1
|
Kintzel PE. Elotuzumab: Empiric analysis of dexamethasone administration schedule. J Oncol Pharm Pract 2024; 30:235-238. [PMID: 37876226 DOI: 10.1177/10781552231207855] [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] [Indexed: 10/26/2023]
Abstract
Elotuzumab in combination with dexamethasone and immunomodulating agents (IMiDs) lenalidomide or pomalidomide is 2nd to 4th line therapy for multiple myeloma. The labelled dosage of dexamethasone for use in conjunction with elotuzumab and IMiDs splits the dexamethasone dose into two administrations, one oral and one intravenous, on the days of each elotuzumab infusion. Administration of split dose dexamethasone on days of elotuzumab administration is based on the registration trials submitted for drug approval and was intended to ensure standard well-timed immunotherapy premedication using pharmacologically equivalent dexamethasone doses for both study arms. Administration of dexamethasone in the manner delineated by the elotuzumab product label adds complexity to the delivery of care. This commentary provides an empirical assessment of established medication safety and effectiveness which supports administration of dexamethasone standard intermittent dose instead of the split dose approach delineated on elotuzumab package insert. Simplification of regimen administration improves medication adherence, reduces the risk of inadvertent omission or duplication of medication therapy, and improves the workflow required for delivery of care.
Collapse
|
2
|
Bourdin V, Bigot W, Vanjak A, Burlacu R, Lopes A, Champion K, Depond A, Amador-Borrero B, Sene D, Comarmond C, Mouly S. Drug-Drug Interactions Involving Dexamethasone in Clinical Practice: Myth or Reality? J Clin Med 2023; 12:7120. [PMID: 38002732 PMCID: PMC10672071 DOI: 10.3390/jcm12227120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Concomitant administration of multiple drugs frequently causes severe pharmacokinetic or pharmacodynamic drug-drug interactions (DDIs) resulting in the possibility of enhanced toxicity and/or treatment failure. The activity of cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp), a drug efflux pump sharing localization and substrate affinities with CYP3A4, is a critical determinant of drug clearance, interindividual variability in drug disposition and clinical efficacy, and appears to be involved in the mechanism of numerous clinically relevant DDIs, including those involving dexamethasone. The recent increase in the use of high doses of dexamethasone during the COVID-19 pandemic have emphasized the need for better knowledge of the clinical significance of drug-drug interactions involving dexamethasone in the clinical setting. We therefore aimed to review the already published evidence for various DDIs involving dexamethasone in vitro in cell culture systems and in vivo in animal models and humans.
Collapse
Affiliation(s)
- Venceslas Bourdin
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - William Bigot
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Anthony Vanjak
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Ruxandra Burlacu
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Amanda Lopes
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Karine Champion
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Audrey Depond
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Blanca Amador-Borrero
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Damien Sene
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM U976, Hôpital Saint-Louis, 75010 Paris, France
| | - Chloe Comarmond
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM U976, Hôpital Saint-Louis, 75010 Paris, France
| | - Stéphane Mouly
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM UMR-S1144, Hôpital Fernand Widal, 75010 Paris, France
| |
Collapse
|
3
|
The genetic heterogeneity and drug resistance mechanisms of relapsed refractory multiple myeloma. Nat Commun 2022; 13:3750. [PMID: 35768438 PMCID: PMC9243087 DOI: 10.1038/s41467-022-31430-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022] Open
Abstract
Multiple myeloma is the second most common hematological malignancy. Despite significant advances in treatment, relapse is common and carries a poor prognosis. Thus, it is critical to elucidate the genetic factors contributing to disease progression and drug resistance. Here, we carry out integrative clinical sequencing of 511 relapsed, refractory multiple myeloma (RRMM) patients to define the disease’s molecular alterations landscape. The NF-κB and RAS/MAPK pathways are more commonly altered than previously reported, with a prevalence of 45–65% each. In the RAS/MAPK pathway, there is a long tail of variants associated with the RASopathies. By comparing our RRMM cases with untreated patients, we identify a diverse set of alterations conferring resistance to three main classes of targeted therapy in 22% of our cohort. Activating mutations in IL6ST are also enriched in RRMM. Taken together, our study serves as a resource for future investigations of RRMM biology and potentially informs clinical management. The genetic factors involved in disease progression and drug resistance in multiple myeloma (MM) are varied and complex. Here, genomic and transcriptomic profiling of 511 relapsed and refractory MM patients reveals genetic alterations in several oncogenic pathways contributing to progression and resistance to MM therapies.
Collapse
|
4
|
Stoeckle JH, Davies FE, Williams L, Boyle EM, Morgan GJ. The evolving role and utility of off-label drug use in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:355-373. [PMID: 36046752 PMCID: PMC9400732 DOI: 10.37349/etat.2021.00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/16/2021] [Indexed: 12/03/2022] Open
Abstract
The treatment landscape for multiple myeloma (MM) has dramatically changed over the last three decades, moving from no US Food and Drug Administration approvals and two active drug classes to over 19 drug approvals and at least eight different active classes. The advances seen in MM therapy have relied on both a structured approach to obtaining new labels and cautious off-label drug use. Although there are country and regional differences in drug approval processes, many of the basic principles behind off-label drug use in MM can be summarized into four main categories: 1) use of a therapy prior to the current approval regulations; 2) widespread use of a therapy following the release of promising clinical trial results but prior to drug approval; 3) use of a cheap therapy supported by clinical safety and efficacy data but without commercial backing; and 4) niche therapies for small well-defined patient populations where large clinical trials with sufficient power may be difficult to perform. This review takes a historical approach to discuss how off-label drug use has helped to shape the current treatment approach for MM.
Collapse
Affiliation(s)
- James H Stoeckle
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Faith E Davies
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Louis Williams
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Eileen M Boyle
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Gareth J Morgan
- Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| |
Collapse
|
5
|
Hofbauer D, Mougiakakos D, Broggini L, Zaiss M, Büttner-Herold M, Bach C, Spriewald B, Neumann F, Bisht S, Nolting J, Zeiser R, Hamarsheh S, Eberhardt M, Vera J, Visentin C, De Luca CMG, Moda F, Haskamp S, Flamann C, Böttcher M, Bitterer K, Völkl S, Mackensen A, Ricagno S, Bruns H. β 2-microglobulin triggers NLRP3 inflammasome activation in tumor-associated macrophages to promote multiple myeloma progression. Immunity 2021; 54:1772-1787.e9. [PMID: 34289378 DOI: 10.1016/j.immuni.2021.07.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/28/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022]
Abstract
As substantial constituents of the multiple myeloma (MM) microenvironment, pro-inflammatory macrophages have emerged as key promoters of disease progression, bone destruction, and immune impairment. We identify beta-2-microglobulin (β2m) as a driver in initiating inflammation in myeloma-associated macrophages (MAMs). Lysosomal accumulation of phagocytosed β2m promotes β2m amyloid aggregation in MAMs, resulting in lysosomal rupture and ultimately production of active interleukin-1β (IL-1β) and IL-18. This process depends on activation of the NLRP3 inflammasome after β2m accumulation, as macrophages from NLRP3-deficient mice lack efficient β2m-induced IL-1β production. Moreover, depletion or silencing of β2m in MM cells abrogates inflammasome activation in a murine MM model. Finally, we demonstrate that disruption of NLRP3 or IL-18 diminishes tumor growth and osteolytic bone destruction normally promoted by β2m-induced inflammasome signaling. Our results provide mechanistic evidence for β2m's role as an NLRP3 inflammasome activator during MM pathogenesis. Moreover, inhibition of NLRP3 represents a potential therapeutic approach in MM.
