151
|
Intercellular Mitochondrial Transfer in the Tumor Microenvironment. Cancers (Basel) 2020; 12:cancers12071787. [PMID: 32635428 PMCID: PMC7407231 DOI: 10.3390/cancers12071787] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/21/2022] Open
Abstract
Cell-to-cell communication is a fundamental process in every multicellular organism. In addition to membrane-bound and released factors, the sharing of cytosolic components represents a new, poorly explored signaling route. An extraordinary example of this communication channel is the direct transport of mitochondria between cells. In this review, we discuss how intercellular mitochondrial transfer can be used by cancer cells to sustain their high metabolic requirements and promote drug resistance and describe relevant molecular players in the context of current and future cancer therapy.
Collapse
|
152
|
Zhou X, Steinhardt MJ, Grathwohl D, Meckel K, Nickel K, Leicht HB, Krummenast F, Einsele H, Rasche L, Kortüm KM. Multiagent therapy with pomalidomide, bortezomib, doxorubicin, dexamethasone, and daratumumab ("Pom-PAD-Dara") in relapsed/refractory multiple myeloma. Cancer Med 2020; 9:5819-5826. [PMID: 32608149 PMCID: PMC7433809 DOI: 10.1002/cam4.3209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/03/2020] [Accepted: 05/16/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Even in the era of novel immunotherapies for multiple myeloma (MM), treatment of late-stage relapsed/refractory (RR) patients remains challenging. The aim of our study was to analyze the efficacy and safety of the five-drug combination pomalidomide, bortezomib, doxorubicin, dexamethasone, and daratumumab ("Pom-PAD-Dara") in RRMM. METHODS We retrospectively analyzed data of 56 patients with RRMM who received Pom-PAD-Dara between September 2016 and May 2019. RESULTS Patients were heavily pretreated with a median of four prior lines of therapy, including autologous and allogenic stem cell transplant in 50 (89%) and six (11%) patients, respectively. The overall response rate (ORR) was 78% and we observed partial remission, very good partial remission, and complete remission in 27 (48%), 13 (23%) and four (7%) patients, respectively. Median progression-free survival was 7 months (95% CI, 3.3-10.7) and the median overall survival was not reached at 24 months. Adverse events grade ≥ 3 were observed 41 (73%) patients and included neutropenia (n = 28, 50%), anemia (n = 22, 39%), thrombocytopenia (n = 21, 38%), and pneumonia (n = 6, 11%). CONCLUSION Pom-PAD-Dara represents a promising multiagent regimen in heavily pretreated RRMM patients with high ORR and an acceptable safety profile.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | | | - Denise Grathwohl
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Katharina Meckel
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Katharina Nickel
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Hans-Benno Leicht
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Franziska Krummenast
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Klaus M Kortüm
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
153
|
Htut TW, Thein KZ, Lawrie A, Tighe J, Preston G. Efficacy of daratumumab combination regimen in patients with multiple myeloma: A combined analysis of phase III randomized controlled trials. EJHAEM 2020; 1:262-266. [PMID: 35847725 PMCID: PMC9175766 DOI: 10.1002/jha2.46] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 11/23/2022]
Abstract
The use of the CD38 monoclonal antibody daratumumab in combination with standard myeloma chemotherapy regimens has been studied extensively in recent years. We undertook an updated meta-analysis of phase III randomized controlled trials (RCT) to determine the efficacy of daratumumab combination regimens. The relative risk for progression was significantly lower in daratumumab-treated cohorts (HR 0.46, 95% CI 0.38-0.55) and this was consistent across newly diagnosed and relapsed cases. No statistically significant improvement was identified in newly diagnosed patients with high-risk cytogenetics and this group remains a therapeutic challenge.
Collapse
Affiliation(s)
- Thura W. Htut
- Department of Haematology, Aberdeen Royal InfirmaryForesterhill Health CampusAberdeenUK
| | - Kyaw Z. Thein
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Alastair Lawrie
- Department of Haematology, Aberdeen Royal InfirmaryForesterhill Health CampusAberdeenUK
| | - Jane Tighe
- Department of Haematology, Aberdeen Royal InfirmaryForesterhill Health CampusAberdeenUK
| | - Gavin Preston
- Department of Haematology, Aberdeen Royal InfirmaryForesterhill Health CampusAberdeenUK
| |
Collapse
|
154
|
Shah N, Aiello J, Avigan DE, Berdeja JG, Borrello IM, Chari A, Cohen AD, Ganapathi K, Gray L, Green D, Krishnan A, Lin Y, Manasanch E, Munshi NC, Nooka AK, Rapoport AP, Smith EL, Vij R, Dhodapkar M. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of multiple myeloma. J Immunother Cancer 2020; 8:e000734. [PMID: 32661116 PMCID: PMC7359060 DOI: 10.1136/jitc-2020-000734] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
Outcomes in multiple myeloma (MM) have improved dramatically in the last two decades with the advent of novel therapies including immunomodulatory agents (IMiDs), proteasome inhibitors and monoclonal antibodies. In recent years, immunotherapy for the treatment of MM has advanced rapidly, with the approval of new targeted agents and monoclonal antibodies directed against myeloma cell-surface antigens, as well as maturing data from late stage trials of chimeric antigen receptor CAR T cells. Therapies that engage the immune system to treat myeloma offer significant clinical benefits with durable responses and manageable toxicity profiles, however, the appropriate use of these immunotherapy agents can present unique challenges for practicing physicians. Therefore, the Society for Immunotherapy of Cancer convened an expert panel, which met to consider the current role of approved and emerging immunotherapy agents in MM and provide guidance to the oncology community by developing consensus recommendations. As immunotherapy evolves as a therapeutic option for the treatment of MM, these guidelines will be updated.
Collapse
Affiliation(s)
- Nina Shah
- Division of Hematology-Oncology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jack Aiello
- Patient Empowerment Network, San Jose, California, USA
| | - David E Avigan
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jesus G Berdeja
- Department of Medicine, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Ivan M Borrello
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center of Johns Hopkins, Baltimore, Maryland, USA
| | - Ajai Chari
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adam D Cohen
- Department of Medicine, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karthik Ganapathi
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lissa Gray
- University of California San Francisco, San Francisco, CA, USA
| | - Damian Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Multiple Myeloma Center for Clinical Research, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Yi Lin
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elisabet Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nikhil C Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ajay K Nooka
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia, USA
| | - Aaron P Rapoport
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Eric L Smith
- Myeloma Service and Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ravi Vij
- Division of Medical Oncology, Siteman Cancer Center, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Madhav Dhodapkar
- School of Medicine, Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
155
|
Premkumar V, Pan S, Lentzsch S, Bhutani D. Use of daratumumab in high risk multiple myeloma: A meta-analysis. EJHAEM 2020; 1:267-271. [PMID: 35847747 PMCID: PMC9175911 DOI: 10.1002/jha2.47] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 11/30/2022]
Abstract
Daratumumab is approved for use in newly diagnosed and relapsed/refractory multiple myeloma (MM), however the patients most likely to benefit from its addition to standard anti-myeloma therapy is unclear. This meta-analysis included 2340 newly diagnosed MM patients (1982 with standard risk and 358 with high risk cytogenetics) and 673 patients with relapsed/refractory MM (513 with standard risk and 160 with high risk cytogenetics) to assess which cytogenetic subgroups derived PFS benefit from Daratumumab. Studies included were the CASSIOPEIA, MAIA and ALCYONE (for newly diagnosed MM) and the CASTOR and POLLUX trials (for relapsed/refractory MM). Daratumumab's addition led to a clear benefit in standard risk newly diagnosed MM (HR 0.43; 95% CI, 0.35-0.53; P < .05) and both high and standard risk relapsed/refractory disease (HR 0.28; 95% CI, 0.21-0.36; P < .05 and HR 0.48; 95% CI, 0.30-0.76; P < .05, respectively). No clear benefit was seen in newly diagnosed high risk MM. These findings fail to demonstrate PFS benefit from Daratumumab's addition in high risk newly diagnosed MM. Data forthcoming from the GRIFFIN and MASTER trials may increase the power of the study and provide a definitive answer. Daratumumab remains important in standard risk upfront and relapsed/refractory MM and high risk relapsed/refractory MM.
Collapse
Affiliation(s)
| | - Samuel Pan
- Columbia University Medical CenterNew YorkNew YorkUSA
| | | | | |
Collapse
|
156
|
Burgos L, Puig N, Cedena MT, Mateos MV, Lahuerta JJ, Paiva B, San-Miguel JF. Measurable residual disease in multiple myeloma: ready for clinical practice? J Hematol Oncol 2020; 13:82. [PMID: 32571377 PMCID: PMC7310444 DOI: 10.1186/s13045-020-00911-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/04/2020] [Indexed: 01/11/2023] Open
Abstract
The landscape of multiple myeloma (MM) has changed considerably in the past two decades regarding new treatments, insight into disease biology and innovation in the techniques available to assess measurable residual disease (MRD) as the most accurate method to evaluate treatment efficacy. The sensitivity and standardization achieved by these techniques together with unprecedented rates of complete remission (CR) induced by new regimens, raised enormous interest in MRD as a surrogate biomarker of patients' outcome and endpoint in clinical trials. By contrast, there is reluctance and general lack of consensus on how to use MRD outside clinical trials. Here, we discuss critical aspects related with the implementation of MRD in clinical practice.
Collapse
Affiliation(s)
- Leire Burgos
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - Noemi Puig
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - María-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - Bruno Paiva
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - Jesús F San-Miguel
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Pamplona, Spain.
| |
Collapse
|
157
|
Schjesvold F. Evolution of diagnostic workup and treatment for multiple myeloma 2013-2019. Eur J Haematol 2020; 105:434-448. [PMID: 32557833 DOI: 10.1111/ejh.13464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To evaluate changes in diagnostic and therapeutic approach in multiple myeloma among Norwegian hematologists in the current decade. METHODS This nationwide study in Norway is based on results of surveys conducted among professionally active hematologists from 2013 to 2019. Every year, respondents participating in the survey suggested treatment regimens used in typical clinical situations in patients with multiple myeloma, as well as diagnostic routines. RESULTS The use of regimens containing alkylators and thalidomide was common at the beginning of the studied period. Later, lenalidomide became the most preferred treatment in most first-line patients. Bortezomib maintained a stable position in the treatment of myeloma in patients with renal insufficiency. The lenalidomide, bortezomib, and dexamethasone combination became the preferred frontline triplet for transplant-ineligible patients and induction therapy before transplant. Nowadays, the relapse after lenalidomide-based treatment is managed using both bortezomib-based therapies and combinations with the newest agents. Together with the therapeutic landscape, the use of diagnostic criteria and workup as well as supportive care changed in the period influenced by local and international guidelines and recommendations. CONCLUSION Norwegian hematologists gradually adopt new clinical concepts, guidelines, and recommendations in their practice.
