37 results on '"Blimark C"'
Search Results
2. Ixazomib-Thalidomide-low dose dexamethasone induction followed by maintenance therapy with ixazomib or placebo in newly diagnosed multiple myeloma patients not eligible for autologous stem cell transplantation; results from the randomized phase II HOVON-126/NMSG 21.13 trial
- Author
-
Zweegman, S., Stege, C.A.M., Haukas, E., Schjesvold, F.H., Levin, M.D., Waage, A., Leys, R.B.L., Klein, S.K., Szatkowski, D., Axelsson, P., Do, T.H., Knut-Bojanowska, D., Spek, E. van der, Svirskaite, A., Klostergaard, A., Salomo, M., Blimark, C., Ypma, P.F., Mellqvist, U.H., Poddighe, P.J., Stevens-Kroef, M.J., Donk, N.W. van de, Sonneveld, P., Hansson, M., Holt, B. van der, Abildgaard, N., Zweegman, S., Stege, C.A.M., Haukas, E., Schjesvold, F.H., Levin, M.D., Waage, A., Leys, R.B.L., Klein, S.K., Szatkowski, D., Axelsson, P., Do, T.H., Knut-Bojanowska, D., Spek, E. van der, Svirskaite, A., Klostergaard, A., Salomo, M., Blimark, C., Ypma, P.F., Mellqvist, U.H., Poddighe, P.J., Stevens-Kroef, M.J., Donk, N.W. van de, Sonneveld, P., Hansson, M., Holt, B. van der, and Abildgaard, N.
- Abstract
Contains fulltext : 229254.pdf (publisher's version ) (Open Access)
- Published
- 2020
3. Melphalan 100 mg/m2 with stem cell support as first-relapse treatment is safe and effective for myeloma patients with long remission after autologous stem cell transplantation: P893
- Author
-
Blimark, C., Veskovski, L., Brune, M., Westin, J., Holmberg, E., Hjorth, M., Rodjer, S., Andersson, P. O., and Mellqvist, U. H.
- Published
- 2011
4. Carfilzomib and dexamethasone maintenance prolong time to progression following salvage ASCT in multiple myeloma
- Author
-
Gregersen, Henrik, Peceliunas, Valdas, Remes, Kari, Schjesvold, Fredrik H., Abildgaard, Niels, Nahi, Hareth, Andersen, N. F., Vangsted, Annette Juul, Klausen, Tobias Wirenfeldt, Helleberg, Carsten, Carlson, K., Frølund, Ulf, Axelsson, Per, Stromberg, Olga, Blimark, C, Linder, Olle, Tsykunova, Galina, Waage, Anders, Hansson, Markus, and Guldbrandsen, N.
- Published
- 2019
5. Carfilzomib and dexamethasone maintenance prolong time to progression following salvage ASCT in multiple myeloma: A randomized phase 2 trial by the nordic myeloma study group
- Author
-
Gregersen, Henrik, Peceliunas, Valdas, Remes, Kari, Schjesvold, Fredrik H., Abildgaard, Niels, Nahi, Hareth, Andersen, N. F., Vangsted, Annette Juul, Klausen, Tobias Wirenfeldt, Helleberg, Carsten, Carlson, K., Frølund, Ulf, Axelsson, Per, Stromberg, Olga, Blimark, C, Linder, Olle, Tsykunova, Galina, Waage, Anders, Hansson, Markus, and Guldbrandsen, N.