Collapse
Affiliation(s)
- Daniel Hofbauer
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | | | - Luca Broggini
- Department of Biosciences, University of Milan, Milan, Italy; Institute of Molecular and Translational Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milano, Italy
| | - Mario Zaiss
- Department of Internal Medicine 3, University Hospital Erlangen, Erlangen, Germany
| | | | - Christian Bach
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Bernd Spriewald
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Frank Neumann
- Department of Internal Medicine 1, Saarland University Medical School, Homburg, Germany
| | - Savita Bisht
- Department of Oncology/Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Jens Nolting
- Department of Oncology/Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Robert Zeiser
- Department of Medicine 1, University of Freiburg, Freiburg, Germany
| | | | - Martin Eberhardt
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Julio Vera
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | | | - Chiara Maria Giulia De Luca
- Divisione di Neurologia 5 - Neuropatologia, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fabio Moda
- Divisione di Neurologia 5 - Neuropatologia, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Stefan Haskamp
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Cindy Flamann
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Martin Böttcher
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Katrin Bitterer
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Stefano Ricagno
- Department of Biosciences, University of Milan, Milan, Italy
| | - Heiko Bruns
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany.
| |
Collapse
|
6
|
Burwick N, Sharma S. Glucocorticoids in multiple myeloma: past, present, and future. Ann Hematol 2018; 98:19-28. [PMID: 30073393 DOI: 10.1007/s00277-018-3465-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Glucocorticoids are a backbone of treatment for multiple myeloma in both the upfront and relapsed/refractory setting. While glucocorticoids have single agent activity in multiple myeloma, in the modern era, they are paired with novel agents to induce high clinical response rates. On the other hand, toxicities of steroid therapy limit high dose delivery and impact patient quality of life. We provide a history of steroid use in multiple myeloma with the aim to understand how steroids have emerged and persisted in the treatment of multiple myeloma. We review mechanisms of glucocorticoid sensitivity and resistance and highlight potential future directions to evaluate steroid responsiveness. Further research in this area will aid in optimizing steroid utilization and help determine when glucocorticoid therapy may no longer benefit patients.
Collapse
Affiliation(s)
- Nicholas Burwick
- VA Puget Sound Health Care System, Seattle, WA, USA. .,Department of Medicine, University of Washington, 1705 NE Pacific St, M/S 358280, Seattle, WA, 98195, USA.
| | | |
Collapse
|
7
|
Lee DH, Fradley MG. Cardiovascular Complications of Multiple Myeloma Treatment: Evaluation, Management, and Prevention. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2018; 20:19. [DOI: 10.1007/s11936-018-0618-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
8
|
Sun T, Wang S, Sun H, Wen J, An G, Li J. Improved survival in multiple myeloma, with a diminishing racial gap and a widening socioeconomic status gap over three decades. Leuk Lymphoma 2017; 59:49-58. [PMID: 28595471 DOI: 10.1080/10428194.2017.1335398] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Multiple myeloma (MM) is estimated to have 30,280 new cases and be associated with 12,590 deaths in 2017. However, quantitative analysis for survival, based on a large population, is lacking. Data were extracted from a total of 33,170 cases from nine registry sites in the Surveillance, Epidemiology, and End Results database. The current study shows that the incidence for MM remained relatively stable between 1981 and 2010, with 4.6, 4.7, and 4.7 per 100,000 persons in each decade. In addition, survival for MM improved each decade with a larger increment in the last two decades, with a narrowing survival gap among races and a widening gap among socioeconomic status (SES) groups. The survival gap changes in races and SES groups may guide clinicians to design better treatment protocols and call for the pressing need for health-care policy to fill the gap among SES groups.
Collapse
Affiliation(s)
- Tiantian Sun
- a Department of Hematology , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Shuncong Wang
- b Department of Oncology , The Fifth Affiliated Hospital of Sun Yat-sen University , Zhuhai , China
| | - Huanhuan Sun
- b Department of Oncology , The Fifth Affiliated Hospital of Sun Yat-sen University , Zhuhai , China
| | - Jianguo Wen
- c Department of Pathology and Genomic Medicine , Houston Methodist Hospital , Houston , TX , USA
| | - Gang An
- d State Key Laboratory of Experimental Hematology , Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College , Tianjin , China
| | - Juan Li
- a Department of Hematology , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| |
Collapse
|
9
|
de la Puente P, Azab AK. Nanoparticle delivery systems, general approaches, and their implementation in multiple myeloma. Eur J Haematol 2017; 98:529-541. [PMID: 28208215 DOI: 10.1111/ejh.12870] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 12/25/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy that remains incurable, with relapse rates >90%. The main limiting factor for the effective use of chemotherapies in MM is the serious side effects caused by these drugs. The emphasis in cancer treatment has shifted from cytotoxic, non-specific chemotherapies to molecularly targeted and rationally designed therapies showing greater efficacy and fewer side effects. Traditional chemotherapy has shown several disadvantages such as lack of targeting capabilities, systemic toxicity, and side effects; low therapeutic index, as well as most anticancer drugs, has poor water solubility. Nanoparticle delivery systems (NPs) are capable of targeting large doses of chemotherapies into the target area while sparing healthy tissues, overcoming the limitations of traditional chemotherapy. Here, we review the current state of the art in nanoparticle-based strategies designed to treat MM. Many nanoparticle delivery systems have been studied for myeloma using non-targeted NPs (liposomes, polymeric NPs, and inorganic NPs), triggered NPs, as well as targeted NPs (VLA-4, ABC drug transporters, bone microenvironment targeting). The results in preclinical and clinical studies are promising; however, there remains much to be learned in the emerging field of nanomedicine in myeloma.
Collapse
Affiliation(s)
- Pilar de la Puente
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Abdel Kareem Azab
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Abstract
Despite recent advances, multiple myeloma remains an incurable disease. Induction therapy followed by autologous transplantation has become the standard of care. The idea of maintenance therapy in multiple myeloma is not new. Starting with chemotherapy in 1975, to interferon in 1998, to novel agents recently, a multitude of agents have been explored in patients with multiple myeloma. In spite of the novel agents, multiple myeloma continues to be an incurable disease with the progression-free survival after autologous transplant rarely exceeding 3 years. The goal of using maintenance therapy has been to improve the outcomes following autologous transplantation by increasing the progression-free survival, deepening remissions and perhaps increasing overall survival. It has been shown that patients with a stringent complete response (CR) have a better outcome [Kapoor et al. 2013]. It is becoming increasingly common to check minimal residual disease (MRD) as a means of assessing depth of response. It has also been shown that patients with no MRD have not only a better progression-free survival but also a better overall survival compared with patients who are MRD positive. This makes it even more important to find agents for maintenance therapy, which can further deepen and maintain responses. Here, we present a comprehensive review of the agents studied as maintenance for multiple myeloma and their efficacy, both in terms of overall survival, progression-free survival and toxicity.
Collapse
Affiliation(s)
- Prerna Mewawalla
- West Penn Hospital, 4800 Friendship Ave Suite 2303, Pittsburgh, PA 15224, USA
| | | |
Collapse
|
11
|
Abstract
Background. Multiple myeloma accounts for 10% of all haematologic malignancies worldwide. In Europe, over 10 000 new cases and nearly 8000 deaths were attributed to multiple myeloma in 2000. Unlike other malignancies, in which surgery and radiation are important treatment modalities, myeloma is exclusively treated with stem cell transplantation and drug therapy, requiring pharmacists to stay abreast of new developments. The melphalan-prednisolone and vincristine-doxorubicin-dexamethasone (VAD) regimens, which have been standard treatments for multiple myeloma over the past few decades, have yielded responses without real survival benefits. Transplantation utilizing high-dose chemotherapy has produced the only meaningful survival benefits for patients with multiple myeloma, but many patients are not candidates for this aggressive treatment option. More effective therapies for multiple myeloma are needed. Objective. To address the mechanisms of action, safety, and efficacy of novel approaches to the treatment of myeloma involving bortezomib, thalidomide and its analogues, lenalidomide and CC-4047 (ActimidTM), and arsenic trioxide as single agents or in combination regimens. Data sources. Published preclinical and primary clinical trial results, as well as scientific or clinical meeting abstracts. The author determined the relevance and subsequent inclusion of the data. Conclusions. Bortezomib is approved in the US and Europe as single-agent therapy for the treatment of relapsed or refractory multiple myeloma. Thalidomide, its analogues, and arsenic trioxide have demonstrated activity and are under investigation in this disease. Further clinical trials of the efficacy and toxicity of these novel agents are ongoing and will further define optimal combinations and sequencing with conventional therapies.