Collapse
Affiliation(s)
- Fredrik Schjesvold
- Oslo Myeloma Center, Oslo University Hospital, Oslo, Norway.,KG Jebsen Center for B cell Malignancies, University of Oslo, Oslo, Norway
| |
Collapse
|
158
|
Daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma: final results from the phase 2 GEN501 and SIRIUS trials. LANCET HAEMATOLOGY 2020; 7:e447-e455. [PMID: 32470437 DOI: 10.1016/s2352-3026(20)30081-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Daratumumab showed encouraging efficacy as monotherapy in patients with heavily pretreated multiple myeloma in the GEN501 and SIRIUS studies. Here we report a pooled, post-hoc final analysis of these two studies. METHODS GEN501 was an open-label, multicentre, phase 1-2, dose escalation and expansion study done in the Netherlands, the USA, Sweden, and Denmark. Eligible patients had multiple myeloma and had relapsed or were refractory to 2 or more previous lines of treatment that included a proteasome inhibitor or an immunomodulatory drug. SIRIUS was an open-label, multicentre, phase 2 study done in Canada, Spain, and the USA, in which eligible patients with multiple myeloma had received 3 or more previous lines of therapy, including a proteasome inhibitor or an immunomodulatory drug, or were double refractory. In both studies, eligible patients were aged 18 years or older and had an Eastern Cooperative Oncology Group performance status of 2 or less. In part 2 of GEN501, patients were given intravenous daratumumab 16 mg/kg once per week for 8 weeks, twice per month for 8 doses, and then once per month until disease progression. In part 2 of SIRIUS, patients received intravenous daratumumab 16 mg/kg once per week for 8 weeks, twice per month for 16 weeks, and once per month until disease progression. The primary endpoints (safety in GEN501 and overall response rate in SIRIUS) have previously been reported. These trials are registered on ClinicalTrials.gov, NCT00574288 (GEN501) and NCT01985126 (SIRIUS). FINDINGS Patients were enrolled in GEN501 from March 27, 2008, until May 30, 2014, and in SIRIUS from Sept 30, 2013, until May 5, 2014. The combined analysis included 148 patients who received daratumumab 16 mg/kg (42 patients in GEN501 part 2; 106 patients in SIRIUS), with a median follow-up of 36·6 months (IQR 34·5-38·2). Patients had received a median of 5 previous lines of therapy (IQR 4-7), and 128 (87%) of 148 patients were double refractory. The overall response rate was 30·4% (95% CI 23·1-38·5), including 20 (14%) of 148 patients with very good partial response or better (8·5-20·1) and seven (5%) patients reporting complete response or better (1·9-9·5). Among 45 responders, the median duration of response was 8·0 months (95% CI 6·5-14·7). Median overall survival was 20·5 months (95% CI 16·6-28·1), with a 3-year overall survival rate of 36·5% (28·4-44·6). The most common grade 3-4 treatment-emergent adverse events (TEAEs) were anaemia (grade 3, 26 [18%] of 148 patients; no grade 4 events) and thrombocytopenia (grade 3, 13 [9%] of 148 patients; grade 4, 8 [5%] of 148 patients). Serious drug-related TEAEs occurred in 13 (9%) of 148 patients. There were no treatment-related deaths. INTERPRETATION In this analysis, daratumumab 16 mg/kg monotherapy showed durable responses and there were no new safety concerns with longer follow-up. FUNDING Janssen Research & Development.
Collapse
|
159
|
Mohyuddin GR, Sigle M, Chandrasekar VT, Aziz M, Abdallah AO, Shune L, McClune B. Impact of anti-CD38 therapy in multiple myeloma with high-risk cytogenetics: systematic review and meta-analysis. Leuk Lymphoma 2020; 61:2519-2522. [DOI: 10.1080/10428194.2020.1772475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | - Monia Sigle
- School of Medicine, University of Kansas, Kansas City, KS, USA
| | | | - Muhammad Aziz
- Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | - Al-Ola Abdallah
- Department of Hematological Malignancies and Cellular Therapeutics, University of Kansas, Kansas City, KS, USA
| | - Leyla Shune
- Department of Hematological Malignancies and Cellular Therapeutics, University of Kansas, Kansas City, KS, USA
| | - Brian McClune
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
160
|
Alegre A, de la Rubia J, Sureda Balari A, Encinas Rodríguez C, Suárez A, Blanchard MJ, Bargay Lleonart J, Rodríguez-Otero P, Insunza A, Palomera L, Peñarrubia MJ, Ríos-Tamayo R, Casado Montero LF, González MS, Potamianou A, Couturier C, Pei H, Hevia H, Milionis I, Gaudig M, Mateos MV. Results of an Early Access Treatment Protocol of Daratumumab Monotherapy in Spanish Patients With Relapsed or Refractory Multiple Myeloma. Hemasphere 2020; 4:e380. [PMID: 32647799 PMCID: PMC7306316 DOI: 10.1097/hs9.0000000000000380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/18/2020] [Indexed: 01/02/2023] Open
Abstract
Daratumumab is a human CD38-targeted monoclonal antibody approved as monotherapy for heavily pretreated relapsed and refractory multiple myeloma. We report findings for the Spanish cohort of an open-label treatment protocol that provided early access to daratumumab monotherapy and collected safety and patient-reported outcomes data for patients with relapsed or refractory multiple myeloma. At 15 centers across Spain, intravenous daratumumab (16 mg/kg) was administered to 73 patients who had ≥3 prior lines of therapy, including a proteasome inhibitor and an immunomodulatory drug, or who were double refractory to both. The median duration of daratumumab treatment was 3.3 (range: 0.03-13.17) months, with a median number of 12 (range: 1-25) infusions. Grade 3/4 treatment-emergent adverse events were reported in 74% of patients and included lymphopenia (28.8%), thrombocytopenia (27.4%), neutropenia (21.9%), leukopenia (19.2%), and anemia (15.1%). Common (>5%) serious treatment-emergent adverse events included respiratory tract infection (9.6%), general physical health deterioration (6.8%), and back pain (5.5%). Infusion-related reactions occurred in 45% of patients. The median change from baseline in all domains of the EQ-5D-5L and EORTC QLQ-C30 was mostly 0. A total of 18 (24.7%) patients achieved a partial response or better, with 10 (13.7%) patients achieving a very good partial response or better. Median progression-free survival was 3.98 months. The results of this early access treatment protocol are consistent with previously reported trials of daratumumab monotherapy and confirm its safety and antitumoral efficacy in Spanish patients with heavily treated relapsed or refractory multiple myeloma. European Clinical Trials Database number: 2015-002993-19.
Collapse
Affiliation(s)
- Adrián Alegre
- Hospital Universitario de La Princesa, Madrid, Spain
| | - Javier de la Rubia
- Hospital Dr. Peset and School of Medicine and Dentistry, Catholic University of Valencia, Valencia, Spain
| | | | - Cristina Encinas Rodríguez
- Hospital General Universitario Gregorio Marañón, Instituto de Investigacion Sanitaria Gregorio Marañón, Madrid, Spain
| | - Alexia Suárez
- Hospital Universitario Gran Canaria Dr. Negrin, Las Palmas, Spain
| | | | | | | | - Andrés Insunza
- Department of Hematology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Luis Palomera
- Hospital Clínico Universitario Lozano Blesa, Instituto de Investigación de Aragón, Zaragoza, Spain
| | | | | | | | | | | | | | - Huiling Pei
- Janssen Research & Development, Horsham, Pennsylvania, United States
| | - Henar Hevia
- Janssen-Cilag Medical Affairs, Madrid, Spain
| | - Iordanis Milionis
- EMEA Medical Affairs, Janssen-Cilag Pharmaceutical SACI, Athens, Greece
| | | | | |
Collapse
|
161
|
Nikolaenko L, Chhabra S, Biran N, Chowdhury A, Hari PN, Krishnan A, Richter J. Graft-Versus-Host Disease in Multiple Myeloma Patients Treated With Daratumumab After Allogeneic Transplantation. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:407-414. [DOI: 10.1016/j.clml.2020.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/11/2020] [Accepted: 01/16/2020] [Indexed: 01/05/2023]
|
162
|
Leblay N, Maity R, Hasan F, Neri P. Deregulation of Adaptive T Cell Immunity in Multiple Myeloma: Insights Into Mechanisms and Therapeutic Opportunities. Front Oncol 2020; 10:636. [PMID: 32432039 PMCID: PMC7214816 DOI: 10.3389/fonc.2020.00636] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has recently emerged as a promising treatment option for multiple myeloma (MM) patients. Profound immune dysfunction and evasion of immune surveillance are known to characterize MM evolution and disease progression. Along with genomic changes observed in malignant plasma cells, the bone marrow (BM) milieu creates a protective environment sustained by the complex interaction of BM stromal cells (BMSCs) and malignant cells that using bidirectional connections and cytokines released stimulate disease progression, drug resistance and enable immune escape. Local immune suppression and T-cell exhaustion are important mediating factors of clinical outcomes and responses to immune-based approaches. Thus, further characterization of the defects present in the immune system of MM patients is essential to develop novel therapies and to repurpose the existing ones. This review seeks to provide insights into the mechanisms that promote tumor escape, cause inadequate T-cell stimulation and impaired cytotoxicity in MM. Furthermore, it highlights current immunotherapies being used to restore adaptive T-cell immune responses in MM and describes strategies created to escape these multiple immune evasion mechanisms.
Collapse
Affiliation(s)
- Noémie Leblay
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Ranjan Maity
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Fajer Hasan
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
163
|
Li N, Liu L, Xiang P, Liang L, Wang J, Wang Y, Luo S, Song Y, Fang B. Addition of low‐dose decitabine to bortezomib and dexamethasone as second‐line therapy in multiple myeloma. Br J Haematol 2020; 189:e258-e262. [PMID: 32346851 DOI: 10.1111/bjh.16686] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ning Li
- Department of Oncology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Lina Liu
- Department of Hematology Henan Institute of Haematology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Pu Xiang
- Department of Hematology Henan Institute of Haematology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Lijie Liang
- Department of Hematology Henan Institute of Haematology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Juan Wang
- Department of Hematology Henan Institute of Haematology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Yaomei Wang
- Department of Hematology Henan Institute of Haematology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Suxia Luo
- Department of Oncology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Yongping Song
- Department of Hematology Henan Institute of Haematology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| | - Baijun Fang
- Department of Hematology Henan Institute of Haematology Henan Cancer Hospital Henan Cancer Hospital Affiliated to Zhengzhou University Zhengzhou University Zhengzhou China
| |
Collapse
|
164
|
Kocoglu MH, Badros AZ. Newly diagnosed multiple myeloma: current treatment strategies, emerging therapeutic approaches and beyond. Expert Rev Hematol 2020; 13:669-686. [PMID: 32290719 DOI: 10.1080/17474086.2020.1756258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION As we have just stepped into a new decade of hopes, the mountain of knowledge learned from multiple myeloma (MM) remains unmatched among cancers. In the last decade alone, this rapid-sequence learning curve has led to regulatory approvals of eight drugs with mechanisms of actions representing five different areas of cell biology some of which made to the frontline setting, sparking debates about how to best sequence them in the treatment continuum of induction, consolidation, and maintenance and gained momentum with the realization of the implications of an effective upfront therapeutic approach with potential impact on survival. AREAS COVERED This review was written with an intent to introduce the reader to the current treatment approach of a newly diagnosed myeloma patient and acquaint with promising targets and mechanistic strategies. Medline and clinicaltrials.gov databases (2000-2020) and relevant meetings (ASH, ASCO, EHA, ESMO, IMW) reports were queried and guidelines (IMWG) were reviewed to distill to expert opinion in an inundating field. EXPERT OPINION Future holds promise with new targets on the horizon. It is likely that the new age of myeloma will belong to quadruplets with the addition of acellular or cellular biologics to first-generation novel agents, leading to new paradigms.
Collapse
Affiliation(s)
- Mehmet H Kocoglu
- Department of Medicine, Division of Oncology, University of Maryland Medical Center , Baltimore, MD, USA
| | - Ashraf Z Badros
- Department of Medicine, Division of Oncology, University of Maryland Medical Center , Baltimore, MD, USA
| |
Collapse
|
165
|
Franssen LE, Stege CAM, Zweegman S, van de Donk NWCJ, Nijhof IS. Resistance Mechanisms Towards CD38-Directed Antibody Therapy in Multiple Myeloma. J Clin Med 2020; 9:E1195. [PMID: 32331242 PMCID: PMC7230744 DOI: 10.3390/jcm9041195] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/10/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
Antibodies targeting CD38 are rapidly changing the treatment landscape of multiple myeloma (MM). CD38-directed antibodies have several mechanisms of action. Fc-dependent immune effector mechanisms include complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and apoptosis. In addition, direct effects and immunomodulatory effects contribute to the efficacy of CD38-directed antibodies. Daratumumab, the first-in-class anti-CD38 monoclonal antibody, is now part of standard treatment regimens of both newly diagnosed as well as relapsed/refractory MM patients. The FDA has recently approved isatuximab in combination with pomalidomide and dexamethasone for relapsed/refractory MM patients after at least two prior therapies. Further, the other CD38-targeting antibodies (i.e., MOR202 and TAK-079) are increasingly used in clinical trials. The shift to front-line treatment of daratumumab will lead to an increase in patients refractory to CD38 antibody therapy already after first-line treatment. Therefore, it is important to gain insight into the mechanisms of resistance to CD38-targeting antibodies in MM, and to develop strategies to overcome this resistance. In the current review, we will briefly describe the most important clinical data and mechanisms of action and will focus in depth on the current knowledge on mechanisms of resistance to CD38-targeting antibodies and potential strategies to overcome this.
