median OS was non-estimable. Despite the longer treatment duration, fewer patients with ≥CR versus VGPR/PR experienced treatment-emergent adverse events that led to discontinuation of carfilzomib-based treatment in ASPIRE or ENDEAVOR. Low serum lactate dehydrogenase was the only factor associated with achieving ≥CR vs patients not achieving CR in ASPIRE in multivariate regression analyses. No association was found between cytogenetic risk status and reaching ≥CR. Carfilzomib treatment may lead to rapid and deep responses, irrespective of most patient characteristics.
Collapse
Affiliation(s)
- Katja Weisel
- Department of Oncology and Hematology, University Medical Center of Hamburg-Eppendorf, Hamburg, Germany.
| | - Maria-Victoria Mateos
- Institute of Biomedical Research of Salamanca (IBSAL), Cancer Research Center-IBMCC (USAL-CSIC), and Hematology Department, University Hospital of Salamanca, Salamanca, Spain
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Michel Delforge
- Department of Hematology, University Hospital (UZ) Leuven, Leuven, Belgium
| | - Gordon Cook
- Department of Haematology, Leeds Cancer Centre, St James's University Hospital, Leeds, UK
| | | | | | - Lucy DeCosta
- Global Biostatistical Science, Amgen Ltd, Cambridge, UK
| | - Philippe Moreau
- Hematology Department, University Hospital Hôtel-Dieu, Nantes, France
| |
Collapse
|
24
|
Cherniawsky HM, Kukreti V, Reece D, Masih-Khan E, McCurdy A, Jimenez-Zepeda VH, Sebag M, Song K, White D, Stakiw J, LeBlanc R, Reiman A, Aslam M, Louzada M, Kotb R, Gul E, Atenafu E, Venner CP. The survival impact of maintenance lenalidomide: an analysis of real-world data from the Canadian Myeloma Research Group national database. Haematologica 2021; 106:1733-1736. [PMID: 33054120 PMCID: PMC8168484 DOI: 10.3324/haematol.2020.259093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
| | - Vishal Kukreti
- Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON
| | - Donna Reece
- Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON
| | - Esther Masih-Khan
- Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada; Canadian Myeloma Research Group, Toronto, Ontario.
| | | | | | - Michael Sebag
- Department of Oncology, Division of Hematology, McGill University, Montreal, QC
| | - Kevin Song
- BC Cancer, Vancouver General Hospital, British Columbia
| | - Darrell White
- Queen Elizabeth II Health Sciences Centre. Dalhousie University, Halifax, Nova Scotia
| | - Julie Stakiw
- Saskatoon Cancer Centre, University of Saskatchewan, Saskatoon, Saskatchewan
| | - Richard LeBlanc
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec
| | - Anthony Reiman
- Department of Oncology, Saint John Regional Hospital, Saint John, NB
| | | | | | - Rami Kotb
- Cancer Care Manitoba, Winnipeg, Manitoba
| | - Engin Gul
- Canadian Myeloma Research Group, Toronto, Ontario
| | | | | |
Collapse
|
25
|
Morris TCM, Drake MB, Kettle PJ, McGuigan T, Leahy M, O’Dwyer M, Enright H, O’Shea T, Popat R, Oakervee HE, Yong K, Cavenagh JD, Cairns DA, Alvarez-Iglesias A, Cook G. How to Simplify the Evaluation of Newly Introduced Chemotherapeutic Interventions in Myeloma. Clin Hematol Int 2021; 3:27-33. [PMID: 34595464 PMCID: PMC8432324 DOI: 10.2991/chi.k.210201.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/26/2021] [Indexed: 11/30/2022] Open
Abstract
When the bortezomib [PS341], adriamycin and dexamethasone (PAD) regimen was first evaluated, the response rate in untreated patients was much superior to that elicited by conventional chemotherapeutic agents. We demonstrated the efficacy of PAD in relapsed or refractory patients by comparing the response rate obtained in 53 patients who received vincristine, adriamycin and dexamethasone (VAD) or equivalent regimen as induction therapy, using a comparative design in which each patient acted as their own control. Whereas 25 patients had a positive response to VAD, 37 patients had a response to PAD ≤ partial remission (PR) (p = 0.023). Using the more stringent response level of very good PR (VGPR) the results favored the PAD regimen very significantly (p = 0.006) (McNemars test). Similar results were seen using paired M-protein levels from individual patient comparisons. As the PAD regimen was subsequently adopted as the re-induction therapy in the British Society for Blood and Marrow Transplantation/United Kingdom Myeloma Forum Myeloma X (Intensive) trial, now concluded, we have retrospectively analyzed the findings from both studies. Comparison of response rates and adverse effects of patients having had previous autologous transplantation (Cohort 1) with the corresponding data from Myeloma X showed close correlation. These findings provide evidence that rapid results may be obtained in the evaluation of newly introduced, and potentially highly effective, anti-tumour agents by direct comparison to the response to the immediately preceding standard regimen, particularly in relatively resistant tumours.
Collapse
Affiliation(s)
| | - Mary B. Drake
- Department of Haematology, Belfast City Hospital, Lisburn Road, Belfast BT9 7AD, UK
| | - Paul J. Kettle
- Department of Haematology, Belfast City Hospital, Lisburn Road, Belfast BT9 7AD, UK
| | - Tracey McGuigan
- Department of Haematology, Belfast City Hospital, Lisburn Road, Belfast BT9 7AD, UK
| | - Maeve Leahy
- Department of Haematology, University Hospital Limerick, Limerick, Ireland
| | - Michael O’Dwyer
- Department of Haematology, University Hospital Galway, Newcastle Road, Galway, Ireland
| | - Helen Enright
- Department of Haematology, Tallaght Hospital, Dublin 24, Ireland
| | - Tanya O’Shea
- Clinical Research Consulting Ltd., 16 College Square, Terenure, Dublin D6WF309, Ireland
| | - Rakesh Popat
- Department of Haematology, University College Hospital, London, UK
| | - Heather E. Oakervee
- Department of Haematology, St Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
| | - Kwee Yong
- Department of Haematology, University College Hospital, London, UK
| | - Jamie D. Cavenagh
- Department of Haematology, St Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
| | - David A. Cairns
- Leeds Cancer Centre, St James’s University Hospital, Leeds LS9 7TF, UK
| | | | - Gordon Cook
- Leeds Cancer Centre, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
26
|
Fais S, Marunaka Y. The Acidic Microenvironment: Is It a Phenotype of All Cancers? A Focus on Multiple Myeloma and Some Analogies with Diabetes Mellitus. Cancers (Basel) 2020; 12:cancers12113226. [PMID: 33147695 PMCID: PMC7693643 DOI: 10.3390/cancers12113226] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is a hematological malignancy characterized by an abnormal clone of plasma cells in the bone marrow. Currently, both the disease progression and its therapy are too often followed by a series of complications and the standard treatment for MM is aimed at improving the quality of life and to prolong progression-free survival (PFS), and overall survival (OS) of patients. This review looks at MM therapies from a different sight. It starts from a clear scientific background, suggesting that many malignant tumors have some common phenotype that isolates the tumor from the rest of the body. Between these phenotypes, extracellular acidity exerts a key role and data collected in the last two decades support the use of anti-acidic drugs in the treatment of cancers, including MM. Lastly, as many cancers, MM has some similarities with type II diabetes mellitus (DM) in the mechanism leading to the extracellular acidity, supporting the use of both a wide panel of proton transporters inhibitors and probably also of some anti-DM drugs in the treatment of both MM and DM. Abstract Multiple myeloma (MM) is a hematological malignancy with a poor prognosis while with a long and progressive outcome. To date, the therapeutic options are restricted to few drugs, including thalidomide or its derivates and autologous transplantation including stem-cell transplantation. More recently, the use of both proteasome inhibitors and monoclonal antibodies have been included in MM therapy, but the clinical results are still under evaluation. Unfortunately, death rates (within the 5-year overall survival rates) are still very high (45%), with no relevant improvement over the past 10 years. Here, we discuss data supporting a new therapeutic approach against MM, based on a common phenotype of tumor malignancies, which is the acidic microenvironment. Extracellular acidity drastically reduces the efficacy of both anti-tumor drugs and the immune reaction against tumors. Pre-clinical data have shown that anti-acidic drugs, such as proton pump inhibitors (PPIs), have a potent cytotoxic effect against human MM cells, thus supporting their use in the treatment of this malignancy. Here, we discuss also similarities between MM and type II diabetes mellitus (DM) with high risk of developing MM, suggesting that both anti-diabetic drugs and a hypocaloric diet may help in curing MM patients.