Collapse
|
12
|
Krishnan SR, Jaiswal R, Brown RD, Luk F, Bebawy M. Multiple myeloma and persistence of drug resistance in the age of novel drugs (Review). Int J Oncol 2016; 49:33-50. [PMID: 27175906 DOI: 10.3892/ijo.2016.3516] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/17/2015] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is a mature B cell neoplasm that results in multi-organ failure. The median age of onset, diverse clinical manifestations, heterogeneous survival rate, clonal evolution, intrinsic and acquired drug resistance have impact on the therapeutic management of the disease. Specifically, the emergence of multidrug resistance (MDR) during the course of treatment contributes significantly to treatment failure. The introduction of the immunomodulatory agents and proteasome inhibitors has seen an increase in overall patient survival, however, for the majority of patients, relapse remains inevitable with evidence that these agents, like the conventional chemotherapeutics are also subject to the development of MDR. Clinical management of patients with MM is currently compromised by lack of a suitable procedure to monitor the development of clinical drug resistance in individual patients. The current MM prognostic measures fail to pick the clonotypic tumor cells overexpressing drug efflux pumps, and invasive biopsy is insufficient in detecting sporadic tumors in the skeletal system. This review summarizes the challenges associated with treating the complex disease spectrum of myeloma, with an emphasis on the role of deleterious multidrug resistant clones orchestrating relapse.
Collapse
Affiliation(s)
- Sabna Rajeev Krishnan
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ritu Jaiswal
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ross D Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Frederick Luk
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Mary Bebawy
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| |
Collapse
|
13
|
An update on molecular biology and drug resistance mechanisms of multiple myeloma. Crit Rev Oncol Hematol 2015; 96:413-24. [DOI: 10.1016/j.critrevonc.2015.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 06/11/2015] [Accepted: 07/07/2015] [Indexed: 12/15/2022] Open
|
14
|
Yang X, Levis M. Pulsed-high-dose dexamethasone as a treatment for pure red cell aplasia following ABO-incompatible allogeneic stem cell transplantation. Transfus Med 2015; 24:246-8. [PMID: 25124073 DOI: 10.1111/tme.12143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 06/09/2014] [Accepted: 07/03/2014] [Indexed: 11/29/2022]
Affiliation(s)
- X Yang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
15
|
Abstract
Multiple myeloma continues to be an incurable disease. The understanding of the disease's pathophysiology has significantly improved over the past few years, partly due to the discovery of the role of immunomodulatory agents and the study of their mechanism of action. Thalidomide, the first of the immunomodulatory family to be used in the management of multiple myeloma, proved not only to be effective in the treatment of multiple myeloma, but also instigated a wide range of in vitro and in vivo studies to define the pathophysiology of the plasma cell dyscrasia. The attention thalidomide has received in the past and recent history has not been without a price. The drug has a side-effect profile that, if managed appropriately, provides the most unique active molecule in the management of the disease, where it maintains the same response rate in newly diagnosed patients as in advanced relapsed/refractory multiple myeloma patients.
Collapse
Affiliation(s)
- Mohamad A Hussein
- Cleveland Clinic Multidisciplinary Multiple Myeloma Research Program, Cleveland, OH 44195, USA.
| |
Collapse
|
16
|
Abstract
Multiple myeloma is a plasma cell malignancy that remains incurable with current treatment approaches including high-dose therapy and autologous stem cell transplantation. Thalidomide represents a major advance in the treatment of this disorder, having demonstrated significant activity in all phases of the disease. Thalidomide exerts its antimyeloma effect through multiple mechanisms including antiangiogenesis, immunomodulation and induction of apoptosis in tumor cells, as well as its effect on the tumor microenvironment. Corticosteroids have formed the mainstay of myeloma therapy for decades along with the alkylating agents and have demonstrated synergy when used in combination with thalidomide. The combination of thalidomide and dexamethasone has demonstrated remarkable activity in the treatment of both newly diagnosed as well as relapsed myeloma, and has become an important addition to the armamentarium of myeloma therapies. Overall responses of approximately 70% have been seen with this combination in patients with newly diagnosed myeloma. The combination is associated with an increased risk of deep vein thrombosis necessitating routine prophylactic anticoagulation. Other drugs have been added to this combination that also result in improved response rates. Currently, this combination is used in newly diagnosed patients as an induction therapy prior to stem cell transplant, for those who fail to achieve adequate response to dexamethasone alone or in whom a relatively rapid response is desired based on clinical presentation. Thalidomide analogs with a better safety profile are currently undergoing evaluation in the clinic.
Collapse
Affiliation(s)
- Shaji Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic and Foundation, Rochester, MN 55905, USA.
| | | |
Collapse
|
17
|
Activity of 129 single-agent drugs in 228 phase I and II clinical trials in multiple myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2013; 14:284-290.e5. [PMID: 24565465 DOI: 10.1016/j.clml.2013.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/17/2013] [Accepted: 12/23/2013] [Indexed: 11/22/2022]
Abstract
BACKGROUND More than 400 preclinical studies report ≥ 1 compound as cytotoxic to multiple myeloma (MM) cells; however, few of these agents became relevant in the clinic. Thus, the utility of such assays in predicting future clinical value is debatable. PATIENTS AND METHODS We examined the application of early-phase trial experiences to predict future clinical adoption. We identified 129 drugs explored as single agents in 228 trials involving 7421 patients between 1961 and 2013. RESULTS All drugs in common use in MM (melphalan, dexamethasone, prednisone, cyclophosphamide, bendamustine, thalidomide, lenalidomide, pomalidomide, bortezomib, carfilzomib, and doxorubicin) demonstrated a best reported response rate of ≥ 22%. Older agents, including teniposide, fotemustine, paclitaxel, and interferon, also appear active by this criterion; however, if mean response rates from all reported trials for an agent are considered, then only drugs with a mean response rate of 15% partial response are in clinical use. CONCLUSION Our analysis suggests that thresholds of 20% for best or 15% for mean response are highly predictive of future clinical success. Below these thresholds, no drug has yet reached regulatory approval or widespread use in the clinic. Thus, this benchmark provides 1 element of the framework for guiding choice of drugs for late-stage clinical testing.
Collapse
|
18
|
|
19
|
Jakubowiak A. Management strategies for relapsed/refractory multiple myeloma: current clinical perspectives. Semin Hematol 2012; 49 Suppl 1:S16-32. [PMID: 22727389 DOI: 10.1053/j.seminhematol.2012.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In the last decade, the introduction of novel agents including the immunomodulatory drugs thalidomide and lenalidomide, and the first-in-class proteasome inhibitor bortezomib, has dramatically improved clinical outcome in patients with relapsed/refractory multiple myeloma (MM) compared to conventional chemotherapy alone. Although combination treatment approaches with traditional cytotoxic agents and novel agents have led to response rates as high as 85% in patients with relapsed/refractory disease, not all patients will respond to established novel agents, and even those who do respond will ultimately relapse or become refractory to currently available regimens. There is no generally accepted standard treatment for patients with relapsed/refractory disease; however, both disease-related (eg, quality and duration of response to previous therapies and the aggressiveness of the relapse) and patient-related (eg, preexisting toxicities, comorbid conditions, quality of life, age, and performance status) factors should be considered when selecting the best treatment option. This article will review up-to-date approaches for managing patients with relapsed/refractory MM, including the efficacy and safety of established novel agents, the use of adjunctive/supportive care, and strategies for tailored treatment.