Collapse
Affiliation(s)
- Laurens E. Franssen
- Department of Hematology, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (C.A.M.S.); (S.Z.); (N.W.C.J.v.d.D.); (I.S.N.)
| | | | | | | | | |
Collapse
|
166
|
Calabretta E, Carlo-Stella C. The Many Facets of CD38 in Lymphoma: From Tumor-Microenvironment Cell Interactions to Acquired Resistance to Immunotherapy. Cells 2020; 9:E802. [PMID: 32225002 PMCID: PMC7226059 DOI: 10.3390/cells9040802] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
The CD38 antigen is expressed in several hematological malignancies, and the anti-CD38 monoclonal antibodies Daratumumab and Isatuximab have an established role in the therapy of multiple myeloma. However, data on the therapeutic utility of CD38 targeting in other lymphoid malignancies are limited. In chronic lymphocytic leukemia, the prognostic significance of CD38 expression is well accepted, and preclinical studies on the use of Daratumumab in monotherapy or combination therapy have demonstrated considerable efficacy. In other lymphoproliferative disorders, preclinical and clinical data have not been as compelling; however, CD38 overexpression likely contributes to resistance to checkpoint inhibitors, prompting numerous clinical trials in Hodgkin and non-Hodgkin lymphoma to investigate whether blocking CD38 enhances the efficacy of checkpoint inhibitors. Furthermore, due to its widespread expression in hematological tumors, CD38 represents an attractive target for cellular therapies such as CAR-T cells. The present review discusses current knowledge of CD38 expression and its implications in various lymphoid malignancies. Furthermore, it addresses current and future therapeutic perspectives, with a particular emphasis on the significance of CD38 interaction with immune cells of the tumor microenvironment. Lastly, results of ongoing studies using anti-CD38 antibodies will be reviewed.
Collapse
Affiliation(s)
- Eleonora Calabretta
- Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, 20089 Milano, Italy;
| | - Carmelo Carlo-Stella
- Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, 20089 Milano, Italy;
- Department of Biomedical Sciences, Humanitas University, Rozzano, 20089 Milano, Italy
| |
Collapse
|
167
|
Kothari S, Hillengass J, McCarthy PL, Holstein SA. Determination of Minimal Residual Disease in Multiple Myeloma: Does It Matter? Curr Hematol Malig Rep 2020; 14:39-46. [PMID: 30671912 DOI: 10.1007/s11899-019-0497-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW The ability to detect minimal residual disease (MRD) in myeloma has improved due to advances in flow cytometry and sequencing methodologies. Here, we evaluate recent clinical trial data and explore the current and future roles of MRD assessment in the context of clinical trial design and clinical practice. RECENT FINDINGS A review of recent phase III studies reveals that achievement of MRD negativity is associated with improved progression-free survival (PFS) and/or overall survival (OS). Treatment arms that are more effective from a PFS or overall response rate perspective are also associated with superior MRD negativity rates. The current standard MRD methodologies are limited by requiring bone marrow samples and refinement of methodologies that can detect disease outside of the bone marrow is needed. Currently, MRD is a prognostic biomarker and further efforts are required to determine whether it can serve as a surrogate endpoint. The use of MRD status to guide treatment decisions is currently not recommended outside the confines of a clinical trial.
Collapse
Affiliation(s)
- Shalin Kothari
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jens Hillengass
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Philip L McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, 986840 Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
168
|
Tachita T, Kinoshita S, Ri M, Aoki S, Asano A, Kanamori T, Yoshida T, Totani H, Ito A, Kusumoto S, Komatsu H, Yamagata K, Kubo K, Tohkin M, Fukuda S, Iida S. Expression, mutation, and methylation of cereblon-pathway genes at pre- and post-lenalidomide treatment in multiple myeloma. Cancer Sci 2020; 111:1333-1343. [PMID: 32061138 PMCID: PMC7156787 DOI: 10.1111/cas.14352] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/03/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Cereblon (CRBN) is a target for immunomodulatory drugs. This study investigated the prognostic value of the expression of CRBN‐pathway genes on the clinical relevance of lenalidomide (Len) treatment and evaluated the levels of CRBN‐binding proteins and mutations in these genes after Len treatment. Forty‐eight primary multiple myeloma cells were collected prior to treatment with Len and dexamethasone (Ld) and 25 paired samples were obtained post‐Ld therapy. These tumor cells were used to determine the expression and mutated forms of the CRBN‐pathway genes. Following normalization with CRBN levels, there was a significantly reduced IKZF1/CRBN ratio in samples that responded poorly to Ld therapy. Moreover, patients with low ratios of IKZF1/CRBN showed a significantly shorter progression‐free survival (PFS) and overall survival (OS) than those with higher ratios. However, patients with high ratios of KPNA2/CRBN showed a significantly shorter PFS and OS than patients with lower ratios. Of the 25 paired samples analyzed, most samples showed a reduction in the expression of CRBN and an increase in IKZF1 gene expression. No mutations were observed in CRBN, IKZF1, or CUL4A genes in the post‐Ld samples. In conclusion, a decreased expression of IKZF1 and increased expression of KPNA2 compared to that of CRBN mRNA predicts poor outcomes of Ld therapy.
Collapse
Affiliation(s)
- Takuto Tachita
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shiori Kinoshita
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Blood Transfusion and Cell Therapy, Nagoya City University Hospital, Nagoya, Japan
| | - Sho Aoki
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Arisa Asano
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Kanamori
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Yoshida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Clinical Oncology, Nagoya Memorial Hospital, Nagoya, Japan
| | - Haruhito Totani
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Asahi Ito
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shigeru Kusumoto
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirokazu Komatsu
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazufumi Yamagata
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kohmei Kubo
- Department of Hematology, Aomori Prefectural Central Hospital, Aomori, Japan
| | - Masahiro Tohkin
- Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Shinsaku Fukuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
169
|
Chari A, Cornell RF, Gasparetto C, Karanes C, Matous JV, Niesvizky R, Lunning M, Usmani SZ, Anderson LD, Chhabra S, Girnius S, Shustik C, Stuart R, Lee Y, Salman Z, Liu E, Valent J. Final analysis of a phase 1/2b study of ibrutinib combined with carfilzomib/dexamethasone in patients with relapsed/refractory multiple myeloma. Hematol Oncol 2020; 38:353-362. [PMID: 32053229 PMCID: PMC7496325 DOI: 10.1002/hon.2723] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/31/2020] [Accepted: 02/08/2020] [Indexed: 01/10/2023]
Abstract
Patients with multiple myeloma (MM) inevitably relapse on initial treatment regimens, and novel combination therapies are needed. Ibrutinib is a first‐in‐class, once‐daily inhibitor of Bruton's tyrosine kinase, an enzyme implicated in growth and survival of MM cells. Preclinical data suggest supra‐additivity or synergy between ibrutinib and proteasome inhibitors (PIs) against MM. This phase 1/2b study evaluated the efficacy and safety of ibrutinib plus the PI carfilzomib and dexamethasone in patients with relapsed/refractory MM (RRMM). In this final analysis, we report results in patients who received the recommended phase 2 dose (RP2D; ibrutinib 840 mg and carfilzomib 36 mg/m2 with dexamethasone), which was determined in phase 1. The primary efficacy endpoint was overall response rate (ORR). Fifty‐nine patients with RRMM received the RP2D (18 in phase 1 and 41 in phase 2b). These patients had received a median of three prior lines of therapy; 69% were refractory to bortezomib, and 90% were refractory to their last treatment. ORR in the RP2D population was 71% (stringent complete response and complete response: 3% each). Median duration of clinical benefit and median duration of response were both 6.5 months. Median progression‐free survival (PFS) was 7.4 months, and median overall survival (OS) was 35.9 months. High‐risk patients had comparable ORR and median PFS (67% and 7.7 months, respectively) to non–high‐risk patients, whose ORR was 73% and median PFS was 6.9 months, whereas median OS in high‐risk patients was 13.9 months and not reached in non–high‐risk patients. The most common grade ≥3 hematologic treatment‐emergent adverse events (TEAEs) were anemia and thrombocytopenia (17% each); the most common grade ≥3 non‐hematologic TEAE was hypertension (19%). In patients with RRMM treated with multiple previous lines of therapy, ibrutinib plus carfilzomib demonstrated anticancer activity within the expected efficacy range. No new safety signals were identified and the combination was well‐tolerated.
Collapse
Affiliation(s)
- Ajai Chari
- Department of Hematology and Medical Oncology, Mount Sinai School of Medicine, New York, New York
| | - Robert F Cornell
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cristina Gasparetto
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Health System, Durham, North Carolina
| | - Chatchada Karanes
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Jeffrey V Matous
- Department of Hematology/Oncology, Colorado Blood Cancer Institute, Denver, Colorado
| | - Ruben Niesvizky
- Department of Medical Oncology, Weill Cornell Medicine, New York, New York
| | - Matthew Lunning
- Division of Oncology & Hematology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Saad Z Usmani
- Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | - Larry D Anderson
- Division of Hematology Oncology, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Saulius Girnius
- Department of Medicine, University of Cincinnati Cancer Institute, Cincinnati, Ohio
| | - Chaim Shustik
- Division of Hematology, Cedars Cancer Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Robert Stuart
- Division of Hematology/Oncology, Medical University of South Carolina, Charleston, South Carolina
| | - Yihua Lee
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Zeena Salman
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Emily Liu
- Pharmacyclics LLC, an AbbVie Company, Sunnyvale, California
| | - Jason Valent
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
170
|
Dimopoulos MA, Niesvizky R, Weisel K, Siegel DS, Hajek R, Mateos MV, Cavo M, Huang M, Zahlten-Kumeli A, Moreau P. Once- versus twice-weekly carfilzomib in relapsed and refractory multiple myeloma by select patient characteristics: phase 3 A.R.R.O.W. study subgroup analysis. Blood Cancer J 2020; 10:35. [PMID: 32152297 PMCID: PMC7062899 DOI: 10.1038/s41408-020-0300-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 12/11/2019] [Accepted: 01/03/2020] [Indexed: 12/29/2022] Open
Abstract
The phase 3 A.R.R.O.W. study demonstrated that treatment with once-weekly carfilzomib (70 mg/m2) and dexamethasone (once-weekly Kd70 mg/m2) improved progression-free survival compared with twice-weekly carfilzomib (27 mg/m2) and dexamethasone (twice-weekly Kd27 mg/m2) in patients with relapsed and refractory multiple myeloma (RRMM; median, 11.2 versus 7.6 months; hazard ratio [HR] = 0.69; 95% confidence interval, 0.54-0.88; P = 0.0029). Once-weekly dosing also improved response rates and depth of response. We performed a subgroup analysis from A.R.R.O.W. according to age (<65, 65-74, or ≥75 years), renal function (creatinine clearance <50, ≥50-<80, or ≥80 mL/min), number of prior therapies (2 or 3), and bortezomib-refractory status (yes or no). Compared with twice-weekly Kd27 mg/m2, once-weekly Kd70 mg/m2 reduced the risk of progression or death (HR = 0.60-0.85) and increased overall response rates in nearly all the examined subgroups, consistent with reports in the overall A.R.R.O.W. population. The safety profiles of once-weekly Kd70 mg/m2 across subgroups were also generally consistent with those in the overall population. Findings from this subgroup analysis generally demonstrate a favorable benefit-risk profile of once-weekly Kd70 mg/m2, further supporting once-weekly carfilzomib dosing as an appropriate treatment option for patients with RRMM, regardless of baseline patient and disease characteristics.