Collapse
Affiliation(s)
- Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), 00161 Rome, Italy
- Correspondence: (S.F.); (Y.M.); Tel.: +39-06-49903195 (S.F.); +81-75-802-0135 (Y.M.)
| | - Yoshinori Marunaka
- Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto 604-8472, Japan
- Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu 525-8577, Japan
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto 602-8566, Japan
- Correspondence: (S.F.); (Y.M.); Tel.: +39-06-49903195 (S.F.); +81-75-802-0135 (Y.M.)
| |
Collapse
|
27
|
Gozzetti A, Raspadori D, Bacchiarri F, Sicuranza A, Pacelli P, Ferrigno I, Tocci D, Bocchia M. Minimal Residual Disease in Multiple Myeloma: State of the Art and Applications in Clinical Practice. J Pers Med 2020; 10:jpm10030120. [PMID: 32927719 PMCID: PMC7565263 DOI: 10.3390/jpm10030120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Novel drugs have revolutionized multiple myeloma therapy in the last 20 years, with median survival that has doubled to up to 8–10 years. The introduction of therapeutic strategies, such as consolidation and maintenance after autologous stem cell transplants, has also ameliorated clinical results. The goal of modern therapies is becoming not only complete remission, but also the deepest possible remission. In this context, the evaluation of minimal residual disease by techniques such as next-generation sequencing (NGS) and next-generation flow (NGF) is becoming part of all new clinical trials that test drug efficacy. This review focuses on minimal residual disease approaches in clinical trials, with particular attention to real-world practices.
Collapse
|
28
|
The impact of response kinetics for multiple myeloma in the era of novel agents. Blood Adv 2020; 3:2895-2904. [PMID: 31594763 DOI: 10.1182/bloodadvances.2019000432] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 07/01/2019] [Indexed: 01/08/2023] Open
Abstract
Rapid remission by induction therapy has long been recognized as an important predictor for long-time survival in acute leukemia. However, the impact of response kinetics on multiple myeloma (MM) seems to be different and remains unexplored. The relationship between response kinetics and outcome were assessed in 626 patients with newly diagnosed MM who were included in a prospective, nonrandomized clinical trial (BDH 2008/02). Patients were assigned to either immunomodulatory drug- or proteasome inhibitor-based therapy. The response depth, time to best response (TBR) and duration of best response (DBR) were collected. Depth of response was associated with superior outcomes, consistent with findings from other studies. However, the early responders (defined as TBR ≤3 months) showed significantly worse survival compared with late responders. We found that patients with rapid complete remission experienced inferior survivals comparable to those attaining a gradual partial remission. Moreover, 4 distinct response kinetics patterns were identified. Patients with gradual and sustained remission ("U-valley" pattern) experienced superior outcomes, whereas poor outcomes were observed in rapid and transient responders ("roller coaster" pattern) (median overall survival, 126 vs 30 months). The effects of response patterns on survival were confirmed in patients at different stages of disease and cytogenetic risk, including transplant-eligible patients and those attaining different extents of response depth. Collectively, our data indicated that slow and gradual response is a favorable prognostic factor in MM. In addition to response depth, the kinetic pattern of response is a simple and powerful predictor for survival even in the era of novel agents.
Collapse
|
29
|
Li Q, Lin J, Chi A, Davies S. Practical considerations of utilizing propensity score methods in clinical development using real-world and historical data. Contemp Clin Trials 2020; 97:106123. [PMID: 32853779 DOI: 10.1016/j.cct.2020.106123] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 11/16/2022]
Abstract
In recent years, with the rapid increase in the volume and accessibility of Real-World-Data (RWD) and Real-World-Evidence (RWE), we have seen the unprecedented opportunities for their use in drug clinical development and life-cycle management. RWD and RWE have demonstrated the significant potential to improve the design, planning, and execution of clinical development. Furthermore, they can feature in the designs as either a substitute or compliment to traditional clinical trials. However, to utilize RWD and RWE appropriately and wisely, it is critical to apply rigorous statistical methodologies that enable the robustness of results to be characterized and ascertained. Several statistical methodologies including exact matching, propensity score methods, matching-adjusted indirect comparisons and meta-analysis have been proposed for analyzing RWD. Among them, propensity score method is one of the most commonly used methods for non-randomized trials with indirect comparison. Although massive methodologies and examples have been published and discussed since propensity score methods were introduced, systematic review and discussion of how to rigorously use propensity score methods in the practical clinical development is still deficient. This paper introduces commonly used and emerging propensity score methods with detailed discussions of their pros and cons. Three different case studies are presented to illustrate the practical considerations of utilizing propensity score methods in the study design and evaluation using real-world and historical data. Additional considerations including selection of patient populations, endpoints, baseline covariates, propensity score methods, sensitivity analysis and practical implementation flow in clinical development will be discussed.
Collapse
Affiliation(s)
- Qing Li
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, 300 Massachusetts Ave, Cambridge, MA 02139, United States.
| | - Jianchang Lin
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, 300 Massachusetts Ave, Cambridge, MA 02139, United States
| | - Andy Chi
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, 300 Massachusetts Ave, Cambridge, MA 02139, United States
| | - Simon Davies
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, 300 Massachusetts Ave, Cambridge, MA 02139, United States
| |
Collapse
|
30
|
Hansen E, Read AF. Cancer therapy: Attempt cure or manage drug resistance? Evol Appl 2020; 13:1660-1672. [PMID: 32821276 PMCID: PMC7428817 DOI: 10.1111/eva.12994] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/12/2020] [Accepted: 03/14/2020] [Indexed: 12/25/2022] Open
Abstract
Cancer treatment is often aimed at achieving rapid, large, and sustained reductions in tumor burden. Even when these strong responses are achieved, treatment frequently fails due to the emergence of drug-resistant cell lineages. Over the last decade, a variety of authors have suggested that treatment should instead be aimed at containing resistance rather than curing the patient. That new philosophy poses a dilemma: how to choose between treatment regimens that can sometimes cure the patient and regimens that can delay progression but not cure the patient? Here, we investigate that choice. We define aspects of the evolution and ecology of tumor dynamics that determine whether it is better to attempt cure or to manage resistance. Even when it is possible to manage resistance and delay progression, this may not be the best treatment option. We show that the best option depends on how "cure" and "delaying progression" are prioritized, and how those priorities will vary among patients. We also discuss the difficulties of comparing in clinical trials traditional strategies that can sometimes successfully cure to alternative approaches where cure is not possible. More generally, where resistance management is possible, there are new challenges in communicating options to patients, setting treatment guidelines, and evaluating data from clinical trials.