Collapse
|
20
|
Heuck CJ, Szymonifka J, Hansen E, Shaughnessy JD, Usmani SZ, van Rhee F, Anaissie E, Nair B, Waheed S, Alsayed Y, Petty N, Bailey C, Epstein J, Hoering A, Crowley J, Barlogie B. Thalidomide in total therapy 2 overcomes inferior prognosis of myeloma with low expression of the glucocorticoid receptor gene NR3C1. Clin Cancer Res 2012; 18:5499-506. [PMID: 22855579 DOI: 10.1158/1078-0432.ccr-12-0019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Because dexamethasone remains a key component of myeloma therapy, we wished to examine the impact of baseline and relapse expression levels of the glucocorticoid receptor gene NR3C1 on survival outcomes in the context of treatment with or without thalidomide. EXPERIMENTAL DESIGN We investigated the clinical impact of gene expression profiling (GEP)-derived expression levels of NR3C1 in 351 patients with GEP data available at baseline and in 130 with data available at relapse, among 668 subjects accrued to total therapy 2 (TT2). RESULTS Low NR3C1 expression levels had a negative impact on progression-free survival (PFS; HR, 1.47; P = 0.030) and overall survival (OS; HR, 1.90; P = 0.002) in the no-thalidomide arm. Conversely, there was a significant clinical benefit of thalidomide for patients with low receptor levels (OS: HR, 0.54; P = 0.015; PFS: HR, 0.54; P = 0.004), mediated most likely by thalidomide's upregulation of NR3C1. In the context of both baseline and relapse parameters, post-relapse survival (PRS) was adversely affected by low NR3C1 levels at relapse in a multivariate analysis (HR, 2.61; P = 0.012). CONCLUSION These findings justify the inclusion of NR3C1 expression data in the work-up of patients with myeloma as it can significantly influence the choice of therapy and, ultimately, OS. The identification of an interaction term between thalidomide and NR3C1 underscores the importance of pharmacogenomic studies in the systematic study of new drugs.
Collapse
|
21
|
Chesi M, Matthews GM, Garbitt VM, Palmer SE, Shortt J, Lefebure M, Stewart AK, Johnstone RW, Bergsagel PL. Drug response in a genetically engineered mouse model of multiple myeloma is predictive of clinical efficacy. Blood 2012; 120:376-85. [PMID: 22451422 PMCID: PMC3398763 DOI: 10.1182/blood-2012-02-412783] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 03/19/2012] [Indexed: 12/25/2022] Open
Abstract
The attrition rate for anticancer drugs entering clinical trials is unacceptably high. For multiple myeloma (MM), we postulate that this is because of preclinical models that overemphasize the antiproliferative activity of drugs, and clinical trials performed in refractory end-stage patients. We validate the Vk*MYC transgenic mouse as a faithful model to predict single-agent drug activity in MM with a positive predictive value of 67% (4 of 6) for clinical activity, and a negative predictive value of 86% (6 of 7) for clinical inactivity. We identify 4 novel agents that should be prioritized for evaluation in clinical trials. Transplantation of Vk*MYC tumor cells into congenic mice selected for a more aggressive disease that models end-stage drug-resistant MM and responds only to combinations of drugs with single-agent activity in untreated Vk*MYC MM. We predict that combinations of standard agents, histone deacetylase inhibitors, bromodomain inhibitors, and hypoxia-activated prodrugs will demonstrate efficacy in the treatment of relapsed MM.
Collapse
Affiliation(s)
- Marta Chesi
- Comprehensive Cancer Center, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kropff M, Baylon HG, Hillengass J, Robak T, Hajek R, Liebisch P, Goranov S, Hulin C, Bladé J, Caravita T, Avet-Loiseau H, Moehler TM, Pattou C, Lucy L, Kueenburg E, Glasmacher A, Zerbib R, Facon T. Thalidomide versus dexamethasone for the treatment of relapsed and/or refractory multiple myeloma: results from OPTIMUM, a randomized trial. Haematologica 2011; 97:784-91. [PMID: 22133776 DOI: 10.3324/haematol.2011.044271] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Thalidomide has potent antimyeloma activity, but no prospective, randomized controlled trial has evaluated thalidomide monotherapy in patients with relapsed/refractory multiple myeloma. DESIGN AND METHODS We conducted an international, randomized, open-label, four-arm, phase III trial to compare three different doses of thalidomide (100, 200, or 400 mg/day) with standard dexamethasone in patients who had received one to three prior therapies. The primary end-point was time to progression. RESULTS In the intent-to-treat population (N=499), the median time to progression was 6.1, 7.0, 7.6, and 9.1 months in patients treated with dexamethasone, and thalidomide 100, 200, and 400 mg/day, respectively; the difference between treatment groups was not statistically significant. In the per-protocol population (n=465), the median time to progression was 6.0, 7.0, 8.0, and 9.1 months, respectively. In patients who had received two or three prior therapies, thalidomide significantly prolonged the time to progression at all dose levels compared to the result achieved with dexamethasone. Response rates and median survival were similar in all treatment groups, but the median duration of response was significantly longer in all thalidomide groups than in the dexamethasone group. Adverse events reported in the thalidomide groups, such as fatigue, constipation and neuropathy, confirmed the known safety profile of thalidomide. CONCLUSIONS Although thalidomide was not superior to dexamethasone in this randomized trial, thalidomide monotherapy may be considered an effective salvage therapy option for patients with relapsed/refractory multiple myeloma, particularly those with a good prognosis and those who have received two or three prior therapies. The recommended starting dose of thalidomide monotherapy is 400 mg/day, which can be rapidly reduced for patients who do not tolerate this treatment. ( CLINICAL TRIAL REGISTRATION NUMBER NCT00452569).
Collapse
Affiliation(s)
- Martin Kropff
- Department of Medicine, University of Muenster, Muenster D-48129, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lonial S, Mitsiades CS, Richardson PG. Treatment options for relapsed and refractory multiple myeloma. Clin Cancer Res 2011; 17:1264-77. [PMID: 21411442 DOI: 10.1158/1078-0432.ccr-10-1805] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment options for patients with relapsed myeloma have benefited from the development of new targeted agents. The use of bortezomib, thalidomide, and lenalidomide have dramatically changed outcomes for patients with relapsed myeloma. New agents are also in development, on the basis of preclinical rationale, as well as combinations of conventional and novel agents. Together each of these treatment approaches are being tested in phase I, II, and III clinical trials, with the goal of prolonged duration of remission and, ultimately, improved overall survival.