- Published
- 2019
6. Oral ixazomib maintenance following autologous stem cell transplantation (TOURMALINE-MM3): a double-blind, randomised, placebo-controlled phase 3 trial
- Author
-
Dimopoulos, M.A. Gay, F. Schjesvold, F. Beksac, M. Hajek, R. Weisel, K.C. Goldschmidt, H. Maisnar, V. Moreau, P. Min, C.K. Pluta, A. Chng, W.-J. Kaiser, M. Zweegman, S. Mateos, M.-V. Spencer, A. Iida, S. Morgan, G. Suryanarayan, K. Teng, Z. Skacel, T. Palumbo, A. Dash, A.B. Gupta, N. Labotka, R. Rajkumar, S.V. Bar, D. Basso, A. Fantl, D. He, S. Horvath, N. Lee, C. Rowlings, P. Taylor, K. Cochrane, T. Kwok, F. Ramanathan, S. Agis, H. Zojer, N. Kentos, A. Offner, F. Van Droogenbroeck, J. Wu, K.L. Maiolino, A. Martinez, G. Zanella, K. Capra, M. Araújo, S. Gregora, E. Pour, L. Scudla, V. Spicka, I. Abildgaard, N. Andersen, N. Jensen, B.A. Helleberg, C. Plesner, T. Salomo, M. Svirskaite, A. Delarue, R. Blau, I. Schieferdecker, A. Teleanu, V. Munder, M. Röllig, C. Salwender, H.-J. Fuhrmann, S. Weisel, K. Duerig, J. Zeis, M. Klein, S. Reimer, P. Schmidt, C. Scheid, C. Mayer, K. Hoffmann, M. Sosada, M. Dimopoulos, A. Delimpasi, S. Kyrtsonis, M.-C. Anagnostopoulos, A. Nagy, Z. Illés, Á. Egyed, M. Borbényi, Z. Mikala, G. Dally, N. Horowitz, N. Gutwein, O. Nemets, A. Vaxman, I. Shvetz, O. Trestman, S. Ruchlemer, R. Nagler, A. Tadmor, T. Rouvio, O. Preis, M. Cavo, M. De Rosa, L. Musto, P. Cafro, A. Tosi, P. Offidani, M. Corso, A. Rossi, G. Liberati, A.M. Bosi, A. Suzuki, K. Nakaseko, C. Ishikawa, T. Matsumoto, M. Nagai, H. Sunami, K. Chou, T. Akashi, K. Takezako, N. Hagiwara, S. Eom, H.S. Jo, D.-Y. Kim, J.S. Lee, J.H. Yoon, S.S. Yoon, D.H. Kim, K. Levin, M.-D. Vellenga, E. Minnema, M. Waage, A. Haukås, E. Grosicki, S. Pluta, A. Robak, T. Marques, H. Bergantim, R. Campilho, F. Chng, W.J. Goh, Y.T. McDonald, A. Rapoport, B. Álvarez Rivas, M.A. De Arriba de La Fuente, F. González Montes, Y. Martin Sanchez, J. Mateos, M.V. Oriol Rocafiguera, A. Rosinol, L. San Miguel, J. Pérez de Oteyza, J. Encinas, C. Alegre-Amor, A. López-Guía, A. Axelsson, P. Carlson, K. Stromberg, O. Hansson, M. Hveding Blimark, C. Mueller, R. Chen, C.-C. Liu, T.-C. Huang, S.-Y. Wang, P.-N. Na Nakorn, T. Prayongratana, K. Unal, A. Goker, H. Sonmez, M. Korenkova, S. Chaidos, A. Oakervee, H. Sati, H. Benjamin, R. Wechalekar, A. Garg, M. Ramasamy, K. Cook, G. Chantry, A. Jenner, M. Buadi, F. Berryman, R. Janakiram, M. TOURMALINE-MM3 study group
- Abstract
Background: Maintenance therapy following autologous stem cell transplantation (ASCT) can delay disease progression and prolong survival in patients with multiple myeloma. Ixazomib is ideally suited for maintenance therapy given its convenient once-weekly oral dosing and low toxicity profile. In this study, we aimed to determine the safety and efficacy of ixazomib as maintenance therapy following ASCT. Methods: The phase 3, double-blind, placebo-controlled TOURMALINE-MM3 study took place in 167 clinical or hospital sites in 30 countries in Europe, the Middle East, Africa, Asia, and North and South America. Eligible participants were adults with a confirmed diagnosis of symptomatic multiple myeloma according to International Myeloma Working Group criteria who had achieved at least a partial response after undergoing standard-of-care induction therapy followed by high-dose melphalan (200 mg/m2) conditioning and single ASCT within 12 months of diagnosis. Patients were randomly assigned in a 3:2 ratio to oral ixazomib or matching