Collapse
Affiliation(s)
- Elsa Hansen
- Department of BiologyCenter for Infectious Disease DynamicsPennsylvania State UniversityUniversity ParkPAUSA
| | - Andrew F. Read
- Department of BiologyCenter for Infectious Disease DynamicsPennsylvania State UniversityUniversity ParkPAUSA
- Department of EntomologyPennsylvania State UniversityUniversity ParkPAUSA
- Huck Institutes of the Life SciencesPennsylvania State UniversityUniversity ParkPAUSA
| |
Collapse
|
31
|
Quinn KM, Kartikasari AER, Cooke RE, Koldej RM, Ritchie DS, Plebanski M. Impact of age-, cancer-, and treatment-driven inflammation on T cell function and immunotherapy. J Leukoc Biol 2020; 108:953-965. [PMID: 32678927 DOI: 10.1002/jlb.5mr0520-466r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/16/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
Many cancers are predominantly diagnosed in older individuals and chronic inflammation has a major impact on the overall health and immune function of older cancer patients. Chronic inflammation is a feature of aging, it can accelerate disease in many cancers and it is often exacerbated during conventional treatments for cancer. This review will provide an overview of the factors that lead to increased inflammation in older individuals and/or individuals with cancer, as well as those that result from conventional treatments for cancer, using ovarian cancer (OC) and multiple myeloma (MM) as key examples. We will also consider the impact of chronic inflammation on immune function, with a particular focus on T cells as they are key targets for novel cancer immunotherapies. Overall, this review aims to highlight specific pathways for potential interventions that may be able to mitigate the impact of chronic inflammation in older cancer patients.
Collapse
Affiliation(s)
- Kylie M Quinn
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia.,Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | | | - Rachel E Cooke
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Rachel M Koldej
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - David S Ritchie
- Australian Cancer Research Foundation (ACRF) Translational Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| |
Collapse
|
32
|
Montefusco V, Corso A, Galli M, Ardoino I, Pezzatti S, Carniti C, Patriarca F, Gherlinzoni F, Zambello R, Sammassimo S, Marcatti M, Nozza A, Crippa C, Cafro AM, Baldini L, Corradini P. Bortezomib, cyclophosphamide, dexamethasone
versus
lenalidomide, cyclophosphamide, dexamethasone in multiple myeloma patients at first relapse. Br J Haematol 2020; 188:907-917. [DOI: 10.1111/bjh.16287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/09/2019] [Indexed: 11/30/2022]
Affiliation(s)
| | - Alessandro Corso
- Division of Hematology Fondazione IRCCS Policlinico San Matteo Università di Pavia PaviaItaly
| | - Monica Galli
- Hematology Papa Giovanni XXIII hospital BergamoItaly
| | - Ilaria Ardoino
- Istituto di Ricerche Farmacologiche “Mario Negri” – IRCCS MilanItaly
| | | | - Cristina Carniti
- Hematology Fondazione IRCCS Istituto Nazionale dei Tumori MilanItaly
| | | | | | - Renato Zambello
- Department of Medicine, Hematology and Clinical Immunology Branch Padua University School of Medicine PaduaItaly
| | | | - Magda Marcatti
- Hematology and Bone Marrow Transplantation Unit IRCCS San Raffaele Scientific Institute MilanItaly
| | - Andrea Nozza
- Oncology and Hematology Department Istituto Clinico Humanitas, Rozzano (MI) MilanItaly
| | | | - Anna Maria Cafro
- Department of Oncology/Hematology Niguarda Ca' Granda Hospital MilanItaly
| | - Luca Baldini
- Hematology/Bone Marrow Transplantation Unit Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico Ca'Granda Ospedale Maggiore PoliclinicoUniversity of Milan MilanItaly
| | - Paolo Corradini
- Hematology Fondazione IRCCS Istituto Nazionale dei Tumori MilanItaly
- Department of Oncology and Hematology University of Milan Milan Italy
| |
Collapse
|
33
|
Mitrović M, Sretenović A, Bila J. The significance of prognostic profiling in the treatment of patients with multiple myeloma. MEDICINSKI PODMLADAK 2020. [DOI: 10.5937/mp71-28137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Multiple myeloma (MM) is a hematological neoplasia characterized by clonal expansion of the most mature B lymphocytes, plasma cells (Plc), in the bone marrow (BM). Due to new treatment modalities, the 5-years survival has improved in the last 15 years, and nowdays ranges from 60-70%. The individual prognostic profile is based on the biological characteristics of the disease, clinical characteristics of patients and therapeutics response characteristics. The biological characteristics of the disease are defined by the clinical stage according to the Durie-Salmon classification and prognostic indices such the international staging system (ISS) and revised ISS (R-ISS). Numerous different mutations of prognostic significance have been discovered applying methods of molecular genetics such as next generation sequencing (NGS). The age and comorbity status of patients are the most important clinical characteristics because they are crucial for therapeutic choice and suitability for treatment with high-dose chemotherapy and autologous stem cell transplantation. The prognostic influence of therapeutic response is very important and the most powerful prognostic factors are achievements of complete remission (CR) and minimal residual disease (MRD) negativity.
Collapse
|
34
|
Petrucci MT, Vozella F. The Anti-CD38 Antibody Therapy in Multiple Myeloma. Cells 2019; 8:E1629. [PMID: 31842517 PMCID: PMC6952883 DOI: 10.3390/cells8121629] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/04/2019] [Accepted: 12/09/2019] [Indexed: 01/08/2023] Open
Abstract
Multiple myeloma (MM) is the second-most common hematologic malignancy after diffuse large B-cell lymphoma. Despite the improvement in response and survival rates following the introduction of novel therapies, only a few patients are cured, and the majority of MM patients experience several relapses and receive multiple lines of treatment. Currently, bortezomib and lenalidomide are the core component of treatment both at the time of diagnosis and at the relapse as well as the new proteasome inhibitors (PIs), such as carfilzomib and ixazomib, and the next-generation immunomodulatory drug, pomalidomide, are now available for patients in relapse. In addition, drugs with a different mechanism of action, such as the histone deacetylase inhibitor and the monoclonal antibodies (MoAb) targeting SLAMF7 or CD38, are a part of the anti-myeloma armamentarium and are very important for heavily pretreated or double refractory to a PI and IMiD patients. In this paper, we focus on the efficacy as well as toxicities of CD38 antibodies used both as a single agent and in combination as multiple myeloma treatment.