Collapse
Affiliation(s)
- Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA.
| | | | | |
Collapse
|
24
|
Morgenroth A, Dinger C, Zlatopolskiy BD, Al-Momani E, Glatting G, Mottaghy FM, Reske SN. Auger electron emitter against multiple myeloma--targeted endo-radio-therapy with 125I-labeled thymidine analogue 5-iodo-4'-thio-2'-deoxyuridine. Nucl Med Biol 2011; 38:1067-77. [PMID: 21982576 DOI: 10.1016/j.nucmedbio.2011.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 02/18/2011] [Accepted: 02/27/2011] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is a plasma cell malignancy characterized by accumulation of malignant, terminally differentiated B cells in the bone marrow. Despite advances in therapy, MM remains an incurable disease. Novel therapeutic approaches are, therefore, urgently needed. Auger electron-emitting radiopharmaceuticals are attractive for targeted nano-irradiation therapy, given that DNA of malignant cells is selectively addressed. Here we evaluated the antimyeloma potential of the Auger electron-emitting thymidine analogue (125)I-labeled 5-iodo-4'-thio-2'-deoxyuridine ([(125)I]ITdU). METHODS Cellular uptake and DNA incorporation of [(125)I]ITdU were determined in fluorodeoxyuridine-pretreated KMS12BM, U266, dexamethasone-sensitive MM1.S and -resistant MM1.R cell lines. The effect of stimulation with interleukin 6 (IL6) or insulin-like growth factor 1 (IGF1) on the intracellular incorporation of [(125)I]ITdU was investigated in cytokine-sensitive MM1.S and MM1.R cell lines. Apoptotic cells were identified using Annexin V. Cleavage of caspase 3 and PARP was visualized by Western blot. DNA fragmentation was investigated using laddering assay. Therapeutic efficiency of [(125)I]ITdU was proven by clonogenic assay. RESULTS [(125)I]ITdU was shown to be efficiently incorporated into DNA of malignant cells, providing a promising mechanism for delivering highly toxic Auger radiation emitters into tumor DNA. [(125)I]ITdU had a potent antimyeloma effect in cell lines representing distinct disease stages and, importantly, in cell lines sensitive or resistant to the conventional therapeutic agent, but was not toxic for normal plasma and bone marrow stromal cells. Furthermore, [(125)I]ITdU abrogated the protective actions of IL6 and IGF1 on MM cells. [(125)I]ITdU induced massive damage in the DNA of malignant plasma cells, which resulted in efficient inhibition of clonogenic growth. CONCLUSION These studies may provide a novel treatment strategy for overcoming resistance to conventional therapy in multiple myeloma.
Collapse
Affiliation(s)
- Agnieszka Morgenroth
- Nuclear Medicine Clinic, University Ulm, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | | | | | | | | | | | | |
Collapse
|
25
|
Messori A, Maratea D, Nozzoli C, Bosi A. The role of bortezomib, thalidomide and lenalidomide in the management of multiple myeloma: an overview of clinical and economic information. PHARMACOECONOMICS 2011; 29:269-285. [PMID: 21395348 DOI: 10.2165/11585930-000000000-00000] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Bortezomib, thalidomide and lenalidomide can be aimed at treating patients with newly diagnosed multiple myeloma (both eligible and ineligible for transplantation) as well as those with relapsed or refractory disease. This review analysed the available clinical and economic data on these three drugs. Irrespective of which of the three agents is considered, the magnitude of the benefit in newly diagnosed cases (transplanted or non-transplanted) tends to be between 10 and 20 months per patient in terms of progression-free survival or survival; the survival benefit is smaller in relapsed or refractory disease. In addition, a single-institution observational analysis evaluated the outcomes in nearly 3000 consecutive patients examined between 1971 and 2006. The survival in patients diagnosed between 2001 and 2006 was longer than that observed in patients diagnosed between 1994 and 2000. This finding supports the conclusion that novel agents provide a survival improvement compared with traditional therapy. Formal cost-effectiveness studies on these three agents are still lacking. A MEDLINE search retrieved only four short papers or letters and no full-length analysis. Hence, the cost effectiveness of these agents needs further investigation, with separate assessments of the different therapeutic settings. In a simplified analysis, we tried to contrast the average cost of treatment for each of the novel agents versus their respective benefit, expressed in quality-adjusted survival. Despite its preliminary nature, our assessment indicates that the cost effectiveness of these three agents is likely to be within commonly accepted pharmacoeconomic thresholds.
Collapse
Affiliation(s)
- Andrea Messori
- Laboratory of Pharmacoeconomics, co Area Vasta Centro, Regional Health System, Florence, Italy.
| | | | | | | |
Collapse
|
26
|
Darda Bayraktar U, Warsch, S, Pereira D. High-dose glucocorticoids improve renal failure reversibility in patients with newly diagnosed multiple myeloma. Am J Hematol 2011; 86:224-7. [PMID: 21264915 DOI: 10.1002/ajh.21922] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
One-fifth of the newly diagnosed multiple myeloma (MM) patients present with renal failure (RF) [1-3]. Glucocorticoids (GCs) may improve RF in MM by (1) rapid reduction of paraprotein production, (2) lessening inflammation and fibrosis in renal parenchyma, and (3) decreasing serum calcium level. We hypothesized that lower dose GCs may be less effective in restoring renal function and retrospectively compared the RF reversibility between the newly diagnosed MM patients who were treated with GCs equivalent to ≥160 mg DX over 4 days (high-dose GC group, n = 16) versus those who were treated with <160 mg (low-dose/no GC group, n = 8). There was no difference in age, baseline calcium, and creatinine levels between the two groups. Renal function was restored in seven patients in the high-dose GC group (44%) and in none of the patients in the low-dose/no GC group (P = 0.026). The only other factor found to impact the RF reversibility was the delay of GC initiation. Four and 1 patients developed a severe infection in the high- and low-dose/no GC groups, respectively. The use of higher dose GCs in the newly diagnosed MM patients who present with RF increases the likelihood of renal function restoration.
Collapse
Affiliation(s)
- Ulas Darda Bayraktar
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Sean Warsch,
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Denise Pereira
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
27
|
Abstract
Despite considerable improvements in first line treatment still the majority of patients experience relapse of multiple myeloma. Treatment decisions for relapse or refractory multiple myeloma should be based on a clinical decision path taking response and adverse events to previous therapy, myeloma specific complications and organ dysfunctions, overall clinical condition, age, cytogenetic information and prognostic factors into account. Bortezomib, thalidomide and lenalidomide have improved the therapeutic armentarium for patients with refractory or relapsed disease and are often used in combination with dexamethasone or chemotherapeutic agents. Combination therapies of novel agents in drug combination regimen are currently under investigation as well. For patients with a disease free survival of 12 month or longer after initial single or tandem high dose therapy and autologous stem cell transplantation (ASCT) repeat of high dose therapy with melphalan and ASCT should be considered in case of relapse. Radiotherapy and osteoplastic procedures can be used as adjunct to systemic therapy to treat local complications in particular vertebral pain caused by osteolytic bone disease. Cytogenetic tests, molecular techniques as gene expression profiling and other diagnostic will lead to a more individualized therapy. The integration of novel compounds into established regimen will be a major challenge for future clinical studies.
Collapse
Affiliation(s)
- Thomas Moehler
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
28
|
Nazha A, Cook R, Vogl DT, Mangan PA, Gardler M, Hummel K, Cunningham K, Luger SM, Porter DL, Schuster S, O'Doherty U, Siegel D, Stadtmauer EA. Stem cell collection in patients with multiple myeloma: impact of induction therapy and mobilization regimen. Bone Marrow Transplant 2010; 46:59-63. [DOI: 10.1038/bmt.2010.63] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
29
|
Palumbo A, Larocca A, Falco P, Sanpaolo G, Falcone AP, Federico V, Canepa L, Crugnola M, Genuardi M, Magarotto V, Petrucci MT, Boccadoro M. Lenalidomide, melphalan, prednisone and thalidomide (RMPT) for relapsed/refractory multiple myeloma. Leukemia 2010; 24:1037-42. [PMID: 20376079 DOI: 10.1038/leu.2010.58] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This multicenter, open-label, non-comparative phase II trial evaluated the safety and efficacy of salvage therapy with lenalidomide, melphalan, prednisone and thalidomide (RMPT) in patients with relapsed/refractory multiple myeloma (MM). Oral lenalidomide (10 mg/day) was administered on days 1-21, and oral melphalan (0.18 mg/kg) and oral prednisone (2 mg/kg) on days 1-4 of each 28-day cycle. Thalidomide was administered at 50 mg/day or 100 mg/day on days 1-28; six cycles were administered in total. Maintenance included lenalidomide 10 mg/day on days 1-21, until unacceptable adverse events or disease progression. Aspirin (100 mg/day) was given as thromboprophylaxis. A total of 44 patients with relapsed/refractory MM were enrolled and 75% achieved at least a partial response (PR), including 32% very good PR (VGPR) and 2% complete response (CR). The 1-year progression-free survival (PFS) was 51% and the 1-year overall survival (OS) from study entry was 72%. Grade 4 hematologic adverse events included neutropenia (18%), thrombocytopenia (7%) and anemia (2%). Grade 3 non-hematologic adverse events were infections (14%), neurological toxicity (4.5%) and fatigue (7%). No grade 3/4 thromboembolic events or peripheral neuropathy were reported. In conclusion, RMPT is an active salvage therapy with good efficacy and manageable side effects. This study represents the basis for larger phase III randomized trials.