placebo on days 1, 8, and 15 in 28-day cycles for 2 years following induction, high-dose therapy, and transplantation. The initial 3 mg dose was increased to 4 mg from cycle 5 if tolerated during cycles 1–4. Randomisation was stratified by induction regimen, pre-induction disease stage, and response post-transplantation. The primary endpoint was progression-free survival (PFS) by intention-to-treat analysis. Safety was assessed in all patients who received at least one dose of ixazomib or placebo, according to treatment actually received. This trial is registered with ClinicalTrials.gov, number NCT02181413, and follow-up is ongoing. Findings: Between July 31, 2014, and March 14, 2016, 656 patients were enrolled and randomly assigned to receive ixazomib maintenance therapy (n=395) or placebo (n=261). With a median follow-up of 31 months (IQR 27·3–35·7), we observed a 28% reduction in the risk of progression or death with ixazomib versus placebo (median PFS 26·5 months [95% CI 23·7–33·8] vs 21·3 months [18·0–24·7]; hazard ratio 0·72, 95% CI 0·58–0·89; p=0·0023). No increase in second malignancies was noted with ixazomib therapy (12 [3%] patients) compared with placebo (eight [3%] patients) at the time of this analysis. 108 (27%) of 394 patients in the ixazomib group and 51 (20%) of 259 patients in the placebo group experienced serious adverse events. During the treatment period, one patient died in the ixazomib group and none died in the placebo group. Interpretation: Ixazomib maintenance prolongs PFS and represents an additional option for post-transplant maintenance therapy in patients with newly diagnosed multiple myeloma. Funding: Millennium Pharmaceuticals, a wholly owned subsidiary of Takeda Pharmaceutical Company. © 2019 Elsevier Ltd
- Published
- 2019
7. Secondary immunodeficiency in lymphoproliferative malignancies
- Author
-
Friman, V, Winqvist, O, Blimark, C, Langerbeins, P, Chapel, H, and Dhalla, F
- Subjects
Male ,Hematologic Neoplasms ,hemic and lymphatic diseases ,Immunologic Deficiency Syndromes ,Humans ,Female ,Multiple Myeloma ,Leukemia, Lymphocytic, Chronic, B-Cell - Abstract
Secondary immunodeficiencies occur as a consequence of various diseases, including hematological malignancies, and the use of pharmacological therapies, such as immunosuppressive, anti-inflammatory, and biological drugs. Infections are the main cause of morbidity and mortality in multiple myeloma (MM) and chronic lymphocytic leukemia (CLL) patients. Recent advances in treatment have prolonged the duration of remission and the time between relapse phases in MM and CLL patients. However, managing multiple relapses and the use of salvage therapies can lead to cumulative immunosuppression and a higher risk of infections. The pathogenesis of immune deficiency secondary to lymphoproliferative malignancy is multifactorial including disease- and treatment-related factors. Supportive treatment, including early vaccination, anti-infective prophylaxis, and replacement immunoglobulin, plays a key role in preventing infections in MM and CLL. This article provides an overview of the basic immunology necessary to understand the pathogenesis of secondary immunodeficiency and the infectious complications in MM and CLL. We also discuss the evidence supporting the role of prophylactic replacement immunoglobulin treatment in patients with antibody failure secondary to MM and CLL and the indications for its use. Copyright © 2016 John WileySons, Ltd.