Collapse
Affiliation(s)
| | - Ruben Niesvizky
- Center for Myeloma, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Katja Weisel
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- University Hospital of Tuebingen, Tuebingen, Germany
| | - David S Siegel
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, USA
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | | | - Michele Cavo
- "Seràgnoli" Institute of Hematology and Medical Oncology, Bologna University School of Medicine, Bologna, Italy
| | | | | | - Philippe Moreau
- Hematology Department, University Hospital Hotel-Dieu, Nantes, France
| |
Collapse
|
171
|
Soekojo CY, Ooi M, de Mel S, Chng WJ. Immunotherapy in Multiple Myeloma. Cells 2020; 9:E601. [PMID: 32138182 PMCID: PMC7140529 DOI: 10.3390/cells9030601] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma is a complex disease and immune dysfunction has been known to play an important role in the disease pathogenesis, progression, and drug resistance. Recent efforts in drug development have been focused on immunotherapies to modify the MM disease process. Here, we summarize the emerging immunotherapies in the MM treatment landscape.
Collapse
Affiliation(s)
| | | | | | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore; (C.Y.S.); (M.O.); (S.d.M.)
| |
Collapse
|
172
|
Bolli N, Genuardi E, Ziccheddu B, Martello M, Oliva S, Terragna C. Next-Generation Sequencing for Clinical Management of Multiple Myeloma: Ready for Prime Time? Front Oncol 2020; 10:189. [PMID: 32181154 PMCID: PMC7057289 DOI: 10.3389/fonc.2020.00189] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Personalized treatment is an attractive strategy that promises increased efficacy with reduced side effects in cancer. The feasibility of such an approach has been greatly boosted by next-generation sequencing (NGS) techniques, which can return detailed information on the genome and on the transcriptome of each patient's tumor, thus highlighting biomarkers of response or druggable targets that may differ from case to case. However, while the number of cancers sequenced is growing exponentially, much fewer cases are amenable to a molecularly-guided treatment outside of clinical trials to date. In multiple myeloma, genomic analysis shows a variety of gene mutations, aneuploidies, segmental copy-number changes, translocations that are extremely heterogeneous, and more numerous than other hematological malignancies. Currently, in routine clinical practice we employ reduced FISH panels that only capture three high-risk features as part of the R-ISS. On the contrary, recent advances have suggested that extending genomic analysis to the full spectrum of recurrent mutations and structural abnormalities in multiple myeloma may have biological and clinical implications. Furthermore, increased efficacy of novel treatments can now produce deeper responses, and standard methods do not have enough sensitivity to stratify patients in complete biochemical remission. Consequently, NGS techniques have been developed to monitor the size of the clone to a sensitivity of up to a cell in a million after treatment. However, even these techniques are not within reach of standard laboratories. In this review we will recapitulate recent advances in multiple myeloma genomics, with special focus on the ones that may have immediate translational impact. We will analyze the benefits and pitfalls of NGS-based diagnostics, highlighting crucial aspects that will need to be taken into account before this can be implemented in most laboratories. We will make the point that a new era in myeloma diagnostics and minimal residual disease monitoring is close and conventional genetic testing will not be able to return the required information. This will mandate that even in routine practice NGS should soon be adopted owing to a higher informative potential with increasing clinical benefits.
Collapse
Affiliation(s)
- Niccolo Bolli
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Bachisio Ziccheddu
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Marina Martello
- Seràgnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Stefania Oliva
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Carolina Terragna
- Seràgnoli Institute of Hematology, Azienda Ospedaliero-Universitaria Sant'Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
173
|
Economic Evaluation of Adding Daratumumab to a Regimen of Bortezomib + Dexamethasone in Relapsed or Refractory Multiple Myeloma: Based on the Latest Updated Analysis of CASTOR. Clin Ther 2020; 42:251-262.e5. [DOI: 10.1016/j.clinthera.2019.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/28/2019] [Accepted: 12/09/2019] [Indexed: 01/22/2023]
|
174
|
Mateos MV, Spencer A, Nooka AK, Pour L, Weisel K, Cavo M, Laubach JP, Cook G, Iida S, Benboubker L, Usmani SZ, Yoon SS, Bahlis NJ, Chiu C, Ukropec J, Schecter JM, Qin X, O'Rourke L, Dimopoulos MA. Daratumumab-based regimens are highly effective and well tolerated in relapsed or refractory multiple myeloma regardless of patient age: subgroup analysis of the phase 3 CASTOR and POLLUX studies. Haematologica 2020; 105:468-477. [PMID: 31221782 PMCID: PMC7012498 DOI: 10.3324/haematol.2019.217448] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/20/2019] [Indexed: 12/17/2022] Open
Abstract
The phase 3 POLLUX and CASTOR studies demonstrated superior benefit of daratumumab plus lenalidomide/dexamethasone or bortezomib/dexamethasone in relapsed/refractory multiple myeloma. Efficacy and safety of daratumumab was analyzed according to age groups of 65 to 74 years and ≥75 years. Patients received ≥1 prior line of therapy. In POLLUX, patients received lenalidomide/dexamethasone ± daratumumab (16 mg/kg weekly, cycles 1-2; every two weeks, cycles 3-6; monthly until progression). In CASTOR, patients received eight cycles of bortezomib/dexamethasone ± daratumumab (16 mg/kg weekly, cycles 1-3; every three weeks, cycles 4-8; monthly until progression). Patients aged >75 years received dexamethasone 20 mg weekly. For patients aged ≥75 years in POLLUX (median follow-up: 25.4 months), daratumumab/lenalido-mide/dexamethasone prolonged progression-free survival versus lenalido-mide/dexamethasone (median: 28.9 versus 11.4 months; hazard ratio, 0.27; 95% confidence interval, 0.10-0.69; P=0.0042) and increased overall response rate (93.1% versus 76.5%; P=0.0740). Neutropenia was the most common grade 3/4 treatment-emergent adverse event (daratumumab: 44.8%; control: 31.4%). Infusion-related reactions occurred in 12 (41.4%) patients. For patients aged ≥75 years in CASTOR (median follow-up: 19.4 months), daratumumab/bortezomib/dexamethasone prolonged progression-free survival versus bortezomib/dexamethasone (median: 17.9 versus 8.1 months; hazard ratio, 0.26; 95% confidence interval, 0.10-0.65; P=0.0022) and increased overall response rate (95.0% versus 78.8%; P=0.1134). Thrombocytopenia was the most common grade 3/4 treatment-emergent adverse event (daratumumab: 45.0%; control: 37.1%). Infusion-related reactions occurred in 13 (65.0%) patients. Similar findings were reported for patients aged 65 to 74 years in both studies. Taken together, this subgroup analysis of efficacy and safety of daratumumab was largely consistent with the overall populations.
Collapse
Affiliation(s)
| | - Andrew Spencer
- Malignant Haematology and Stem Cell Transplantation Service, Alfred Health-Monash University, Melbourne, Australia
| | - Ajay K Nooka
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Ludek Pour
- University Hospital Brno, Brno, Czech Republic
| | - Katja Weisel
- University Medical Center of Hamburg-Eppendorf, Hamburg, Germany
| | - Michele Cavo
- Institute of Hematology Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Jacob P Laubach
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Gordon Cook
- St James's Institute of Oncology, Leeds Teaching Hospitals NHS Trust and University of Leeds, Leeds, United Kingdom
| | - Shinsuke Iida
- Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Lotfi Benboubker
- Service d'Hématologie et Thérapie Cellulaire, Hôpital Bretonneau, Centre Hospitalier Régional Universitaire (CHRU), Tours, France
| | - Saad Z Usmani
- Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Nizar J Bahlis
- Tom Baker Cancer Center, University of Calgary, Calgary, Alberta, Canada
| | | | - Jon Ukropec
- Janssen Global Medical Affairs, Horsham, PA, USA
| | | | - Xiang Qin
- Janssen Research and Development, LLC, Spring House, PA, USA
| | - Lisa O'Rourke
- Janssen Research and Development, LLC, Spring House, PA, USA
| | | |
Collapse
|
175
|
Ntanasis-Stathopoulos I, Gavriatopoulou M, Terpos E. Antibody therapies for multiple myeloma. Expert Opin Biol Ther 2020; 20:295-303. [PMID: 31944131 DOI: 10.1080/14712598.2020.1717464] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Multiple myeloma (MM) is characterized by the uncontrollable proliferation of plasma cells and the excessive production of a specific type of immunoglobulin. Immune system is deregulated in MM and, thus, immunotherapy is a promising therapeutic strategy.Areas covered: The first approach is to use monoclonal antibodies that recognize specific antigens on the surface of myeloma cells, such as CD38 and B-cell maturation antigen. Upon binding to their target, monoclonal antibodies activate the immune cells to destroy the malignant cell. Anti-CD38 molecules as part of highly effective combination regimens have been approved in both newly diagnosed and relapsed/refractory patients and have significantly changed the myeloma treatment landscape in the recent years. Another strategy is to use antibodies that bind both to a molecule on the surface of the myeloma cell and another molecule on the surface of a T-cell (bispecific antibodies). Consecutively, the T-cell comes close to and recognizes the myeloma cell. These have shown promising results in heavily pre-treated patients.Expert opinion: Antibody therapy has significantly enhanced the armamentarium against MM. Further research should focus on tailoring the combination regimens based on disease and patient characteristics in order to optimize the efficacy and safety.
Collapse
Affiliation(s)
- Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, Alexandra General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, Alexandra General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, Alexandra General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
176
|
Multiple Myeloma and Thrombosis: Prophylaxis and Risk Prediction Tools. Cancers (Basel) 2020; 12:cancers12010191. [PMID: 31940972 PMCID: PMC7016665 DOI: 10.3390/cancers12010191] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/19/2022] Open
Abstract
Thromboembolism in multiple myeloma (MM) patients remains a common complication that renders the optimization of our thromboprophylaxis practice necessary. This review aims to make clear the need for the development of more accurate risk assessment tools and means of thrombosis prevention. Current clinical practice is guided by available guidelines published by the IMWG in 2014, but the extent to which these are implemented is unclear. Recently, several groups developed clinical scores for thrombosis risk in MM in an attempt to improve risk stratification, but these have not been validated or used in clinical practice so far. Research in this field is increasingly focusing on understanding the unique coagulation profile of the MM patient, and data on potential biomarkers that accurately reflect hypercoagulability is emerging. Finally, promising evidence on the effectiveness of direct oral anticoagulants (DOACs) in the context of thrombosis prevention in MM patients is increasingly becoming available. The critical appraisal of the above research areas will establish the necessity of combining disease-specific clinical risk factors with coagulation biomarkers to allow more effective risk stratification that will eventually lead to the reduction of this significant complication. Results from ongoing clinical trials on the role of DOACs are much anticipated.