Collapse
Affiliation(s)
- Maria Teresa Petrucci
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto I, Sapienza University of Rome, Via Benevento 6, 00161 Rome, Italy;
| | | |
Collapse
|
35
|
Samala RV, Valent J, Noche N, Lagman R. Palliative Care in Patients With Multiple Myeloma. J Pain Symptom Manage 2019; 58:1113-1118. [PMID: 31326505 DOI: 10.1016/j.jpainsymman.2019.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Renato V Samala
- Palliative Medicine Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| | - Jason Valent
- Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Natalee Noche
- Palliative Medicine Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ruth Lagman
- Palliative Medicine Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
36
|
Case of Stroke from Cerebral Vasculitis following Carfilzomib, Lenalidomide, and Dexamethasone Therapy in a Patient with Relapsing Multiple Myeloma. Case Rep Hematol 2019; 2019:5180424. [PMID: 31885953 PMCID: PMC6907054 DOI: 10.1155/2019/5180424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/22/2019] [Accepted: 11/05/2019] [Indexed: 12/24/2022] Open
Abstract
Lenalidomide, a synthetic derivation of thalidomide, in recent years, has been the backbone of multiple myeloma treatment leading to improved survival. Common adverse effects from lenalidomide-based regimens include hypertension, heart disease, and venous thromboembolism. Hence, thromboprophylaxis is recommended to reduce the risk of stroke. We report a case of stroke from cerebral vasculitis in a patient receiving carfilzomib, lenalidomide, and dexamethasone for relapsing multiple myeloma, not previously published. Medical oncologists should be aware of other causes of stroke among multiple myeloma patients receiving a lenalidomide-based regimen to prevent its occurrence.
Collapse
|
37
|
Rosiñol L, Oriol A, Rios R, Sureda A, Blanchard MJ, Hernández MT, Martínez-Martínez R, Moraleda JM, Jarque I, Bargay J, Gironella M, de Arriba F, Palomera L, González-Montes Y, Martí JM, Krsnik I, Arguiñano JM, González ME, González AP, Casado LF, López-Anglada L, Paiva B, Mateos MV, San Miguel JF, Lahuerta JJ, Bladé J. Bortezomib, lenalidomide, and dexamethasone as induction therapy prior to autologous transplant in multiple myeloma. Blood 2019; 134:1337-1345. [PMID: 31484647 PMCID: PMC6888142 DOI: 10.1182/blood.2019000241] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 08/02/2019] [Indexed: 12/27/2022] Open
Abstract
Achieving and maintaining a high-quality response is the treatment goal for patients with newly diagnosed multiple myeloma (NDMM). The phase 3 PETHEMA/GEM2012 study, in 458 patients aged ≤65 years with NDMM, is evaluating bortezomib (subcutaneous) + lenalidomide + dexamethasone (VRD) for 6 cycles followed by autologous stem cell transplant (ASCT) conditioned with IV busulfan + melphalan vs melphalan and posttransplant consolidation with 2 cycles of VRD. We present grouped response analysis of induction, transplant, and consolidation. Responses deepened over time; in patients who initiated cycle 6 of induction (n = 426), the rates of a very good partial response or better were 55.6% by cycle 3, 63.8% by cycle 4, 68.3% by cycle 5, and 70.4% after induction. The complete response rate of 33.4% after induction in the intent-to-treat (ITT) population, which was similar in the 92 patients with high-risk cytogenetics (34.8%), also deepened with further treatment (44.1% after ASCT and 50.2% after consolidation). Rates of undetectable minimal residual disease (median 3 × 10-6 sensitivity) in the ITT population also increased from induction (28.8%) to transplant (42.1%) and consolidation (45.2%). The most common grade ≥3 treatment-emergent adverse events during induction were neutropenia (12.9%) and infection (9.2%). Grade ≥2 peripheral neuropathy (grouped term) during induction was 17.0%, with a low frequency of grade 3 (3.7%) and grade 4 (0.2%) events. VRD is an effective and well-tolerated regimen for induction in NDMM with deepening response throughout induction and over the course of treatment. This trial was registered at www.clinicaltrials.gov as #NCT01916252 and EudraCT as #2012-005683-10.
Collapse
Affiliation(s)
- Laura Rosiñol
- Hospital Clínic, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Albert Oriol
- Institut Català d'Oncologia I Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Rafael Rios
- Hospital Virgen de las Nieves, Granada, Spain
| | - Anna Sureda
- Institut Català d'Oncologia-Hospitalet, Barcelona, Spain
| | | | | | | | - Jose M Moraleda
- Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | - Juan Bargay
- Hospital Son Llatzer, Palma de Mallorca, Spain
| | | | - Felipe de Arriba
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, IMIB-Arrixaca, Universidad de Murcia, Murcia, Spain
| | | | | | | | | | | | | | | | | | | | - Bruno Paiva
- Clínica Universidad de Navarra, CIMA, CIBERONC, IDISNA, Pamplona, Spain; and
| | - Maria-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Jesus F San Miguel
- Clínica Universidad de Navarra, CIMA, CIBERONC, IDISNA, Pamplona, Spain; and
| | | | - Joan Bladé
- Hospital Clínic, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
38
|
Liu YT, Zhang K, Li CC, Hu XS, Jiang J, Hao XZ, Wang Y, Li JL, Xing PY, Yang S, Zhang X, Wang GQ, Cai SL, Shi YK. Depth of Response was Associated with Progression-Free Survival in Patients with Advanced Non-small Cell Lung Cancer treated with EGFR-TKI. J Cancer 2019; 10:5108-5113. [PMID: 31602263 PMCID: PMC6775600 DOI: 10.7150/jca.33450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/28/2019] [Indexed: 11/05/2022] Open
Abstract
Background: Response Evaluation Criteria in Solid Tumors (RECIST) has been widely utilized to evaluate new therapeutic strategies in cancer. However, RECIST fails to assess the heterogeneity of response in highly active therapies. Depth of response (DepOR), defined as the maximum percentage change in tumor size compared with baseline, may provide a new strategy to evaluate disease response. In the present study, we studied the association between DepOR and progression-free survival (PFS) in patients with advanced non-small cell lung cancer (NSCLC) treated with epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). Methods: Advanced NSCLC patients harboring EGFR driver mutation (L858R or exon 19 deletion) treated with EGFR-TKI from August 2014 to July 2017 from two sites were retrospetively collected for analysis. Patients were divided into four groups by DepOR (Q1 = 1-25%, Q2 = 26-50%, Q3 = 51-75%, Q4 = 76-100%). Kaplan-Meier curves were plotted for PFS against DepOR and the hazard ratio (HR) was determined through univariable and multivariable cox regression models. Results: In total, 265 patients were included for analysis. The number of patients in Group Q1-Q4 were 91 (34.3%), 73 (27.5%), 65 (24.5%) and 36 (13.6%), respectively. A greater DepOR was significantly associated with a longer PFS (Log-rank P<0.0001). The HRs (95% CI) for PFS comparing patients with different DepOR status were 0.58 (0.42-0.80) for Q2, 0.49 (0.35-0.69) for Q3, and 0.33 (0.22-0.50) for Q4, all compared with patients in Q1. DepOR as a continuous variable was also associated with prolonged PFS (HR, 0.20; 95% CI, 0.13-0.33; P<0.001). Additionally, in the multivariable cox regression model, abnormal LDH, brain metastasis and male were found to be associated with worse PFS outcomes (P<0.05). Conclusion: A greater DepOR is significantly associated with PFS benefit in advanced NSCLC treated with EGFR-TKI, suggesting that it may be a useful clinical outcome to evaluate the response of targeted therapy.