- Published
- 2016
8. Improved progression free survival with bortezomib consolidation after high dose melphalan: results of a randomised phase III trial
- Author
-
Mellquist, U. H., Gimsing, P, Hjertner, O, Lenhoff, S, Laane, E, Remes, K, Steingrimsdottir, H, Abildgaard , Niels, Ahlberg, L, Blimark, C, Carlsson, K, Dahl, I M, Forsberg, K, Andersen, N F, Gedde-Dahl, T, Gregersen, H, Gruber, A, Guldbrandsen, N, Haukas, E, Juliusson, G, Lindas, R, Linder, N, Nahi, N, Silvennoinen, R, Swedin, A, Toffner-Clausen, N, Turesson, I, Waage, A, and Westin, J
- Published
- 2011
9. THE REVII TRIAL : LENALIDOMIDE AND DEXAMETHASONE AS SECOND LINE TREATMENT IN MYELOMA FOLLOWED BY EXTENDED LENALIDOMID VS LEN/DEX
- Author
-
Lund, J., Gruber, A., Lauri, B., Blimark, C., Swedin, A., Hansson, M., Forsberg, Karin, Ahlberg, L., Carlsson, C., Waage, A., Gimsing, P., Vangsted, A. J., Frolund, U., Hardling, M., Mellqvist, U. H., Nahi, H., Lund, J., Gruber, A., Lauri, B., Blimark, C., Swedin, A., Hansson, M., Forsberg, Karin, Ahlberg, L., Carlsson, C., Waage, A., Gimsing, P., Vangsted, A. J., Frolund, U., Hardling, M., Mellqvist, U. H., and Nahi, H.
- Published
- 2014
10. MULTIPLE MYELOMA PATIENTS THAT DEVELOP SECOND PRIMARY MALIGNANCY HAVE A WORSE PROGNOSIS THAN MULTIPLE MYELOMA PATIENTS TREATED BEFORE THE INTRODUCTION OF NOVEL AGENTS : A POPULATION BASED-STUDY
- Author
-
Jonsdottir, G., Lund, S. H., Landgren, O., Bjorkholm, M., Turesson, I., Wahlin, Anders, Blimark, C., Hultcrantz, M., Porwit, A., Kristinsson, S. Y., Jonsdottir, G., Lund, S. H., Landgren, O., Bjorkholm, M., Turesson, I., Wahlin, Anders, Blimark, C., Hultcrantz, M., Porwit, A., and Kristinsson, S. Y.
- Published
- 2014
11. Multiple myeloma and infections: a population-based study on 9253 multiple myeloma patients
- Author
-
Blimark, C., primary, Holmberg, E., additional, Mellqvist, U.-H., additional, Landgren, O., additional, Bjorkholm, M., additional, Hultcrantz, M., additional, Kjellander, C., additional, Turesson, I., additional, and Kristinsson, S. Y., additional
- Published
- 2014
- Full Text
- View/download PDF
12. Monoclonal gammopathy of undetermined significance and risk of infections: a population-based study
- Author
-
Kristinsson, S. Y., primary, Tang, M., additional, Pfeiffer, R. M., additional, Bjorkholm, M., additional, Goldin, L. R., additional, Blimark, C., additional, Mellqvist, U.-H., additional, Wahlin, A., additional, Turesson, I., additional, and Landgren, O., additional
- Published
- 2011
- Full Text
- View/download PDF
13. Patterns of survival and causes of death following a diagnosis of monoclonal gammopathy of undetermined significance: a population-based study
- Author
-
Kristinsson, S. Y., primary, Bjorkholm, M., additional, Andersson, T. M-L, additional, Eloranta, S., additional, Dickman, P. W., additional, Goldin, L. R., additional, Blimark, C., additional, Mellqvist, U.-H., additional, Wahlin, A., additional, Turesson, I., additional, and Landgren, O., additional
- Published
- 2009
- Full Text
- View/download PDF
14. MULTIPLE MYELOMA PATIENTS THAT DEVELOP SECOND PRIMARY MALIGNANCY HAVE A WORSE PROGNOSIS THAN MULTIPLE MYELOMA PATIENTS TREATED BEFORE THE INTRODUCTION OF NOVEL AGENTS: A POPULATION BASED-STUDY
- Author
-
Jonsdottir, G., Sigrún Helga Lund, Landgren, O., Bjorkholm, M., Turesson, I., Wahlin, A., Blimark, C., Hultcrantz, M., Porwit, A., and Kristinsson, S. Y.