Collapse
|
177
|
Mateos MV, Cavo M, Blade J, Dimopoulos MA, Suzuki K, Jakubowiak A, Knop S, Doyen C, Lucio P, Nagy Z, Pour L, Cook M, Grosicki S, Crepaldi A, Liberati AM, Campbell P, Shelekhova T, Yoon SS, Iosava G, Fujisaki T, Garg M, Krevvata M, Chen Y, Wang J, Kudva A, Ukropec J, Wroblewski S, Qi M, Kobos R, San-Miguel J. Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): a randomised, open-label, phase 3 trial. Lancet 2020; 395:132-141. [PMID: 31836199 DOI: 10.1016/s0140-6736(19)32956-3] [Citation(s) in RCA: 298] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/20/2019] [Accepted: 11/28/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Standard-of-care treatment for patients with newly diagnosed multiple myeloma includes combination therapies for patients who are not eligible for autologous stem-cell transplantation. At the primary analysis for progression-free survival of the phase 3 ALCYONE trial, progression-free survival was significantly longer with daratumumab in combination with bortezomib, melphalan, and prednisone (D-VMP) versus bortezomib, melphalan, and prednisone (VMP) alone in patients with transplant-ineligible, newly diagnosed multiple myeloma. Here we report updated efficacy and safety results from a prespecified, interim, overall survival analysis of ALCYONE with more than 36 months of follow-up. METHODS ALCYONE was a multicentre, randomised, open-label, active-controlled, phase 3 trial that enrolled patients between Feb 9, 2015, and July 14, 2016, at 162 sites in 25 countries across North America, South America, Europe, and the Asia-Pacific region. Patients were eligible for inclusion if they had newly diagnosed multiple myeloma and were ineligible for high-dose chemotherapy with autologous stem-cell transplantation, because of their age (≥65 years) or because of substantial comorbidities. Patients were randomly assigned in a 1:1 ratio and by permuted block randomisation to receive D-VMP or VMP. An interactive web-based randomisation system was used. Randomisation was stratified by International Staging System disease stage, geographical region, and age. There was no masking to treatment assignments. All patients received up to nine 6-week cycles of subcutaneous bortezomib (1·3 mg/m2 of body surface area on days 1, 4, 8, 11, 22, 25, 29, and 32 of cycle one and on days 1, 8, 22, and 29 of cycles two through nine), oral melphalan (9 mg/m2 once daily on days 1 through 4 of each cycle), and oral prednisone (60 mg/m2 once daily on days 1 through 4 of each cycle). Patients in the D-VMP group also received intravenous daratumumab (16 mg/kg of bodyweight, once weekly during cycle one, once every 3 weeks in cycles two through nine, and once every 4 weeks thereafter as maintenance therapy until disease progression or unacceptable toxicity). The primary endpoint was progression-free survival, which has been reported previously. Results presented are from a prespecified interim analysis for overall survival. The primary analysis population (including for overall survival) was the intention-to-treat population of all patients who were randomly assigned to treatment. The safety population included patients who received any dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT02195479. FINDINGS 706 patients were randomly assigned to treatment groups (350 to the D-VMP group, 356 to the VMP group). At a median follow-up of 40·1 months (IQR 37·4-43·1), a significant benefit in overall survival was observed for the D-VMP group. The hazard ratio (HR) for death in the D-VMP group compared with the VMP group was 0·60 (95% CI 0·46-0·80; p=0·0003). The Kaplan-Meier estimate of the 36-month rate of overall survival was 78·0% (95% CI 73·2-82·0) in the D-VMP group and 67·9% (62·6-72·6) in the VMP group. Progression-free survival, the primary endpoint, remained significantly improved for the D-VMP group (HR 0·42 [0·34-0·51]; p<0·0001). The most frequent adverse events during maintenance daratumumab monotherapy in patients in the D-VMP group were respiratory infections (54 [19%] of 278 patients had upper respiratory tract infections; 42 [15%] had bronchitis, 34 [12%] had viral upper respiratory tract infections), cough (34 [12%]), and diarrhoea (28 [10%]). INTERPRETATION D-VMP prolonged overall survival in patients with newly diagnosed multiple myeloma who were ineligible for stem-cell transplantation. With more than 3 years of follow-up, the D-VMP group continued to show significant improvement in progression-free survival, with no new safety concerns. FUNDING Janssen Research & Development.
Collapse
Affiliation(s)
- Maria-Victoria Mateos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain.
| | - Michele Cavo
- Seràgnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Joan Blade
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | | | - Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | | | - Stefan Knop
- Würzburg University Medical Center, Würzburg, Germany
| | - Chantal Doyen
- CHU UCL Namur, Université catholique de Louvain, Yvoir, Belgium
| | - Paulo Lucio
- Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Zsolt Nagy
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Ludek Pour
- University Hospital Brno, Brno, Czech Republic
| | - Mark Cook
- University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Sebastian Grosicki
- Department of Hematology and Cancer Prevention, Chorzów School of Public Health, Medical University of Silesia, Katowice, Poland
| | - Andre Crepaldi
- Clínica de tratamento e pesquisa em Hematologia e Oncologia, Cuiaba, Brazil
| | | | | | | | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | | | | | - Mamta Garg
- Department of Haematology, Leicester Royal Infirmary, Leicester, UK
| | | | - Ying Chen
- Janssen Research & Development, Raritan, NJ, USA
| | | | - Anupa Kudva
- Janssen Research & Development, Raritan, NJ, USA
| | - Jon Ukropec
- Janssen Global Medical Affairs, Horsham, PA, USA
| | | | - Ming Qi
- Janssen Research & Development, Spring House, PA, USA
| | - Rachel Kobos
- Janssen Research & Development, Raritan, NJ, USA
| | - Jesus San-Miguel
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Centro de Investigación Biomédica en Red de Oncología (CIBERONC), Pamplona, Spain
| |
Collapse
|
178
|
Oriol A, Abril L, Ibarra G, Senin A. Limited treatment options in refractory multiple myeloma: promising therapeutic developments. Expert Rev Anticancer Ther 2020; 20:31-44. [PMID: 31865804 DOI: 10.1080/14737140.2020.1708721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Introduction: Combinations of proteasome inhibitors, immunomodulators, and monoclonal antibodies are highly active against multiple myeloma. Consequently, several combinations have moved from the relapsed to the front-line setting. In the context of lenalidomide and bortezomib being used upfront, salvage options need to be evaluated.Areas covered: This manuscript reviews available data for the treatment of patients progressing on optimal frontline strategies, with a focus on the role of second-generation proteasome inhibitors and immunomodulators, monoclonal antibodies and immunotherapy.Expert opinion: Remarkable progress has been made in myeloma treatment due to the integration of immunomodulators, proteasome inhibitors and more recently monoclonal antibodies in the front-line setting. However, we work on the assumption that most individuals will eventually relapse. Optimized upfront therapy negatively selects more resistant patients among still relapsing individuals. Bortezomib and lenalidomide-exposed patients are under-represented in trials leading to currently approved combinations. Evidence needs to be reviewed taking into account how the improvement of frontline therapy has modified the characteristics of patients at the time of relapse. Second generation immunomodulatory agents and proteasome inhibitors, monoclonal antibodies and other agents have shown efficacy in this new landscape. Immunotherapeutic agents, including CAR-T cells are promising for patients failing standard combinations, despite current data are still immature.
Collapse
Affiliation(s)
- Albert Oriol
- Hematology Service and Hemato-Oncology Clinical Trial Unit, Institut Català d'Oncologia, Badalona, Barcelona, Spain.,Josep Carreras Institute and Germans Trias i Pujol Hospital, Badalona, Barcelona, Spain
| | - Laura Abril
- Hematology Service and Hemato-Oncology Clinical Trial Unit, Institut Català d'Oncologia, Badalona, Barcelona, Spain.,Josep Carreras Institute and Germans Trias i Pujol Hospital, Badalona, Barcelona, Spain
| | - Gladys Ibarra
- Hematology Service and Hemato-Oncology Clinical Trial Unit, Institut Català d'Oncologia, Badalona, Barcelona, Spain.,Josep Carreras Institute and Germans Trias i Pujol Hospital, Badalona, Barcelona, Spain
| | - Alicia Senin
- Hematology Service and Hemato-Oncology Clinical Trial Unit, Institut Català d'Oncologia, Badalona, Barcelona, Spain.,Josep Carreras Institute and Germans Trias i Pujol Hospital, Badalona, Barcelona, Spain
| |
Collapse
|
179
|
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: 1.0] [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
|
180
|
Kiesewetter B, Cherny NI, Boissel N, Cerisoli F, Dafni U, de Vries EGE, Ghia P, Gökbuget N, González-Calle V, Huntly B, Jäger U, Latino NJ, Douillard JY, Malcovati L, Mateos MV, Ossenkoppele GJ, Porkka K, Raderer M, Ribera JM, Scarfò L, Wester R, Zygoura P, Sonneveld P. EHA evaluation of the ESMO-Magnitude of Clinical Benefit Scale version 1.1 (ESMO-MCBS v1.1) for haematological malignancies. ESMO Open 2020; 5:e000611. [PMID: 31958292 PMCID: PMC7003483 DOI: 10.1136/esmoopen-2019-000611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Value frameworks in oncology have not been validated for the assessment of treatments in haematological malignancies, but to avoid overlaps and duplications it appears reasonable to build up experience on existing value frameworks, such as the European Society for Medical Oncology-Magnitude of Clinical Benefit Scale (ESMO-MCBS). METHODS Here we present the results of the first feasibility testing of the ESMO-MCBS v1.1 for haematological malignancies based on the grading of 80 contemporary studies for acute leukaemia, chronic leukaemia, lymphoma, myeloma and myelodysplastic syndromes. The aims were (1) to evaluate the scorability of data, (2) to evaluate the reasonableness of the generated grades for clinical benefit using the current version and (3) to identify shortcomings in the ESMO-MCBS v1.1 that require amendments to improve the efficacy and validity of the scale in grading new treatments in the management of haematological malignancies. RESULTS In general, the ESMO-MCBS v1.1 was found to be widely applicable to studies in haematological malignancies, generating scores that were judged as reasonable by European Hematology Association (EHA) experts. A small number of studies could either not be graded or were not appropriately graded. The reasons, related to the differences between haematological and solid tumour malignancies, are identified and described. CONCLUSIONS Based on the findings of this study, ESMO and EHA are committed to develop a version of the ESMO-MCBS that is validated for haematological malignancies. This development process will incorporate all of the usual stringencies for accountability of reasonableness that have characterised the development of the ESMO-MCBS including field testing, statistical modelling, evaluation for reasonableness and openness to appeal and revision. Applying such a scale will support future public policy decision-making regarding the value of new treatments for haematological malignancies and will provide insights that could be helpful in the design of future clinical trials.
Collapse
Affiliation(s)
- Barbara Kiesewetter
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Nathan I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nicolas Boissel
- Department of Hematology, Hospital Saint-Louis, Paris, Île-de-France, France
- Adolescent and Young Adult Hematology Unit, Diderot University Paris Faculty of Medicine, Paris, Île-de-France, France
| | - Francesco Cerisoli
- European Hematology Association, Den Haag, Zuid-Holland, The Netherlands
| | - Urania Dafni
- Laboratory of Biostatistics, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
- Frontier Science Foundation-Hellas, Frontier Science Foundation-Hellas, Athens, Greece
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Paolo Ghia
- Strategic Research Program on CLL, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Lombardia, Italy
- Universita Vita e Salute San Raffaele, Milano, Lombardia, Italy
| | - Nicola Gökbuget
- Department of Hematology/Oncology, Goethe University, Frankfurt am Main, Hessen, Germany
| | - Verónica González-Calle
- Department of Hematology and Instituto de Investigación Biomédica de Salamanca-IBSAL, University Hospital of Salamanca, Salamanca, Castilla y León, Spain
| | - Brian Huntly
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Ulrich Jäger
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | | | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, Pavia, Lombardia, Italy
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Lombardia, Italy
| | - María-Victoria Mateos
- Department of Hematology and Instituto de Investigación Biomédica de Salamanca-IBSAL, University Hospital of Salamanca, Salamanca, Castilla y León, Spain
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Centre Amsterdam, Amsterdam, Noord-Holland, The Netherlands
| | - Kimmo Porkka
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Markus Raderer
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Josep-Maria Ribera
- Clinical Hematology Department, ICO-Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Catalunya, Spain
| | - Lydia Scarfò
- Strategic Research Program on CLL, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Lombardia, Italy
- Universita Vita e Salute San Raffaele, Milano, Lombardia, Italy
| | - Ruth Wester
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Zuid-Holland, The Netherlands
| | - Panagiota Zygoura
- Frontier Science Foundation-Hellas, Frontier Science Foundation-Hellas, Athens, Greece
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Zuid-Holland, The Netherlands
- Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, Zuid-Holland, The Netherlands
| |
Collapse
|
181
|
Lessons Learned from Proteasome Inhibitors, the Paradigm for Targeting Protein Homeostasis in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1243:147-162. [PMID: 32297217 DOI: 10.1007/978-3-030-40204-4_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Targeting aberrant protein homeostasis (proteostasis) in cancer is an attractive therapeutic strategy. However, this approach has thus far proven difficult to bring to clinical practice, with one major exception: proteasome inhibition. These small molecules have dramatically transformed outcomes for patients with the blood cancer multiple myeloma. However, these agents have failed to make an impact in more common solid tumors. Major questions remain about whether this therapeutic strategy can be extended to benefit even more patients. Here we discuss the role of the proteasome in normal and tumor cells, the basic, preclinical, and clinical development of proteasome inhibitors, and mechanisms proposed to govern both intrinsic and acquired resistance to these drugs. Years of study of both the mechanism of action and modes of resistance to proteasome inhibitors reveal these processes to be surprisingly complex. Here, we attempt to draw lessons from experience with proteasome inhibitors that may be relevant for other compounds targeting proteostasis in cancer, as well as extending the reach of proteasome inhibitors beyond blood cancers.