Collapse
Affiliation(s)
- A Palumbo
- Divisione di Ematologia dell'Università di Torino, AOU S. Giovanni Battista, Turin, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sinha S, Rajkumar SV, Lacy MQ, Hayman SR, Buadi FK, Dispenzieri A, Dingli D, Kyle RA, Gertz MA, Kumar S. Impact of dexamethasone responsiveness on long term outcome in patients with newly diagnosed multiple myeloma. Br J Haematol 2009; 148:853-8. [PMID: 19958361 DOI: 10.1111/j.1365-2141.2009.08023.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dexamethasone (Dex), alone or in combination, is commonly used for treating multiple myeloma. Dex as single agent for initial therapy of myeloma results in overall response rates of 50-60%. It is unclear whether steroid responsiveness reflects any biological characteristic that impacts long-term outcome. We studied a cohort of 182 patients with newly diagnosed myeloma seen between March 1998 and June 2007, initially treated with single-agent Dex for at least 4 weeks. The median age at diagnosis was 63 years (range, 39-81) and the median estimated survival was 55 months. At a median duration of therapy of 15 weeks, 91 (50%) patients had a partial response or better, 80 (44%) had less than partial response and the remaining (6%) patients were not evaluable. The median overall survival from diagnosis for the responders was 75 months compared to 71 months for remaining patients, P = 0.6.There was no correlation between baseline disease characteristics and Dex responsiveness. While overall survival was longer for the 130 (70%) patients who proceeded to an autologous stem cell transplant, no correlation was found between survival and Dex responsiveness among either group. Among this cohort of patients with myeloma, failure to respond to single agent steroid did not have an adverse impact on eventual outcome.
Collapse
Affiliation(s)
- Shirshendu Sinha
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Laubach JP, Mahindra A, Mitsiades CS, Schlossman RL, Munshi NC, Ghobrial IM, Carreau N, Hideshima T, Anderson KC, Richardson PG. The use of novel agents in the treatment of relapsed and refractory multiple myeloma. Leukemia 2009; 23:2222-32. [PMID: 19741729 DOI: 10.1038/leu.2009.179] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Although outcomes for patients with multiple myeloma (MM) have improved over the past decade, the disease remains incurable and even patients who respond well to induction therapy ultimately relapse and require additional treatment. Conventional chemotherapy and high-dose therapy with stem cell transplantation (SCT) have historically been utilized in the management of relapsed MM, but in recent years the immunomodulatory drugs (IMiDs) thalidomide and lenalidomide, as well as the proteasome inhibitor bortezomib, have assumed a primary role in this setting. This review focuses on the role of thalidomide, lenalidomide and bortezomib in relapsed and refractory MM, with additional discussion dedicated to emerging drugs in relapsed MM that may prove beneficial to patients with this disease.
Collapse
Affiliation(s)
- J P Laubach
- Department of Medical Oncology, Harvard Medical School, Dana Farber Cancer Institute, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Laubach JP, Mitsiades CS, Hideshima T, Schlossman R, Chauhan D, Munshi N, Ghobrial I, Carreau N, Anderson KC, Richardson PG. Bortezomib in the management of multiple myeloma. Cancer Manag Res 2009; 1:107-17. [PMID: 21188129 PMCID: PMC3004671 DOI: 10.2147/cmar.s4555] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by clonal expansion of plasma cells within the bone marrow, the presence of a serum and/or urine monoclonal protein, lytic bone lesions, and anemia. On a cellular level, the disease is characterized by complex interactions between tumor cells and the surrounding bone marrow microenvironment. Understanding of the relationship between malignant plasma cells and the microenvironment has sparked ongoing efforts to develop targeted therapeutic agents for treatment of this disease. The successful development of the first-in-class small-molecule proteasome inhibitor bortezomib occurred as a result of these efforts. This review focuses on the rationale for bortezomib therapy in the treatment of patients with newly diagnosed and relapsed MM, important treatment-related side effects, and future directions for use of bortezomib and other, emerging proteasome inhibitors.
Collapse
Affiliation(s)
- Jacob P Laubach
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Multiple myeloma is characterised by clonal proliferation of malignant plasma cells, and mounting evidence indicates that the bone marrow microenvironment of tumour cells has a pivotal role in myeloma pathogenesis. This knowledge has already expanded treatment options for patients with multiple myeloma. Prototypic drugs thalidomide, bortezomib, and lenalidomide have each been approved for the treatment of this disease by targeting both multiple myeloma cells and the bone marrow microenvironment. Although benefit was first shown in relapsed and refractory disease, improved overall response, duration of response, and progression-free and overall survival can be achieved when these drugs are part of first-line regimens. This treatment framework promises to improve outcome not only for patients with multiple myeloma, but also with other haematological malignancies and solid tumours.
Collapse
Affiliation(s)
- Marc S Raab
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
The introduction of several novel and active treatments and improvements in supportive care of myeloma patients has resulted in a prolongation of the survival of these patients. However, myeloma remains an incurable disease and almost all patients will relapse. Effective management of the relapsing/refractory disease incorporates several different strategies, depending on prior treatments, responses, and duration of responses, as well as residual toxicity, age, and physical condition. High-dose dexamethasone still has a role in the management of disease complications such as cytopenias, renal impairment, or spinal cord compression until another agent is added. High-dose therapy may be considered for selected patients who have a long-term treatment-free interval after their first transplantation. Allogeneic transplantation is limited to selected young patients, preferably with an HLA-matched donor. However, the backbone of current strategies for the management of relapsed/refractory myeloma includes the novel agents thalidomide, bortezomib, and lenalidomide. These agents, either with dexamethasone or in combination with chemotherapy, have shown significant activity both in relapsed and in refractory patients. Based on the results of phase III trials, lenalidomide and bortezomib have increased the post-relapse survival and are active in patients who have received prior novel agents; lenalidomide is active in thalidomide-pretreated or bortezomib-pretreated patients and bortezomib alone or in combination with chemotherapy is active in thalidomide/lenalidomide-pretreated patients. Combinations of novel agents show synergistic activity and may overcome drug resistance. Finally, special consideration is needed for the management of patients with renal impairment or other poor prognostic features.
Collapse
Affiliation(s)
- Efstathios Kastritis
- Department of Clinical Therapeutics, University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | | | | |
Collapse
|
35
|
Reece DE, Leitch HA, Atkins H, Voralia M, Canning LA, LeBlanc R, Belch AR, White D, Kovacs MJ. Treatment of relapsed and refractory myeloma. Leuk Lymphoma 2009; 49:1470-85. [DOI: 10.1080/10428190802064941] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Podar K, Tai YT, Hideshima T, Vallet S, Richardson PG, Anderson KC. Emerging therapies for multiple myeloma. Expert Opin Emerg Drugs 2009; 14:99-127. [PMID: 19249983 DOI: 10.1517/14728210802676278] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma cell malignancy clinically characterized by osteolytic lesions, immunodeficiency, and renal disease. There are an estimated 750,000 people diagnosed with MM worldwide, with a median overall survival of 3 - 5 years. Besides chromosomal aberrations, translocations, and mutations in essential growth and tumor-suppressor genes, accumulating data strongly highlight the pathophysiologic role of the bone marrow (BM) microenvironment in MM pathogenesis. Based on this knowledge, several novel agents have been identified, and treatment options in MM have fundamentally changed during the last decade. Thalidomide, bortezomib, and lenalidomide have been incorporated into conventional cytotoxic and transplantation regimens, first in relapsed and refractory and now also in newly diagnosed MM. Despite these significant advances, there remains an urgent need for more efficacious and tolerable drugs. Indeed, a plethora of preclinical agents awaits translation from the bench to the bedside. This article reviews the scientific rationale of new therapy regimens and newly identified therapeutic agents - small molecules as well as therapeutic antibodies - that hold promise to further improve outcome in MM.