15. Oral ixazomib maintenance following autologous stem cell transplantation (TOURMALINE-MM3): a double-blind, randomised, placebo-controlled phase 3 trial
- Author
-
Meletios A Dimopoulos, Francesca Gay, Fredrik Schjesvold, Meral Beksac, Roman Hajek, Katja Christina Weisel, Hartmut Goldschmidt, Vladimir Maisnar, Philippe Moreau, Chang Ki Min, Agnieszka Pluta, Wee-Joo Chng, Martin Kaiser, Sonja Zweegman, Maria-Victoria Mateos, Andrew Spencer, Shinsuke Iida, Gareth Morgan, Kaveri Suryanarayan, Zhaoyang Teng, Tomas Skacel, Antonio Palumbo, Ajeeta B Dash, Neeraj Gupta, Richard Labotka, S Vincent Rajkumar, Daniel Bar, Alfredo Basso, Dorotea Fantl, Simon He, Neomi Horvath, Cindy Lee, Phillip Rowlings, Kerry Taylor, Tara Cochrane, Fiona Kwok, Sundreswran Ramanathan, Hermine Agis, Niklas Zojer, Alain Kentos, Fritz Offner, Jan Van Droogenbroeck, Ka Lung Wu, Angelo Maiolino, Gracia Martinez, Karla Zanella, Marcelo Capra, Sérgio Araújo, Evzen Gregora, Ludek Pour, Vlastimil Scudla, Ivan Spicka, Niels Abildgaard, Niels Andersen, Bo Amdi Jensen, Carsten Helleberg, Torben Plesner, Morten Salomo, Asta Svirskaite, Richard Delarue, Igor Blau, Aneta Schieferdecker, Veronica Teleanu, Markus Munder, Christoph Röllig, Han-Juergen Salwender, Stephan Fuhrmann, Katja Weisel, Jan Duerig, Matthias Zeis, Stefan Klein, Peter Reimer, Christian Schmidt, Christof Scheid, Karin Mayer, Martin Hoffmann, Markus Sosada, Athanasios Dimopoulos, Sosana Delimpasi, Mary-Christine Kyrtsonis, Achilleas Anagnostopoulos, Zsolt Nagy, Árpád Illés, Miklós Egyed, Zita Borbényi, Gabor Mikala, Najib Dally, Netanel Horowitz, Odit Gutwein, Anatoly Nemets, Iuliana Vaxman, Olga Shvetz, Svetlana Trestman, Rosa Ruchlemer, Arnon Nagler, Tamar Tadmor, Ory Rouvio, Meir Preis, Michele Cavo, Luca De Rosa, Pellegrino Musto, Anna Cafro, Patrizia Tosi, Massimo Offidani, Alessandro Corso, Giuseppe Rossi, Anna Marina Liberati, Alberto Bosi, Kenshi Suzuki, Chiaki Nakaseko, Takayuki Ishikawa, Morio Matsumoto, Hirokazu Nagai, Kazutaka Sunami, Takaaki Chou, Koichi Akashi, Naoki Takezako, Shotaro Hagiwara, Hyeon Seok Eom, Deog-Yeon Jo, Jin Seok Kim, Jae Hoon Lee, Sung Soo Yoon, Dok Hyun Yoon, Kihyun Kim, Mark-David Levin, Edo Vellenga, Monique Minnema, Anders Waage, Einar Haukås, Sebastian Grosicki, Andrzej Pluta, Tadeusz Robak, Herlander Marques, Rui Bergantim, Fernando Campilho, Wee Joo Chng, Yeow Tee Goh, Andrew McDonald, Bernado Rapoport, Miguel Angel Álvarez Rivas, Felipe De Arriba de La Fuente, Yolanda González Montes, Jesus Martin Sanchez, Maria Victoria Mateos, Albert Oriol Rocafiguera, Laura Rosinol, Jesús San Miguel, Jaime Pérez de Oteyza, Cristina Encinas, Adrian Alegre-Amor, Ana López-Guía, Per Axelsson, Kristina Carlson, Olga Stromberg, Markus