Collapse
|
182
|
Therapeutic Monoclonal Antibodies and Antibody Products: Current Practices and Development in Multiple Myeloma. Cancers (Basel) 2019; 12:cancers12010015. [PMID: 31861548 PMCID: PMC7017131 DOI: 10.3390/cancers12010015] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/26/2022] Open
Abstract
Immunotherapy is the latest innovation for the treatment of multiple myeloma (MM). Monoclonal antibodies (mAbs) entered the clinical practice and are under evaluation in clinical trials. MAbs can target highly selective and specific antigens on the cell surface of MM cells causing cell death (CD38 and CS1), convey specific cytotoxic drugs (antibody-drug conjugates), remove the breaks of the immune system (programmed death 1 (PD-1) and PD-ligand 1/2 (L1/L2) axis), or boost it against myeloma cells (bi-specific mAbs and T cell engagers). Two mAbs have been approved for the treatment of MM: the anti-CD38 daratumumab for newly-diagnosed and relapsed/refractory patients and the anti-CS1 elotuzumab in the relapse setting. These compounds are under investigation in clinical trials to explore their synergy with other anti-MM regimens, both in the front-line and relapse settings. Other antibodies targeting various antigens are under evaluation. B cell maturation antigens (BCMAs), selectively expressed on plasma cells, emerged as a promising target and several compounds targeting it have been developed. Encouraging results have been reported with antibody drug conjugates (e.g., GSK2857916) and bispecific T cell engagers (BiTEs®), including AMG420, which re-directs T cell-mediated cytotoxicity against MM cells. Here, we present an overview on mAbs currently approved for the treatment of MM and promising compounds under investigation.
Collapse
|
183
|
Moreau P, Dimopoulos MA, Yong K, Mikhael J, Risse ML, Asset G, Martin T. Isatuximab plus carfilzomib/dexamethasone versus carfilzomib/dexamethasone in patients with relapsed/refractory multiple myeloma: IKEMA Phase III study design. Future Oncol 2019; 16:4347-4358. [PMID: 31833394 DOI: 10.2217/fon-2019-0431] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although the treatment of relapsed/refractory multiple myeloma has improved dramatically over the past decade, the disease remains incurable; therefore, additional therapies are needed. Novel combination therapies incorporating monoclonal antibodies have shown significant promise. Here we describe the design of a Phase III study (NCT03275285, IKEMA), which is evaluating isatuximab plus carfilzomib and low-dose dexamethasone, versus carfilzomib/dexamethasone in relapsed/refractory multiple myeloma. The primary end point is progression-free survival. Responses are being determined by an independent review committee using 2016 International Myeloma Working Group criteria, and safety will be assessed throughout. The first patient was recruited in November 2017, and the last patient was recruited in March 2019; 302 patients have been randomized, and the study is ongoing. Clinical trial registration: NCT03275285.
Collapse
Affiliation(s)
- Philippe Moreau
- Department of Hematology, University Hospital, Allée de l'Ile Gloriette, Nantes, 44093, France
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National & Kapodistrian University of Athens, Stadiou 5 10562 Athens, Greece
| | - Kwee Yong
- Department of Hematology, University College London, London, UK
| | - Joseph Mikhael
- Department of Medicine, Translational Genomics Research Institute, City of Hope Cancer Center, Phoenix, AZ, AZ 85028, USA
| | | | | | - Thomas Martin
- Department of Hematology, University of California at San Francisco, San Francisco, CA, CA 94143, USA
| |
Collapse
|
184
|
Manasanch EE. What to do with minimal residual disease testing in myeloma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:137-141. [PMID: 31808833 PMCID: PMC6913484 DOI: 10.1182/hematology.2019000080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The role and use of minimal residual disease (MRD) testing has changed significantly over the past few years as it has become part of the routine care for response assessment in multiple myeloma. The most widely used standardized methods to assess MRD in myeloma in the bone marrow are multicolor flow cytometry and next-generation sequencing. Importantly, the depth of MRD negativity in the bone marrow correlates with improved progression-free survival and overall survival in myeloma. Whole-body position emission tomography-computed tomography and magnetic resonance imaging are also used to evaluate patchy and extramedullary disease, which may not be readily visible through bone marrow assessment. This article reviews a clinical case in which MRD testing, both in bone marrow and in functional imaging, is part of the standard of care. It also reviews the different modalities of MRD testing and current practice guidelines. Finally, patients with myeloma may be tested for MRD after treatment because this is part of the routine response assessment according to International Myeloma Working Group criteria and correlates with clinical outcomes. Important questions such as when to stop therapy for sustained MRD-negative patients or whether to change treatments for patients who go from MRD negative to positive without other evidence of disease relapse are being evaluated in clinical trials and remain controversial.
Collapse
Affiliation(s)
- Elisabet E Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
185
|
Leng S, Bhutani D, Lentzsch S. How I treat a refractory myeloma patient who is not eligible for a clinical trial. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:125-136. [PMID: 31808850 PMCID: PMC6913488 DOI: 10.1182/hematology.2019000016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Myeloma patients not eligible for clinical trials have many treatment options. Choosing the next best therapy starts with careful assessment of the biology and dynamics of the disease at relapse, as well as the condition and situation of the patient. Fit patients should be considered for triplet regimens, whereas intermediate and frail patients warrant dose-reduced triplets or doublets. An indolent serologic relapse may be treated with dose intensification, especially in a maintenance situation, whereas a rapid relapse requires a more aggressive approach with drug class change or a second-generation immunomodulatory drug (IMID) or proteasome inhibitor (PI). Monoclonal antibodies, in combination with PIs and IMIDs, have proven highly efficacious in early and late relapse. Key elements of supportive care include infection prevention, bone health, thromboprophylaxis, and management of active symptoms, such as pain and distress.
Collapse
Affiliation(s)
- Siyang Leng
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Divaya Bhutani
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Suzanne Lentzsch
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
186
|
Lin Q, Zhao J, Song Y, Liu D. Recent updates on CAR T clinical trials for multiple myeloma. Mol Cancer 2019; 18:154. [PMID: 31684964 PMCID: PMC6829852 DOI: 10.1186/s12943-019-1092-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 10/18/2019] [Indexed: 12/22/2022] Open
Abstract
Proteasome inhibitors, immunomodulatory agents and monoclonal antibodies have dramatically changed the natural history of multiple myeloma (MM). However, most patients eventually suffer a relapse and succumb to the disease. Chimeric antigen receptor (CAR) engineered T cells targeting B cell maturation antigen (BCMA), CD138, CS1 glycoprotein antigen (SLAMF7) and light chains are in active development for therapy of refractory /relapsed (RR) MM. CD19- targeted CAR T cells in conjunction with autologous stem cell transplantation also showed activity in RRMM. Dual- target CAR T cells are in clinical trials for RRMM. This review summarized the recent updates of ongoing CAR T clinical trials for multiple myeloma.
Collapse
Affiliation(s)
- Quande Lin
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Juanjuan Zhao
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Yongping Song
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Delong Liu
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| |
Collapse
|
187
|
Eleutherakis-Papaiakovou E, Gavriatopoulou M, Ntanasis-Stathopoulos I, Kastritis E, Terpos E, Dimopoulos MA. Elotuzumab in combination with pomalidomide and dexamethasone for the treatment of multiple myeloma. Expert Rev Anticancer Ther 2019; 19:921-928. [DOI: 10.1080/14737140.2019.1685879] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
188
|
Monoclonal Antibody Therapies in Multiple Myeloma: A Challenge to Develop Novel Targets. JOURNAL OF ONCOLOGY 2019; 2019:6084012. [PMID: 31781214 PMCID: PMC6875016 DOI: 10.1155/2019/6084012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022]
Abstract
The treatment options in multiple myeloma (MM) has changed dramatically over the past decade with the development of novel agents such as proteasome inhibitors (PIs); bortezomib and immunomodulatory drugs (IMiDs); thalidomide, and lenalidomide which revealed high efficacy and improvement of overall survival (OS) in MM patients. However, despite these progresses, most patients relapse and become eventually refractory to these therapies. Thus, the development of novel, targeted immunotherapies has been pursued aggressively. Recently, next-generation PIs; carfilzomib and ixazomib, IMiD; pomalidomide, histone deacetylase inhibitor (HDADi); panobinostat and monoclonal antibodies (MoAbs); and elotuzumab and daratumumab have emerged, and especially, combination of mAbs plus novel agents has led to dramatic improvements in the outcome of MM patients. The field of immune therapies has been accelerating in the treatment of hematological malignancies and has also taken center stage in MM. This review focuses on an overview of current status of novel MoAb therapy including bispecific T-cell engager (BiTE) antibody (BsAb), antibody-drug conjugate (ADC), and chimeric antigen receptor (CAR) T cells, in relapsed or refractory MM (RRMM). Lastly, investigational novel MoAb-based therapy to overcome immunotherapy resistance in MM is shown.
Collapse
|
189
|
O'Steen S, Comstock ML, Orozco JJ, Hamlin DK, Wilbur DS, Jones JC, Kenoyer A, Nartea ME, Lin Y, Miller BW, Gooley TA, Tuazon SA, Till BG, Gopal AK, Sandmaier BM, Press OW, Green DJ. The α-emitter astatine-211 targeted to CD38 can eradicate multiple myeloma in a disseminated disease model. Blood 2019; 134:1247-1256. [PMID: 31395601 PMCID: PMC6788008 DOI: 10.1182/blood.2019001250] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/31/2019] [Indexed: 12/30/2022] Open
Abstract
Minimal residual disease (MRD) has become an increasingly prevalent and important entity in multiple myeloma (MM). Despite deepening responses to frontline therapy, roughly 75% of MM patients never become MRD-negative to ≤10-5, which is concerning because MRD-negative status predicts significantly longer survival. MM is highly heterogeneous, and MRD persistence may reflect survival of isolated single cells and small clusters of treatment-resistant subclones. Virtually all MM clones are exquisitely sensitive to radiation, and the α-emitter astatine-211 (211At) deposits prodigious energy within 3 cell diameters, which is ideal for eliminating MRD if effectively targeted. CD38 is a proven MM target, and we conjugated 211At to an anti-CD38 monoclonal antibody to create an 211At-CD38 therapy. When examined in a bulky xenograft model of MM, single-dose 211At-CD38 at 15 to 45 µCi at least doubled median survival of mice relative to untreated controls (P < .003), but no mice achieved complete remission and all died within 75 days. In contrast, in a disseminated disease model designed to reflect low-burden MRD, 3 studies demonstrated that single-dose 211At-CD38 at 24 to 45 µCi produced sustained remission and long-term survival (>150 days) for 50% to 80% of mice, where all untreated mice died in 20 to 55 days (P < .0001). Treatment toxicities were transient and minimal. These data suggest that 211At-CD38 offers the potential to eliminate residual MM cell clones in low-disease-burden settings, including MRD. We are optimistic that, in a planned clinical trial, addition of 211At-CD38 to an autologous stem cell transplant (ASCT) conditioning regimen may improve ASCT outcomes for MM patients.