Collapse
Affiliation(s)
- Yu-Tao Liu
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Kai Zhang
- Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, P.R. China
| | - Cheng-Cheng Li
- The Medical Department, 3D Medicines Inc, Shanghai, P.R. China
| | - Xing-Sheng Hu
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Jun Jiang
- Department of radiology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Xue-Zhi Hao
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Yan Wang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Jun-Ling Li
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Pu-Yuan Xing
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Sheng Yang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Xin Zhang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Guo-Qiang Wang
- The Medical Department, 3D Medicines Inc, Shanghai, P.R. China
| | - Shang-Li Cai
- The Medical Department, 3D Medicines Inc, Shanghai, P.R. China
| | - Yuan-Kai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/ National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
39
|
Avet-Loiseau H, Ludwig H, Landgren O, Paiva B, Morris C, Yang H, Zhou K, Ro S, Mateos MV. Minimal Residual Disease Status as a Surrogate Endpoint for Progression-free Survival in Newly Diagnosed Multiple Myeloma Studies: A Meta-analysis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 20:e30-e37. [PMID: 31780415 DOI: 10.1016/j.clml.2019.09.622] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/05/2019] [Accepted: 09/29/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Therapeutic advances have greatly extended survival times in patients with multiple myeloma, necessitating increasingly lengthy trials when using survival outcomes as primary endpoints. A surrogate endpoint that can more rapidly predict survival could accelerate drug development. We conducted a meta-analysis to evaluate minimal residual disease (MRD) status as a valid progression-free survival (PFS) surrogate in patients with newly diagnosed multiple myeloma (NDMM). MATERIALS AND METHODS We searched abstracts in PubMed, The American Society of Hematology, and the European Hematology Association for "myeloma," "minimal residual disease," and "clinical trial." Because of the need to evaluate the treatment effect on MRD response, only randomized studies for subjects with NDMM were included. Details on the MRD-tested populations were required. The meta-analysis was performed by principles outlined at the 2013 United States Food and Drug Administration workshop on MRD in acute myeloid leukemia.42 For samples that were not measured for MRD and within the subset specified for MRD assessment, their MRD status was imputed from the samples that had known MRD status. Patients that were excluded from planned MRD assessment were considered MRD-positive. RESULTS Six randomized studies, representing 3283 patients and 2208 MRD samples, met analysis inclusion criteria. MRD negativity rates ranged from 0.06 to 0.70. The treatment effect on the odds ratio for MRD-negative response strongly correlated with the hazard ratio for PFS with a coefficient of determination for the weighted regression line of 0.97. Our meta-analysis suggested that MRD status met both the Prentice criteria for PFS surrogacy. CONCLUSIONS These results support the claim that MRD status can be used as a surrogate for PFS in NDMM.
Collapse
Affiliation(s)
- Hervé Avet-Loiseau
- Unité de Génomique du Myélome, University of Toulouse, Toulouse, France.
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Wilhelminenspital, Vienna, Austria
| | - Ola Landgren
- Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - Chris Morris
- Global Biostatistical Science, Amgen, Inc, Thousand Oaks, CA
| | - Hui Yang
- Global Biostatistical Science, Amgen, Inc, Thousand Oaks, CA
| | - Kefei Zhou
- Global Biostatistical Science, Amgen, Inc, Thousand Oaks, CA
| | - Sunhee Ro
- Global Biostatistical Science, Amgen, Inc, Thousand Oaks, CA
| | | |
Collapse
|
40
|
Zhang Y, Xiong X, Fu Z, Dai H, Yao F, Liu D, Deng S, Hu C. Whole-body diffusion-weighted MRI for evaluation of response in multiple myeloma patients following bortezomib-based therapy: A large single-center cohort study. Eur J Radiol 2019; 120:108695. [PMID: 31589995 DOI: 10.1016/j.ejrad.2019.108695] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/14/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE To determine the feasibility of whole-body diffusion-weighted imaging (WB-DWI) MRI for evaluation of response in patients with multiple myeloma (MM) following bortezomib-based therapy and to explore the direction of apparent diffusion coefficient (ADC) changes upon treatment. METHOD Seventy-two MM patients who underwent WB-DWI MRI before and after bortezomib-based chemotherapy (21 weeks) were evaluated retrospectively. The estimated tumor volume (eTV) and ADCmean values before and after chemotherapy were calculated and compared between deep and non-deep responders. Predictive value of baseline ADCmean was calculated to predict the trend of ADCmean change following treatment. RESULTS Fifty-five patients were classified as deep responders, and 17 cases were assigned as non-deep responders. For 327 focal lesions (FLs), the ADCmean value was significantly increased from baseline to post-treatment. However, the ADCmean value was significantly decreased following treatment in 846 representative diffuse lesions. Diffuse lesions showed a significantly decreased ADCmean value in deep responders, whereas no significant variation in ADCmean value in FLs was found between deep and non-deep responders. Baseline ADCmean at a specific value (0.808 × 10-3 mm2/s) yielded a maximum specificity (68.05%) and sensitivity (54.09%) in predicting increase of post-treatment ADCmean. CONCLUSIONS The ADCmean value was significantly decreased in MM patients with diffuse pattern, while it was significantly increased in those with focal pattern following bortezomib-based treatment. WB-DWI MRI could be used to discriminate deep response to induction treatment in MM patients with diffuse infiltration pattern. Baseline ADCmean value might have a potential to predict the trend of ADCmean change following treatment.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Xing Xiong
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zhengzheng Fu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Hui Dai
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Feirong Yao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Dong Liu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
41
|
Gertz MA. Management of induction failures in newly diagnosed transplant-eligible multiple myeloma. Leuk Lymphoma 2019; 61:1-3. [PMID: 31502884 DOI: 10.1080/10428194.2019.1663424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
42
|
Lewis KD, Larkin J, Ribas A, Flaherty KT, McArthur GA, Ascierto PA, Dréno B, Yan Y, Wongchenko M, McKenna E, Zhu Q, Mun Y, Hauschild A. Impact of depth of response on survival in patients treated with cobimetinib ± vemurafenib: pooled analysis of BRIM-2, BRIM-3, BRIM-7 and coBRIM. Br J Cancer 2019; 121:522-528. [PMID: 31417188 PMCID: PMC6889491 DOI: 10.1038/s41416-019-0546-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This pooled analysis investigated the prognostic value of depth of response in two cohorts of patients with BRAFV600-mutated metastatic melanoma treated with vemurafenib or cobimetinib plus vemurafenib. METHODS The data were pooled from BRIM-2, BRIM-3, BRIM-7 and coBRIM. Association of depth of response with survival was estimated by Cox proportional hazards regression, adjusted for clinically relevant covariates. Depth of response was analysed in previously identified prognostic subgroups based on disease characteristics and gene signatures. RESULTS Greater tumour reduction and longer time to maximal response were significantly associated with longer progression-free survival (PFS) and overall survival (OS) when evaluated as continuous variables. Patients with the deepest responses had long-lasting survival outcomes (median PFS: 14 months; OS: 32 months with vemurafenib; not estimable with cobimetinib plus vemurafenib). Cobimetinib plus vemurafenib improved depth of response versus vemurafenib monotherapy regardless of other prognostic factors, including gene signatures. CONCLUSIONS Greater depth of response was associated with improved survival, supporting its utility as a measure of treatment efficacy in melanoma and further evaluation of its incorporation into existing prognostic models. Cobimetinib plus vemurafenib improved outcomes across quartiles of response regardless of prognostic factors or gene signatures and provided durable survival benefits in patients with deep responses.