Collapse
Affiliation(s)
- Klaus Podar
- Dana-Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Boston, MA 02115, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Barlogie B, Alexanian R. Prognostic factors in multiple myeloma: the M.D. Anderson experience. Eur J Haematol Suppl 2009; 51:84-7. [PMID: 2627993 DOI: 10.1111/j.1600-0609.1989.tb01498.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Palva IP, Ahrenberg P, Harja KA, Almqvist A, Elonen E, Hallman H, Hänninen A, Ilvonen M, Isomaa B, Jouppila J, Järvenpää E, Järventie G, Kilpi H, Koivunen E, Kätkä K, Kääriäinen M, Lahtinen R, Laitinen A, Lehtinen M, Mäkelä H, Nyländen P, Nyman D, Oivanen T, Pakkala S, Pelliniemi TT, Pulli T, Rajamäki A, Ruutu T, Savola J, Soininen K, Timonen T, Wasastjerna C, Vilpo J. Intensive chemotherapy with combinations containing anthracyclines for refractory and relapsing multiple myeloma. Eur J Haematol 2009. [DOI: 10.1111/j.1600-0609.1990.tb00362.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
39
|
Abstract
This report summarizes a broad experience in the treatment of patients with multiple myeloma resistant to standard chemotherapy. The VAD regimen has induced remissions in about 50% of relapsing patients but in only about 25% of previously unresponsive patients. In patients resistant to VAD, high-dose therapies with intravenous melphalan, a CBV combination (cyclophosphamide-BCNU-VP-16) or an EDAP regimen (VP-16 -platinum) produced responses in about 40% of patients. However, these treatments usually required autologous bone marrow or blood stem cell support and the median duration of control was only 6 months. With an even more intensive program using high-dose melphalan and total body irradiation supported by autologous bone marrow, all patients who survived the early treatment period responded for a median duration of about 1 year. Results indicated a dose-response effect of chemoradiotherapy on VAD-resistant myeloma with the potential that such intensive regimens will prolong disease-free survival time.
Collapse
|
40
|
Gertz MA. New targets and treatments in multiple myeloma: Src family kinases as central regulators of disease progression. Leuk Lymphoma 2009; 49:2240-5. [PMID: 19052970 DOI: 10.1080/10428190802475311] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple myeloma is a malignant condition that most commonly occurs in the seventh decade of life. Recent improvements in treatment may result in a more favourable outlook for recently diagnosed patients. Multiple myeloma is an incurable clonal B-cell malignancy, which is initially responsive to conventional chemotherapy; one-third of the patients achieve complete remission but multidrug resistance eventually develops. Although autologous stem cell transplantation remains an important option, many older patients are less tolerant to the toxicity associated with conditioning treatment, as well as being intrinsically less likely to do well after transplantation. Most patients eventually relapse with or without transplantation, and salvage therapy is only moderately effective. Thalidomide and subsequently, lenalidomide and bortezomib, have demonstrated improved outcomes for these patients, as well as proving efficacious in front-line regimens. A deeper understanding of the molecular mechanisms underlying multiple myeloma has given rise to novel targeted approaches. This review will focus in particular on Src-dependent signalling pathways, reflecting the expanding realisation of the critical and ubiquitous role of Src family kinases (SFKs) in normal and abnormal hematopoiesis.
Collapse
Affiliation(s)
- Morie A Gertz
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
41
|
|
42
|
Hazarika M, Rock E, Williams G, Dagher R, Sridhara R, Booth B, Farrell A, Justice R, Pazdur R. Lenalidomide in combination with dexamethasone for the treatment of multiple myeloma after one prior therapy. Oncologist 2008; 13:1120-7. [PMID: 18922829 DOI: 10.1634/theoncologist.2008-0077] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Lenalidomide (CC-5013, Revlimid; Celgene Corporation, Summit, NJ), a thalidomide analogue, was granted approval by the U.S. Food and Drug Administration (FDA) on June 29, 2006, for use in combination with dexamethasone in patients with multiple myeloma (MM) who have received at least one prior therapy. The FDA approved lenalidomide with a restricted distribution program, RevAssist. EXPERIMENTAL DESIGN In two randomized, double-blind, multicenter studies, the combination of lenalidomide and dexamethasone (LD) was compared with placebo and dexamethasone (PD) in patients with MM who had received at least one prior therapy. The primary endpoint was time to progression (TTP). RESULTS Following a prespecified interim analysis of TTP, an independent data-monitoring committee advised the sponsor to halt the two studies. For both studies, the interim analysis for efficacy revealed a statistically significant longer TTP with LD than with PD. The most clinically relevant grade 3 and 4 adverse events that occurred more frequently in the LD arm were neutropenia, thrombocytopenia, deep vein thrombosis, pulmonary embolism, and atrial fibrillation. Thrombotic or thromboembolic events, including deep vein thrombosis, pulmonary embolism, thrombosis, and intracranial venous sinus thrombosis were reported more frequently in patients treated with LD than with PD. CONCLUSIONS The FDA approved lenalidomide based on interim results from two multicenter, placebo-controlled, randomized trials comparing the combination of LD with PD that revealed a longer TTP with LD than with PD. The major toxicity observed during these trials was myelosuppression. The serious toxicities included thromboembolic events. Lenalidomide is only available under the RevAssist Program.
Collapse
Affiliation(s)
- Maitreyee Hazarika
- Office of Oncology Drug Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland 20993-0002, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Redzepovic J, Weinmann G, Ott I, Gust R. Current Trends in Multiple Myeloma Management. J Int Med Res 2008; 36:371-86. [DOI: 10.1177/147323000803600301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Treatment of multiple myeloma, a B-cell cancer, is usually palliative, however, as a result of intensive clinical research there are numerous new treatment options available today. The present review summarizes non-transplant treatment options for multiple myeloma on the basis of available publications. Treatment with new substances, such as immunomodulatory agents, farnesyl transferase inhibitors and apoptosis stimulators, and their mechanisms of action are discussed. In addition to this systematic review of the available evidence on multiple myeloma therapy we have also summarized current recommendations from national and international organizations on aspects of the treatment of multiple myeloma. This should enable readers to see different points of view at a glance and, hopefully, will provide a basis for translation of the available evidence into the best possible therapy.
Collapse
Affiliation(s)
- J Redzepovic
- Institute of Pharmacy, Free University of Berlin, Berlin, Germany
| | | | - I Ott
- Institute of Pharmacy, Free University of Berlin, Berlin, Germany
| | - R Gust
- Institute of Pharmacy, Free University of Berlin, Berlin, Germany
| |
Collapse
|
44
|
AID-dependent activation of a MYC transgene induces multiple myeloma in a conditional mouse model of post-germinal center malignancies. Cancer Cell 2008; 13:167-80. [PMID: 18242516 DOI: 10.1016/j.ccr.2008.01.007] [Citation(s) in RCA: 293] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 11/27/2007] [Accepted: 01/08/2008] [Indexed: 11/29/2022]
Abstract
By misdirecting the activity of Activation-Induced Deaminase (AID) to a conditional MYC transgene, we have achieved sporadic, AID-dependent MYC activation in germinal center B cells of Vk*MYC mice. Whereas control C57BL/6 mice develop benign monoclonal gammopathy with age, all Vk*MYC mice progress to an indolent multiple myeloma associated with the biological and clinical features highly characteristic of the human disease. Furthermore, antigen-dependent myeloma could be induced by immunization with a T-dependent antigen. Consistent with these findings in mice, more frequent MYC rearrangements, elevated levels of MYC mRNA, and MYC target genes distinguish human patients with multiple myeloma from individuals with monoclonal gammopathy, implicating a causal role for MYC in the progression of monoclonal gammopathy to multiple myeloma.