Collapse
Affiliation(s)
- Shyril O'Steen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Melissa L Comstock
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Johnnie J Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Donald K Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, WA; and
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, WA; and
| | - Jon C Jones
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Aimee Kenoyer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Margaret E Nartea
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Yukang Lin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Brian W Miller
- Department of Radiation Oncology, School of Medicine, University of Colorado, Aurora, CO
| | - Theodore A Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Sherilyn A Tuazon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Ajay K Gopal
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Brenda M Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| | - Damian J Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine and
| |
Collapse
|
190
|
Mateos MV, Sonneveld P, Hungria V, Nooka AK, Estell JA, Barreto W, Corradini P, Min CK, Medvedova E, Weisel K, Chiu C, Schecter JM, Amin H, Qin X, Ukropec J, Kobos R, Spencer A. Daratumumab, Bortezomib, and Dexamethasone Versus Bortezomib and Dexamethasone in Patients With Previously Treated Multiple Myeloma: Three-year Follow-up of CASTOR. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 20:509-518. [PMID: 32482541 DOI: 10.1016/j.clml.2019.09.623] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/23/2019] [Accepted: 09/29/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND In the phase III CASTOR study in relapsed or refractory multiple myeloma, daratumumab, bortezomib, and dexamethasone (D-Vd) demonstrated significant clinical benefit versus Vd alone. Outcomes after 40.0 months of median follow-up are discussed. PATIENTS AND METHODS Eligible patients had received ≥ 1 line of treatment and were administered bortezomib (1.3 mg/m2) and dexamethasone (20 mg) for 8 cycles with or without daratumumab (16 mg/kg) until disease progression. RESULTS Of 498 patients in the intent-to-treat (ITT) population (D-Vd, n = 251; Vd, n = 247), 47% had 1 prior line of treatment (1PL; D-Vd, n = 122; Vd, n = 113). Median progression-free survival (PFS) was significantly prolonged with D-Vd versus Vd in the ITT population (16.7 vs. 7.1 months; hazard ratio [HR], 0.31; 95% confidence interval [CI], 0.25-0.40; P < .0001) and the 1PL subgroup (27.0 vs. 7.9 months; HR, 0.22; 95% CI, 0.15-0.32; P < .0001). In lenalidomide-refractory patients, the median PFS was 7.8 versus 4.9 months (HR, 0.44; 95% CI, 0.28-0.68; P = .0002) for D-Vd (n = 60) versus Vd (n = 81). Minimal residual disease (MRD)-negativity rates (10-5) were greater with D-Vd versus Vd (ITT: 14% vs. 2%; 1PL: 20% vs. 3%; both P < .0001). PFS2 was significantly prolonged with D-Vd versus Vd (ITT: HR, 0.48; 95% CI, 0.38-0.61; 1PL: HR, 0.35; 95% CI, 0.24-0.51; P < .0001). No new safety concerns were observed. CONCLUSION After 3 years, D-Vd maintained significant benefits in patients with relapsed or refractory multiple myeloma with a consistent safety profile. D-Vd provided the greatest benefit at first relapse and increased MRD-negativity rates.
Collapse
Affiliation(s)
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC, Rotterdam, The Netherlands
| | - Vania Hungria
- Irmandade Da Santa Casa De Misericordia De São Paulo, São Paulo, Brazil
| | - Ajay K Nooka
- Winship Cancer Institute, Emory University, Atlanta, GA
| | - Jane A Estell
- Haematology Department, Concord Cancer Centre, Concord Hospital, University of Sydney, Concord NSW, Australia
| | | | - Paolo Corradini
- Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy
| | - Chang-Ki Min
- Seoul St. Mary's Hospital, Seoul, The Republic of Korea
| | | | - Katja Weisel
- University Medical Center of Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Himal Amin
- Janssen Research & Development, LLC, Raritan, NJ
| | - Xiang Qin
- Janssen Research & Development, LLC, Spring House, PA
| | - Jon Ukropec
- Janssen Research & Development, LLC, Spring House, PA
| | - Rachel Kobos
- Janssen Research & Development, LLC, Spring House, PA
| | - Andrew Spencer
- Malignant Haematology and Stem Cell Transplantation Service, Alfred Health-Monash University, Melbourne, Australia.
| |
Collapse
|
191
|
Baertsch MA, Lutz R, Raab MS, Weinhold N, Goldschmidt H. Meeting report of the 7th Heidelberg Myeloma Workshop: today and tomorrow. J Cancer Res Clin Oncol 2019; 145:2445-2455. [PMID: 31407112 DOI: 10.1007/s00432-019-02998-w] [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: 06/09/2019] [Accepted: 08/05/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE The 7th Heidelberg Myeloma Workshop was held on April 5th and 6th, 2019 at the University Hospital Heidelberg. METHODS AND RESULTS Main topics of the meeting were (1) diagnostics and prognostic factors, (2) role of immunotherapy in multiple myeloma (MM), (3) current therapy of MM, (4) biology and genomics of MM as well as (5) novel treatment concepts. A debate on the status of minimal residual disease (MRD) driven therapy was held. CONCLUSION Diagnostics and treatment of newly diagnosed and relapsed MM are continuously evolving. While advances in the field of (single cell) genetic analysis now allow for characterization of the disease at an unprecedented resolution, immunotherapeutic approaches and MRD testing are at the forefront of the current clinical trial landscape.
Collapse
Affiliation(s)
- M A Baertsch
- Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany.
| | - R Lutz
- Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - M S Raab
- Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - N Weinhold
- Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - H Goldschmidt
- Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
192
|
NK Cells in the Treatment of Hematological Malignancies. J Clin Med 2019; 8:jcm8101557. [PMID: 31569769 PMCID: PMC6832953 DOI: 10.3390/jcm8101557] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells have the innate ability to kill cancer cells, however, tumor cells may acquire the capability of evading the immune response, thereby leading to malignancies. Restoring or potentiation of this natural antitumor activity of NK cells has become a relevant therapeutic approach in cancer and, particularly, in hematological cancers. The use of tumor-specific antibodies that promote antibody-dependent cell-mediated cytotoxicity (ADCC) through the ligation of CD16 receptor on NK cells has become standard for many hematologic malignancies. Hematopoietic stem cell transplantation is another key therapeutic strategy that harnesses the alloreactivity of NK cells against cancer cells. This strategy may be refined by adoptive transfer of NK cells that may be previously expanded, activated, or redirected (chimeric antigen receptor (CAR)-NK cells) against cancer cells. The antitumor activity of NK cells can also be boosted by cytokines or immunostimulatory drugs such as lenalidomide or pomalidomide. Finally, targeting immunosubversive mechanisms developed by hematological cancers and, in particular, using antibodies that block NK cell inhibitory receptors and checkpoint proteins are novel promising therapeutic approaches in these malignant diseases.
Collapse
|
193
|
Kortüm KM, Einsele H. [Diagnostic and therapeutic considerations on recurrence of multiple myeloma : A current overview]. Internist (Berl) 2019; 60:34-41. [PMID: 30536028 DOI: 10.1007/s00108-018-0528-9] [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: 10/27/2022]
Abstract
The treatment of multiple myeloma remains in a state of profound change. Over the past decades both disease-free survival and overall survival have been significantly prolonged by the approval of new drugs; however, despite high response rates and achievement of deep responses to primary treatment, recurrence of the disease is still expected in nearly all patients treated. Fortunately, good treatment options for myeloma patients in relapse are also currently available and the possible combinations of approved substances are numerous. Patient-specific criteria, such as primary response, comorbidities and treatment-associated toxicity can thus be taken into account more frequently in the selection of a suitable treatment of recurrences; however, the lack of comparative studies of new substances and extensive interindividual disease heterogeneity continue to make it difficult to select the best treatment of recurrence in a specific case. Therefore, the treatment of recurrence of multiple myeloma, especially for patients with high-risk features, remains a clinical challenge. This review article deliberately dispenses with the commonly used combination of mere study results and a more practical approach should be taught for the rational planning of treatment for recurrent multiple myeloma. This includes new insights into tumor evolution and taking current developments in the drug treatment of multiple myeloma into account.
Collapse
Affiliation(s)
- K M Kortüm
- Medizinische Klinik 2, Universitätsklinikum Würzburg, Oberdürrbacher Str. 6, 97074, Würzburg, Deutschland.
| | - H Einsele
- Medizinische Klinik 2, Universitätsklinikum Würzburg, Oberdürrbacher Str. 6, 97074, Würzburg, Deutschland
| |
Collapse
|
194
|
Hollmann S, Moldaver D, Goyert N, Grima D, Maiese EM. Letter--The Authors Respond. J Manag Care Spec Pharm 2019; 25:1028-1030. [PMID: 31456500 PMCID: PMC10398007 DOI: 10.18553/jmcp.2019.25.9.1028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
DISCLOSURES No additional funding was received for the writing of this letter. The published study referred to in this letter was funded by Janssen Scientific Affairs, which employs Maiese and funded Cornerstone Research Group, a health economic consulting group, to conduct the study. Grima is a founding partner of Cornerstone Research Group, which employs Hollmann, Goyert, and Moldaver.
Collapse
Affiliation(s)
| | | | - Nik Goyert
- Cornerstone Research Group Burlington, Ontario, Canada
| | - Daniel Grima
- Cornerstone Research Group Burlington, Ontario, Canada
| | | |
Collapse
|
195
|
Relapsed refractory multiple myeloma: a comprehensive overview. Leukemia 2019; 33:2343-2357. [PMID: 31455853 DOI: 10.1038/s41375-019-0561-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/28/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023]
Abstract
Most patients with relapsed/refractory multiple myeloma (RRMM) have been treated with drug combinations including a proteasome inhibitor (PI) and/or an immunomodulatory drug (IMiD). The goal of therapy for such patients is therefore to achieve disease control with acceptable toxicity and patient-defined decent quality of life. Physicians face a difficult task not only deciding who to treat, but also when to treat and how to treat, utilizing knowledge of previously administered therapies, patient comorbidities, potential adverse events, and patient wishes to make such a critical decision. New drugs and combination regimens are continuously underway thus broadening the options for therapy and giving way to a more individualized approach for patients with RRMM. The integration of novel agents into the treatment paradigm has shifted the perception of multiple myeloma (MM) from an incurable, fatal disease to a manageable, chronic one. This comprehensive review addresses the results and challenges posed by many of the newer agents for the treatment of RRMM. It attempts to propose a universal strategy for optimal therapy decision-making thus answering three simple fundamental questions-when to treat, how to treat, and how long to treat for.