Collapse
Affiliation(s)
- Karl D Lewis
- Department of Medicine, University of Colorado Comprehensive Cancer Center, Aurora, CO, 80045, USA.
| | - James Larkin
- Skin Unit, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Antoni Ribas
- Departments of Medicine and Hematology and Oncology, Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Keith T Flaherty
- Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Grant A McArthur
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Department of Oncology, University of Melbourne, Parkville, VIC, 3000, Australia
| | - Paolo A Ascierto
- Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, 80131, Italy
| | - Brigitte Dréno
- Department of Oncology, Nantes University, Nantes, 44093, France
| | - Yibing Yan
- Product Development Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Matthew Wongchenko
- Product Development Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Edward McKenna
- Medical Affairs, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Qian Zhu
- Product Development Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Yong Mun
- Product Development Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, D-24105, Germany
| |
Collapse
|
43
|
Jones JR, Weinhold N, Ashby C, Walker BA, Wardell C, Pawlyn C, Rasche L, Melchor L, Cairns DA, Gregory WM, Johnson D, Begum DB, Ellis S, Sherborne AL, Cook G, Kaiser MF, Drayson MT, Owen RG, Jackson GH, Davies FE, Greaves M, Morgan GJ. Clonal evolution in myeloma: the impact of maintenance lenalidomide and depth of response on the genetics and sub-clonal structure of relapsed disease in uniformly treated newly diagnosed patients. Haematologica 2019; 104:1440-1450. [PMID: 30733268 PMCID: PMC6601103 DOI: 10.3324/haematol.2018.202200] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/30/2019] [Indexed: 12/27/2022] Open
Abstract
The emergence of treatment resistant sub-clones is a key feature of relapse in multiple myeloma. Therapeutic attempts to extend remission and prevent relapse include maximizing response and the use of maintenance therapy. We used whole exome sequencing to study the genetics of paired samples taken at presentation and at relapse from 56 newly diagnosed patients, following induction therapy, randomized to receive either lenalidomide maintenance or observation as part of the Myeloma XI trial. Patients included were considered high risk, relapsing within 30 months of maintenance randomization. Patients achieving a complete response had predominantly branching evolutionary patterns leading to relapse, characterized by a greater mutational burden, an altered mutational profile, bi-allelic inactivation of tumor suppressor genes, and acquired structural aberrations. Conversely, in patients achieving a partial response, the evolutionary features were predominantly stable with a similar mutational and structural profile seen at both time points. There were no significant differences between patients relapsing after lenalidomide maintenance versus observation. This study shows that the depth of response is a key determinant of the evolutionary patterns seen at relapse. This trial is registered at clinicaltrials.gov identifier: 01554852.
Collapse
Affiliation(s)
- John R Jones
- Department of Haematology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Niels Weinhold
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Cody Ashby
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brian A Walker
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chris Wardell
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Charlotte Pawlyn
- Department of Haematology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Leo Rasche
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - David A Cairns
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, UK
| | - Walter M Gregory
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, UK
| | | | - Dil B Begum
- The Institute of Cancer Research, London, UK
| | - Sidra Ellis
- The Institute of Cancer Research, London, UK
| | - Amy L Sherborne
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gordon Cook
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Martin F Kaiser
- Department of Haematology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Mark T Drayson
- Clinical Immunology, School of Immunity and Infection, University of Birmingham, UK
| | - Roger G Owen
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Graham H Jackson
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Faith E Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mel Greaves
- The Institute of Cancer Research, London, UK
| | - Gareth J Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
44
|
Nass SJ, Rothenberg ML, Pentz R, Hricak H, Abernethy A, Anderson K, Gee AW, Harvey RD, Piantadosi S, Bertagnolli MM, Schrag D, Schilsky RL. Accelerating anticancer drug development - opportunities and trade-offs. Nat Rev Clin Oncol 2019; 15:777-786. [PMID: 30275514 DOI: 10.1038/s41571-018-0102-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The traditional approach to drug development in oncology, with discrete phases of clinical testing, is becoming untenable owing to expansion of the precision medicine paradigm, whereby patients are stratified into multiple subgroups according to the underlying cancer biology. Seamless approaches to drug development in oncology hold great promise of accelerating the accessibility of novel therapeutic agents to the public but are also accompanied by important trade-offs, including the limited availability of information on the clinical benefit and safety of novel agents at the time of market entry. In this Perspectives article, we describe several opportunities, in the form of novel trial designs or modelling strategies, to improve the efficiency of drug development in oncology, as well as new mechanisms to obtain information about anticancer therapies throughout their life cycle, such as innovative functional imaging techniques or the use of real-world clinical data.
Collapse
Affiliation(s)
- Sharyl J Nass
- Health and Medicine Division, National Academies of Sciences, Engineering and Medicine, Washington, DC, USA.
| | - Mace L Rothenberg
- Global Product Development, Pfizer Oncology, Pfizer, New York, NY, USA
| | - Rebecca Pentz
- Department of Hematology & Medical Oncology, Emory University School of Medicine, and Winship Cancer Institute, Atlanta, GA, USA
| | - Hedvig Hricak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Kenneth Anderson
- Lebow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amanda Wagner Gee
- Health and Medicine Division, National Academies of Sciences, Engineering and Medicine, Washington, DC, USA
| | - R Donald Harvey
- Department of Hematology & Medical Oncology, Emory University School of Medicine, and Winship Cancer Institute, Atlanta, GA, USA
| | - Steven Piantadosi
- Department of Surgery, Brigham and Women's Cancer Center, Boston, MA, USA
| | | | - Deborah Schrag
- Division of Population Sciences, Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | |
Collapse
|
45
|
Cook G, Royle K, O'Connor S, Cairns DA, Ashcroft AJ, Williams CD, Hockaday A, Cavenagh JD, Snowden JA, Ademokun D, Tholouli E, Andrews VE, Jenner M, Parrish C, Yong K, Cavet J, Hunter H, Bird JM, Pratt G, Drayson MT, Brown JM, Morris TCM. The impact of cytogenetics on duration of response and overall survival in patients with relapsed multiple myeloma (long-term follow-up results from BSBMT/UKMF Myeloma X Relapse [Intensive]): a randomised, open-label, phase 3 trial. Br J Haematol 2019; 185:450-467. [PMID: 30729512 PMCID: PMC6519200 DOI: 10.1111/bjh.15782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/11/2018] [Indexed: 12/04/2022]
Abstract
The Myeloma X trial (ISCRTN60123120) registered patients with relapsed multiple myeloma. Participants were randomised between salvage autologous stem cell transplantation (ASCT) or weekly cyclophosphamide following re-induction therapy. Cytogenetic analysis performed at trial registration defined t(4;14), t(14;16) and del(17p) as high-risk. The effect of cytogenetics on time to progression (TTP) and overall survival was investigated. At 76 months median follow-up, ASCT improved TTP compared to cyclophosphamide (19 months (95% confidence interval [95% CI] 16-26) vs. 11 months (9-12), hazard ratio [HR]: 0·40, 95% CI: 0·29-0·56, P < 0·001), on which the presence of any single high-risk lesion had a detrimental impact [likelihood ratio test (LRT): P = 0·011]. ASCT also improved OS [67 months (95% CI 59-not reached) vs. 55 months (44-67), HR: 0·64, 95% CI: 0·42-0·99, P = 0·0435], with evidence of a detrimental impact with MYC rearrangement (LRT: P = 0·021). Twenty-one (24·7%) cyclophosphamide patients received an ASCT post-trial, median OS was not reached (95% CI: 39-not reached) for these participants compared to 31 months (22-39), in those who did not receive a post-trial ASCT. The analysis further supports the benefit of salvage ASCT, which may still be beneficial after second relapse in surviving patients. There is evidence that this benefit reduces in cytogenetic high-risk patients, highlighting the need for targeted study in this patient group.