Collapse
|
45
|
Abstract
Studies of bortezomib, thalidomide, and lenalidomide have shown promising clinical activity in relapsed/refractory multiple myeloma (MM). Bortezomib alone and in combination with other agents is associated with high response rates, consistently high rates of complete response, and a predictable and manageable profile of adverse events. Thalidomide-based regimens have also shown substantial clinical activity. The accumulating experience from ongoing trials of bortezomib/lenalidomide/dexamethasone combinations in patients who have relapsed/refractory or newly diagnosed MM will provide critical information that will determine the possible role of this combination as the basic backbone for combination regimens for management of advanced MM.
Collapse
|
46
|
Ogawa Y, Tobinai K, Ogura M, Ando K, Tsuchiya T, Kobayashi Y, Watanabe T, Maruyama D, Morishima Y, Kagami Y, Taji H, Minami H, Itoh K, Nakata M, Hotta T. Phase I and II pharmacokinetic and pharmacodynamic study of the proteasome inhibitor bortezomib in Japanese patients with relapsed or refractory multiple myeloma. Cancer Sci 2008; 99:140-4. [PMID: 17970782 PMCID: PMC11159153 DOI: 10.1111/j.1349-7006.2007.00638.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2007] [Revised: 09/06/2007] [Accepted: 09/09/2007] [Indexed: 11/30/2022] Open
Abstract
The purpose of this phase I and II study was to evaluate the safety, pharmacokinetics, pharmacodynamics, and efficacy of bortezomib in Japanese patients with relapsed or refractory multiple myeloma. This was a dose-escalation study designed to determine the recommended dose for Japanese patients (phase I) and to investigate the antitumor activity and safety (phase II) of bortezomib administered on days 1, 4, 8, and 11 every 21 days. Thirty-four patients were enrolled. A dose-limiting toxicity was febrile neutropenia, which occurred in one of six patients in the highest-dose cohort in phase I and led to the selection of 1.3 mg/m(2) as the recommended dose. Adverse events >or= grade 3 were rare except for hematological toxicities, although there was one fatal case of interstitial lung disease. The overall response rate was 30% (95% confidence interval, 16-49%). Pharmacokinetic evaluation showed a biexponential decline, characterized by a rapid distribution followed by a longer elimination, after dose administration, whereas the area under the concentration-time curve increased proportionately with the dose. Bortezomib was effective in Japanese patients with relapsed or refractory multiple myeloma. A favorable tolerability profile was also seen, although the potential for pulmonary toxicity should be monitored closely. The pharmacokinetic and pharmacodynamic profiles of bortezomib in the present study warrant further investigations, including more relevant administration schedules.
Collapse
Affiliation(s)
- Yoshiaki Ogawa
- Department of Hematology and Oncology, Tokai University School of Medicine, 143, Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The advent of new therapies for multiple myeloma brings new hope for patients but also new side effects. Emerging information about the risks of supportive care therapies, including long-term, high-intensity bisphosphonate use and erythropoiesis-stimulating agents, is examined. As the number of drugs in the myeloma armamentarium grows, so does the list of possible side effects and interactions. With current progress, not only are there more complications to consider but patients are also living longer and the risk for delayed complications is becoming more relevant. The author provides perspective about the risks for the most active and commonly used single-agent and combination myeloma therapies.
Collapse
Affiliation(s)
- Angela Dispenzieri
- Division of Hematology, Mayo Clinic, 200 First Street, Rochester, MN 55905, USA.
| |
Collapse
|
48
|
Pant S, Copelan EA. Hematopoietic Stem Cell Transplantation in Multiple Myeloma. Biol Blood Marrow Transplant 2007; 13:877-85. [PMID: 17640590 DOI: 10.1016/j.bbmt.2007.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Accepted: 05/01/2007] [Indexed: 10/23/2022]
Abstract
High-dose therapy and autologous hematopoietic stem cell transplantation are standard early treatment of patients with multiple myeloma. Tandem transplantation appears to provide additional benefit, particularly in patients with limited response to initial transplantation. Myeloablative allogeneic transplantation provides the only potential for cure, but has been largely abandoned because of high mortality rates. Newer and better induction regimens, rigorous analysis of results with autologous and allogeneic transplantation, and the development of risk-adapted stratification provide the impetus for this critical evaluation of the role of hematopoietic stem cell transplantation in multiple myeloma.
Collapse
Affiliation(s)
- Shubham Pant
- Division of Hematology and Oncology, Ohio State University, Columbus, Ohio 44195, USA
| | | |
Collapse
|
49
|
Cavallo F, Ambrosini MT, Rus C, Boccadoro M, Palumbo A. The treatment of the elderly multiple myeloma patients. Leuk Lymphoma 2007; 48:469-80. [PMID: 17454586 DOI: 10.1080/10428190601059852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Multiple myeloma (MM) is an incurable malignancy of the plasma cells. Most patients are diagnosed when they are older than 65 years. Therapeutic options include chemotherapy, using either established (e.g. melphalan) or newly available (e.g. thalidomide) drugs and high-dose treatment with stem-cell support (autologous as well as allogeneic). Recent research has focused on defining the target population for the different therapeutic approaches, taking into account pre-treatment characteristics of patients, particularly age, and aims to balance treatment benefit with potential adverse events. In this review we present the data available on the most recent trials dealing with the treatment of elderly MM patients.
Collapse
Affiliation(s)
- Federica Cavallo
- Divisione di Ematologia dell'Universita' di Torino, Azienda Ospedaliera San Giovanni Battista, Torino, Italy
| | | | | | | | | |
Collapse
|
50
|
Armand JP, Burnett AK, Drach J, Harousseau JL, Löwenberg B, San Miguel J. The emerging role of targeted therapy for hematologic malignancies: update on bortezomib and tipifarnib. Oncologist 2007; 12:281-90. [PMID: 17405892 DOI: 10.1634/theoncologist.12-3-281] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
As therapy for hematologic malignancy evolves, new regimens and novel agents that target specific cellular processes allow a more optimistic prognosis for many patients. Bortezomib and tipifarnib are two new, targeted treatments for hematologic malignancies. Bortezomib, a proteasome inhibitor, has shown impressive efficacy in patients with relapsed multiple myeloma and as initial treatment, including before autologous stem cell transplantation. It has been studied as monotherapy and in combination with standard treatments such as dexamethasone, and with newer agents such as the immunomodulators thalidomide and lenalidomide; response is encouraging, even in patients who have relapsed after previously receiving components of a regimen as single agents. Bortezomib is generally well tolerated, including in combination with novel and conventional agents. Tipifarnib is a specific inhibitor of farnesyltransferase. Clinical trials in patients with high-risk acute leukemias and myelodysplastic syndromes have demonstrated good efficacy with tipifarnib. Continued investigation with these new, targeted treatments will further define their use as treatment options in patients with hematologic cancer.
Collapse
Affiliation(s)
- Jean-Pierre Armand
- Institut Gustave-Roussy, Dept. of Medicine, 39 Rue Camille-Desmoulins, Villejuif Cedex 94805, France.
| | | | | | | | | | | |
Collapse
|