Collapse
|
196
|
Lovas S, Varga G, Farkas P, Masszi T, Wohner N, Bereczki Á, Adamkovich N, Borbényi Z, Szomor Á, Alizadeh H, Szaleczky E, Wolf K, Schneider T, Plander M, Szendrei T, Csacsovszki O, Csukly Z, Rajnics P, Egyed M, Nagy Z, Rejtő L, Illés Á, Mikala G, Váróczy L. Real-world data on the efficacy and safety of daratumumab treatment in Hungarian relapsed/refractory multiple myeloma patients. Int J Hematol 2019; 110:559-565. [PMID: 31392600 DOI: 10.1007/s12185-019-02715-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/02/2019] [Accepted: 08/02/2019] [Indexed: 01/08/2023]
Abstract
Daratumumab is a human anti-CD38 monoclonal antibody used in the treatment of refractory and relapsed multiple myeloma. We investigated the efficacy and safety of daratumumab therapy in a real-world setting. Ninety-nine Hungarian patients were included; 48 received monotherapy, while lenalidomide and bortezomib combinations were administered in 29 and 19 cases, respectively. Overall response rate was assessable in 88 patients, with 12 complete, 10 very good partial, 34 partial, and seven minor responses. At a median duration of follow-up of 18.6 months, median progression-free survival (PFS) among all patients was 17.0 months. These values were inferior in the bortezomib combination and monotherapy groups. Patients with early-stage disease (ISS1) had better survival results than those with stage 2 or 3 myeloma (p = 0.009). Heavily pretreated patients had inferior PFS compared to those with 1-3 therapies (p = 0.035). Patients with impaired renal function had PFS results comparable with those having no kidney involvement. There were 10 fatal infections, and the most frequent adverse events were mild infusion-associated reactions and hematologic toxicities. Our results confirm that daratumumab is an effective treatment option for relapsed/refractory MM with an acceptable safety profile in patients with normal and impaired renal function.
Collapse
Affiliation(s)
- Szilvia Lovas
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Gergely Varga
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Farkas
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Masszi
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Nikolett Wohner
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Ágnes Bereczki
- 2nd Department of Internal Medicine and Cardiology Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Nóra Adamkovich
- 2nd Department of Internal Medicine and Cardiology Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Zita Borbényi
- 2nd Department of Internal Medicine and Cardiology Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Árpád Szomor
- 1st Department of Internal Medicine, University of Pécs, Pecs, Hungary
| | - Hussain Alizadeh
- 1st Department of Internal Medicine, University of Pécs, Pecs, Hungary
| | | | | | | | - Márk Plander
- Department of Hematology, Markusovszky Teaching Hospital, Szombathely, Hungary
| | - Tamás Szendrei
- Department of Hematology, Markusovszky Teaching Hospital, Szombathely, Hungary
| | - Ottó Csacsovszki
- Department of Hematology and Stem Cell Transplantation, South-Pest Central Hospital, National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Zoltán Csukly
- Department of Hematology and Stem Cell Transplantation, South-Pest Central Hospital, National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Péter Rajnics
- Department of Hematology, Mór Kaposi Teaching Hospital, Kaposvár, Hungary
| | - Miklós Egyed
- Department of Hematology, Mór Kaposi Teaching Hospital, Kaposvár, Hungary
| | - Zsolt Nagy
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - László Rejtő
- Department of Hematology, Jósa András Teaching Hospital, Nyíregyháza, Hungary
| | - Árpád Illés
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary
| | - Gábor Mikala
- Department of Hematology and Stem Cell Transplantation, South-Pest Central Hospital, National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - László Váróczy
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., 4032, Debrecen, Hungary.
| |
Collapse
|
197
|
Bal S, Weaver A, Cornell RF, Costa LJ. Challenges and opportunities in the assessment of measurable residual disease in multiple myeloma. Br J Haematol 2019; 186:807-819. [PMID: 31364160 DOI: 10.1111/bjh.16130] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Treatment response assessment in multiple myeloma (MM) relies on the detection of paraprotein in serum and/or urine, bone marrow morphology and immunohistochemistry. With remarkable advances in therapy, particularly in the newly diagnosed setting, achievement of complete remission became frequent, creating the need to identify smaller amounts of residual disease and understand their prognostic and therapeutic implications. Measurable residual disease (MRD) can be assessed primarily by flow cytometry and next generation sequencing and state-of-the-art assays have sensitivity approaching 1 in 106 cells. This review discusses the existing challenges in utilizing MRD to inform management of MM and highlights open research questions and opportunities as MRD is more routinely incorporated into clinical practice for patients with MM.
Collapse
Affiliation(s)
- Susan Bal
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allison Weaver
- Division of Hematology and Oncology, Vanderbilt University, Nashville, TN, USA
| | - Robert F Cornell
- Division of Hematology and Oncology, Vanderbilt University, Nashville, TN, USA
| | - Luciano J Costa
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
198
|
|
199
|
Hofmeister CC, Nooka A, Kaufman JL, Lonial S. Registering a CD38 antibody upfront for multiple myeloma. Lancet 2019; 394:3-4. [PMID: 31171418 DOI: 10.1016/s0140-6736(19)31250-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/13/2019] [Indexed: 11/26/2022]
Affiliation(s)
| | - Ajay Nooka
- Winship Cancer Institute of Emory University, Atlanta, GA 30307, USA
| | | | - Sagar Lonial
- Winship Cancer Institute of Emory University, Atlanta, GA 30307, USA
| |
Collapse
|
200
|
Moreau P, Attal M, Hulin C, Arnulf B, Belhadj K, Benboubker L, Béné MC, Broijl A, Caillon H, Caillot D, Corre J, Delforge M, Dejoie T, Doyen C, Facon T, Sonntag C, Fontan J, Garderet L, Jie KS, Karlin L, Kuhnowski F, Lambert J, Leleu X, Lenain P, Macro M, Mathiot C, Orsini-Piocelle F, Perrot A, Stoppa AM, van de Donk NW, Wuilleme S, Zweegman S, Kolb B, Touzeau C, Roussel M, Tiab M, Marolleau JP, Meuleman N, Vekemans MC, Westerman M, Klein SK, Levin MD, Fermand JP, Escoffre-Barbe M, Eveillard JR, Garidi R, Ahmadi T, Zhuang S, Chiu C, Pei L, de Boer C, Smith E, Deraedt W, Kampfenkel T, Schecter J, Vermeulen J, Avet-Loiseau H, Sonneveld P. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet 2019; 394:29-38. [PMID: 31171419 DOI: 10.1016/s0140-6736(19)31240-1] [Citation(s) in RCA: 685] [Impact Index Per Article: 114.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/13/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Bortezomib, thalidomide, and dexamethasone (VTd) plus autologous stem-cell transplantation is standard treatment in Europe for transplant-eligible patients with newly diagnosed multiple myeloma. We evaluated whether the addition of daratumumab to VTd before and after autologous stem-cell transplantation would improve stringent complete response rate in patients with newly diagnosed multiple myeloma. METHODS In this two-part, randomised, open-label, phase 3 CASSIOPEIA trial, we recruited transplant-eligible patients with newly diagnosed multiple myeloma at 111 European sites. Patients were randomly assigned (1:1) to receive four pre-transplant induction and two post-transplant consolidation cycles of VTd alone (VTd group) or in combination with daratumumab (D-VTd group). The primary endpoint of part 1 was stringent complete response assessed 100 days after transplantation. Part 2 (maintenance) is ongoing. The trial is registered with ClinicalTrials.gov, number NCT02541383. FINDINGS Between Sept 22, 2015, and Aug 1, 2017, 1085 patients were enrolled at 111 European sites and were randomly assigned to the D-VTd group (n=543) or the VTd group (n=542). At day 100 after transplantation, 157 (29%) of 543 patients in the D-VTd group and 110 (20%) of 542 patients in the VTd group in the intention-to-treat population had achieved a stringent complete response (odds ratio 1·60, 95% CI 1·21-2·12, p=0·0010). 211 (39%) patients in the D-VTd group versus 141 (26%) in the VTd group achieved a complete response or better, and 346 (64%) of 543 versus 236 (44%) of 542 achieved minimal residual disease-negativity (10-5 sensitivity threshold, assessed by multiparametric flow cytometry; both p<0·0001). Median progression-free survival from first randomisation was not reached in either group (hazard ratio 0·47, 95% CI 0·33-0·67, p<0·0001). 46 deaths on study were observed (14 vs 32, 0·43, 95% CI 0·23-0·80). The most common grade 3 or 4 adverse events were neutropenia (28% vs 15%), lymphopenia (17% vs 10%), and stomatitis (13% vs 16%). INTERPRETATION D-VTd before and after autologous stem-cell transplantation improved depth of response and progression-free survival with acceptable safety. CASSIOPEIA is the first study showing the clinical benefit of daratumumab plus standard of care in transplant-eligible patients with newly diagnosed multiple myeloma. FUNDING The Intergroupe Francophone du Myélome and Dutch-Belgian Cooperative Trial Group for Hematology Oncology.
Collapse
Affiliation(s)
| | - Michel Attal
- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Cyrille Hulin
- Department of Hematology, Hopital Haut Leveque, University Hospital Bordeaux, Bordeaux, France
| | | | | | - Lotfi Benboubker
- Hôpital de Bretonneau, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Marie C Béné
- Hematology Biology, University Hospital Hôtel Dieu, Nantes, France
| | - Annemiek Broijl
- Erasmus University Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Hélène Caillon
- Biochemistry Laboratory, Hospital of Nantes, Nantes, France
| | - Denis Caillot
- Hôpital Du Bocage, Centre Hospitalier Universitaire Dijon, Dijon, France
| | - Jill Corre
- Unite de Genomique du Myelome, Institut universitaire du cancer de Toulouse Oncopole, Toulouse, France
| | | | - Thomas Dejoie
- Biochemistry Laboratory, Hospital of Nantes, Nantes, France
| | - Chantal Doyen
- Centre Hospitalier Universitaire UCL Namur-site Godinne, Yvoir, Belgium
| | - Thierry Facon
- Service des Maladies du Sang, Hôpital Claude Huriez, Lille, France
| | | | | | - Laurent Garderet
- Team Proliferation and Differentiation of Stem Cells, Centre de Recherche Saint-Antoine, Inserm, Sorbonne Université, Paris, France; Département d'Hématologie et de Thérapie Cellulaire, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Lionel Karlin
- Service d'Hématologie Clinique, Centre Hospitalier Lyon-Sud, Lyon, France
| | | | - Jérôme Lambert
- Biostatistical Department, Hôpital Saint Louis, Paris, France
| | - Xavier Leleu
- Hôpital la Milétrie, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Pascal Lenain
- Centre de Lutte Contre le Cancer-Centre Henri Becquerel, Rouen, France
| | | | | | | | - Aurore Perrot
- Hematology Department, Vandoeuvre Les Nancy, University Hospitals Nancy, France
| | | | - Niels Wcj van de Donk
- Department of Hematology, University Medical Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Soraya Wuilleme
- Hematology Biology, University Hospital Hôtel Dieu, Nantes, France
| | - Sonja Zweegman
- Department of Hematology, University Medical Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Brigitte Kolb
- Hôpital Robert Debré, Centre Hospitalier Universitaire de Reims, Reims, France
| | | | - Murielle Roussel
- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Mourad Tiab
- Centre Hospitalier Départemental Vendée, La Roche sur Yon, France
| | | | - Nathalie Meuleman
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | - Jean Paul Fermand
- Service d'Immuno-Hématologie, Département d'Immunologie Clinique, Inserm and Intergroupe Francophone du Myélome, Hôpital Saint-Louis, Paris, France
| | | | | | - Reda Garidi
- Hospital Center de Saint-Quentin, Saint Quentin, France
| | | | - Sen Zhuang
- Janssen Research & Development, Raritan, NJ, USA
| | | | - Lixia Pei
- Janssen Research & Development, Raritan, NJ, USA
| | - Carla de Boer
- Janssen Research & Development, LLC, Leiden, Netherlands
| | - Elena Smith
- Janssen Research & Development, LLC, High Wycombe, UK
| | | | | | | | | | - Hervé Avet-Loiseau
- Unite de Genomique du Myelome, Institut universitaire du cancer de Toulouse Oncopole, Toulouse, France
| | - Pieter Sonneveld
- Erasmus University Medical Center Cancer Institute, Rotterdam, Netherlands
| |
Collapse
|