Collapse
MESH Headings
- Aged
- Antineoplastic Agents, Alkylating/therapeutic use
- Chromosomes, Human, Pair 14/genetics
- Chromosomes, Human, Pair 14/ultrastructure
- Chromosomes, Human, Pair 16/genetics
- Chromosomes, Human, Pair 16/ultrastructure
- Chromosomes, Human, Pair 17/genetics
- Chromosomes, Human, Pair 17/ultrastructure
- Chromosomes, Human, Pair 4/genetics
- Chromosomes, Human, Pair 4/ultrastructure
- Clinical Trials, Phase III as Topic/statistics & numerical data
- Combined Modality Therapy
- Cyclophosphamide/therapeutic use
- Disease-Free Survival
- Female
- Follow-Up Studies
- Hematopoietic Stem Cell Transplantation
- Humans
- In Situ Hybridization, Fluorescence
- Kaplan-Meier Estimate
- Male
- Middle Aged
- Multicenter Studies as Topic/statistics & numerical data
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/mortality
- Multiple Myeloma/therapy
- Proportional Hazards Models
- Randomized Controlled Trials as Topic/statistics & numerical data
- Salvage Therapy
- Sequence Deletion
- Translocation, Genetic
- Transplantation, Autologous
Collapse
Affiliation(s)
- Gordon Cook
- Leeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
- Clinical Trials Research UnitLeeds Institute of Clinical Trials ResearchUniversity of LeedsLeedsUK
| | - Kara‐Louise Royle
- Clinical Trials Research UnitLeeds Institute of Clinical Trials ResearchUniversity of LeedsLeedsUK
| | | | - David A. Cairns
- Clinical Trials Research UnitLeeds Institute of Clinical Trials ResearchUniversity of LeedsLeedsUK
| | | | - Cathy D. Williams
- Department of HaematologyCentre for Clinical HaematologyNottingham City HospitalsNottinghamUK
| | - Anna Hockaday
- Clinical Trials Research UnitLeeds Institute of Clinical Trials ResearchUniversity of LeedsLeedsUK
| | | | - John A. Snowden
- Department of HaematologySheffield Teaching Hospitals NHS Foundation TrustSheffieldUK
| | | | - Eleni Tholouli
- Department of HaematologyManchester Royal InfirmaryManchesterUK
| | | | - Matthew Jenner
- University Hospital Southampton NHS FoundationSouthamptonUK
| | | | - Kwee Yong
- Department of HaematologyUniversity College HospitalLondonUK
| | - Jim Cavet
- Department of HaematologyThe Christie NHS Foundation TrustManchesterUK
| | - Hannah Hunter
- Department of HaematologyPlymouth Hospitals TrustPlymouthUK
| | - Jenny M. Bird
- Department of HaematologyUniversity Hospitals Bristol NHS TrustBristolUK
| | - Guy Pratt
- Department of HaematologyHeart of England NHS TrustBirminghamUK
| | | | - Julia M. Brown
- Clinical Trials Research UnitLeeds Institute of Clinical Trials ResearchUniversity of LeedsLeedsUK
| | | | | |
Collapse
|
46
|
Phase I trial of isatuximab monotherapy in the treatment of refractory multiple myeloma. Blood Cancer J 2019; 9:41. [PMID: 30926770 PMCID: PMC6440961 DOI: 10.1038/s41408-019-0198-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
This phase I dose-escalation/expansion study evaluated isatuximab (anti-CD38 monoclonal antibody) monotherapy in patients with relapsed/refractory multiple myeloma (RRMM). Patients progressing on or after standard therapy received intravenous isatuximab (weekly [QW] or every 2 weeks [Q2W]). The primary objective was to determine the maximum tolerated dose (MTD) of isatuximab. Overall, 84 patients received ≥ 1 dose of isatuximab. The MTD was not reached; no cumulative adverse reactions were noted. The most frequent adverse events were infusion reactions (IRs), occurring in 37/73 patients (51%) following introduction of mandatory prophylaxis. IRs were mostly grade 1/2, occurred predominantly during Cycle 1, and led to treatment discontinuation in two patients. CD38 receptor occupancy reached a plateau of 80% with isatuximab 20 mg/kg (highest dose tested) and was associated with clinical response. In patients receiving isatuximab ≥ 10 mg/kg, overall response rate (ORR) was 23.8% (15/63), including one complete response. In high-risk patients treated with isatuximab 10 mg/kg (QW or Q2W), ORR was 16.7% (3/18). Median (range) duration of response at doses ≥ 10 mg/kg was 25 (8–30) weeks among high-risk patients versus 36 (6–85) weeks for other patients. In conclusion, isatuximab demonstrated a manageable safety profile and clinical activity in patients with RRMM.
Collapse
|
47
|
Bashir Q, Thall PF, Milton DR, Fox PS, Kawedia JD, Kebriaei P, Shah N, Patel K, Andersson BS, Nieto YL, Valdez BC, Parmar S, Rondon G, Delgado R, Hosing C, Popat UR, Oran B, Ciurea SO, Lin P, Weber DM, Thomas SK, Lee HC, Manasanch EE, Orlowski RZ, Williams LA, Champlin RE, Qazilbash MH. Conditioning with busulfan plus melphalan versus melphalan alone before autologous haemopoietic cell transplantation for multiple myeloma: an open-label, randomised, phase 3 trial. LANCET HAEMATOLOGY 2019; 6:e266-e275. [PMID: 30910541 DOI: 10.1016/s2352-3026(19)30023-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Retrospective studies suggest that conditioning therapy with busulfan plus melphalan could result in longer progression-free survival compared with melphalan alone in patients with multiple myeloma undergoing autologous haemopoietic cell transplantation (auto-HCT). We aimed to test this hypothesis in a randomised trial. METHODS The primary objective of the study was to compare progression-free survival with conditioning of busulfan plus melphalan with melphalan alone in patients with multiple myeloma. Patients with newly diagnosed multiple myeloma who were eligible for cell transplantation, aged 70 years or younger, with at least stable disease, were randomly assigned (1:1) to treatment. Patients received either busulfan plus melphalan, with a test dose of busulfan 32 mg/m2 followed by pharmacokinetically adjusted doses on days -7, -6, -5, and -4 to achieve a target daily area under the curve (AUC) of 5000 mmol-minute and melphalan 70 mg/m2 per day on days -2 and -1 (total melphalan dose 140 mg/m2), or a melphalan dose of 200 mg/m2 on day -2. Randomisation was performed via a Clinical Trial Conduct Website at the University of Texas MD Anderson Cancer Center. The accrual is complete and final results are presented here. The study is registered with ClinicalTrials.gov, number NCT01413178. FINDINGS Between Oct 12, 2011, and March 22, 2017, 205 patients were assessed for eligibility and randomly assigned to treatment. The primary analysis of progression-free survival was measured in 202 patients who received treatment: 104 patients in the busulfan plus melphalan group and 98 patients in the melphalan alone group. 90 days after auto-HCT, 102 (98%) of 104 patients given busulfan plus melphalan and 95 (97%) of 98 patients given melphalan alone achieved partial response or better. The median follow-up in the busulfan plus melphalan group was 22·6 months (IQR 15·2-47·1) and 20·2 months (IQR 8·8-46·6) in the melphalan alone group. Median progression-free survival was 64·7 months (32·9-64·7) with busulfan plus melphalan versus 43·5 months (19·9-not estimated) with melphalan alone (hazard ratio 0·53 [95% CI 0·30-0·91]; p=0·022). There were no treatment-related deaths by day 100 in either group. Grade 2-3 mucositis was observed in 77 (74%) of 104 patients in the busulfan plus melphalan group versus 14 (14%) of 98 patients in the melphalan alone group. INTERPRETATION These findings, if confirmed in other ongoing studies, suggest that busulfan plus melphalan could replace melphalan alone as the conditioning regimen for auto-HCT in patients with newly diagnosed myeloma. FUNDING This study was funded in part by the National Institutes of Health (NIH) through MD Anderson's Cancer Center Support Grant (CA016672).
Collapse
Affiliation(s)
- Qaiser Bashir
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA.
| | - Peter F Thall
- Department of Biostatistics, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Denái R Milton
- Department of Biostatistics, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Patricia S Fox
- Department of Biostatistics, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Jitesh D Kawedia
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Nina Shah
- Department of Hematology and Blood and Marrow Transplant, University of California San Francisco, San Francisco, CA, USA
| | - Krina Patel
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Borje S Andersson
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Yago L Nieto
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Ben C Valdez
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Simrit Parmar
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Gabriela Rondon
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Ruby Delgado
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Chitra Hosing
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Uday R Popat
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Betul Oran
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Stefan O Ciurea
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Pei Lin
- Department of Hematopathology, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Donna M Weber
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Sheeba K Thomas
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Hans C Lee
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Elisabet E Manasanch
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Loretta A Williams
- Department of Symptom Research, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation & Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| |
Collapse
|
48
|
Bangi E. A Drosophila Based Cancer Drug Discovery Framework. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1167:237-248. [PMID: 31520359 DOI: 10.1007/978-3-030-23629-8_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In recent years, there has been growing interest in using Drosophila for drug discovery as it provides a unique opportunity to screen small molecules against complex disease phenotypes in a whole animal setting. Furthermore, gene-compound interaction experiments that combine compound feeding with complex genetic manipulations enable exploration of compound mechanisms of response and resistance to an extent that is difficult to achieve in other experimental models. Here, I discuss how compound screening and testing approaches reported in Drosophila fit into the current cancer drug discovery pipeline. I then propose a framework for a Drosophila-based cancer drug discovery strategy which would allow the Drosophila research community to effectively leverage the power of Drosophila to identify candidate therapeutics and push our discoveries into the clinic.
Collapse
Affiliation(s)
- Erdem Bangi
- Department of Biological Science, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
49
|
Gopalakrishnan S, D'Souza A, Scott E, Fraser R, Davila O, Shah N, Gale RP, Kamble R, Diaz MA, Lazarus HM, Savani BN, Hildebrandt GC, Solh M, Freytes CO, Lee C, Kyle RA, Usmani SZ, Ganguly S, Assal A, Berdeja J, Kanate AS, Dhakal B, Meehan K, Kindwall-Keller T, Saad A, Locke F, Seo S, Nishihori T, Gergis U, Gasparetto C, Mark T, Nieto Y, Kumar S, Hari P. Revised International Staging System Is Predictive and Prognostic for Early Relapse (<24 months) after Autologous Transplantation for Newly Diagnosed Multiple Myeloma. Biol Blood Marrow Transplant 2018; 25:683-688. [PMID: 30579965 DOI: 10.1016/j.bbmt.2018.12.141] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/12/2018] [Indexed: 11/15/2022]
Abstract
The revised International Staging System (R-ISS) combines ISS with genetic markers and lactate dehydrogenase and can prognosticate newly diagnosed multiple myeloma (MM). Early relapse (<24 months) after upfront autologous hematopoietic cell transplantation (AHCT) strongly predicts inferior overall survival (OS). We examined the ability of R-ISS in predicting early relapse and its independent prognostic effect on postrelapse survival after an early relapse. Using the Center for International Blood and Marrow Transplant Research database we identified MM patients receiving first AHCT within 18 months after diagnosis with available R-ISS stage at diagnosis (n = 628). Relative risks of relapse/progression, progression-free survival (PFS), and OS were calculated with the R-ISS group as a predictor in multivariate analysis. Among early relapsers, postrelapse survival was tested to identify factors affecting postrelapse OS. The cumulative incidence of early relapse was 23%, 39%, and 50% for R-ISS I, R-ISS II, and R-ISS III, respectively (P < .001). Shorter PFS and OS were seen with higher stage R-ISS. R-ISS was independently predictive for inferior postrelapse OS among early relapsers, as was the presence of ≥3 comorbidities and the use of ≥2 induction chemotherapy lines. R-ISS stage at diagnosis predicts early post-AHCT relapse and independently affects postrelapse survival among early relapsers.
Collapse
Affiliation(s)
| | - Anita D'Souza
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Emma Scott
- Center for Hematologic Malignancies, The Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Raphael Fraser
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Omar Davila
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nina Shah
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert Peter Gale
- Hematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Rammurti Kamble
- Division of Hematology and Oncology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Miguel Angel Diaz
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Hillard M Lazarus
- Seidman Cancer Center, Division of Hematology Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia
| | | | - Cindy Lee
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | - Saad Z Usmani
- Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute/Atrium Health, Charlotte, North Carolina
| | - Siddhartha Ganguly
- Division of Hematological Malignancy and Cellular Therapeutics, University of Kansas Health System, Kansas City, Kansas
| | - Amer Assal
- Columbia University Medical Center, New York, New York
| | | | - Abraham S Kanate
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, West Virginia
| | - Binod Dhakal
- Medical College of Wisconsin, Division of Hematology Oncology; Milwaukee, Wisconsin
| | - Kenneth Meehan
- Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire
| | - Tamila Kindwall-Keller
- Division of Hematology/Oncology, University of Virginia Health System, Charlottesville, Virginia
| | - Ayman Saad
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Frederick Locke
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sachiko Seo
- Department of Hematology & Oncology, National Cancer Research Center East, Chiba, Japan
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Usama Gergis
- Hematolgic Malignancies & Bone Marrow Transplant, Department of Medical Oncology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | | | - Tomer Mark
- University of Colorado Hospital, Division of Hematology Oncology; Aurora, Colorado
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Parameswaran Hari
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
50
|
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: 1.7] [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]
|