Hansson, Cecile Hveding Blimark, Rouven Mueller, Chih-Cheng Chen, Ta-Chih Liu, Shang-Yi Huang, Po-Nan Wang, Thanyaphong Na Nakorn, Kannadit Prayongratana, Ali Unal, Hakan Goker, Mehmet Sonmez, Sybiryna Korenkova, Aristeidis Chaidos, Heather Oakervee, Hamdi Sati, Reuben Benjamin, Ashutosh Wechalekar, Mamta Garg, Karthik Ramasamy, Gordon Cook, Andrew Chantry, Matthew Jenner, Francis Buadi, Robert Berryman, Murali Janakiram, Takeda Pharmaceutical Company, Dimopoulos MA1, Gay F2, Schjesvold F3, Beksac M4, Hajek R5, Weisel KC6, Goldschmidt H7, Maisnar V8, Moreau P9, Min CK10, Pluta A11, Chng WJ12, Kaiser M13, Zweegman S14, Mateos MV15, Spencer A16, Iida S17, Morgan G18, Suryanarayan K19, Teng Z19, Skacel T19, Palumbo A20, Dash AB19, Gupta N19, Labotka R19, Rajkumar SV21, TOURMALINE-MM3 study group. Bar D, Basso A, Fantl D, He S, Horvath N, Lee C, Rowlings P, Taylor K, Spencer A, Cochrane T, Kwok F, Ramanathan S, Agis H, Zojer N, Kentos A, Offner F, Van Droogenbroeck J, Wu KL, Maiolino A, Martinez G, Zanella K, Capra M, Araújo S, Gregora E, Hajek R, Maisnar V, Pour L, Scudla V, Spicka I, Abildgaard N, Andersen N, Jensen BA, Helleberg C, Plesner T, Salomo M, Svirskaite A, Delarue R, Moreau P, Blau I, Goldschmidt H, Schieferdecker A, Teleanu V, Munder M, Röllig C, Salwender HJ, Fuhrmann S, Weisel K, Duerig J, Zeis M, Klein S, Reimer P, Schmidt C, Scheid C, Mayer K, Hoffmann M, Sosada M, Dimopoulos A, Delimpasi S, Kyrtsonis MC, Anagnostopoulos A, Nagy Z, Illés Á, Egyed M, Borbényi Z, Mikala G, Dally N, Horowitz N, Gutwein O, Nemets A, Vaxman I, Shvetz O, Trestman S, Ruchlemer R, Nagler A, Tadmor T, Rouvio O, Preis M, Gay F, Cavo M, De Rosa L, Musto P, Cafro A, Tosi P, Offidani M, Corso A, Rossi G, Liberati AM, Bosi A, Suzuki K, Iida S, Nakaseko C, Ishikawa T, Matsumoto M, Nagai H, Sunami K, Chou T, Akashi K, Takezako N, Hagiwara S, Eom HS, Jo DY, Kim JS, Lee JH, Min CK, Yoon SS, Yoon DH, Kim K, Zweegman S, Levin MD, Vellenga E, Minnema M, Schjesvold F, Waage A, Haukås E, Grosicki S, Pluta A, Robak T, Marques H, Bergantim R, Campilho F, Chng WJ, Goh YT, McDonald A, Rapoport B, Álvarez Rivas MA, De Arriba de La Fuente F, González Montes Y, Martin Sanchez J, Mateos MV, Oriol Rocafiguera A, Rosinol L, San Miguel J, Pérez de Oteyza J, Encinas C, Alegre-Amor A, López-Guía A, Axelsson P, Carlson K, Stromberg O, Hansson M, Hveding Blimark C, Mueller R, Chen CC, Liu TC, Huang SY, Wang PN, Na Nakorn T, Prayongratana K, Beksac M, Unal A, Goker H, Sonmez M, Korenkova S, Chaidos A, Oakervee H, Sati H, Benjamin R, Wechalekar A, Garg M, Kaiser M, Ramasamy K, Cook G, Chantry A, Jenner M, Buadi F, Berryman R, Janakiram M., Guided Treatment in Optimal Selected Cancer Patients (GUTS), Stem Cell Aging Leukemia and Lymphoma (SALL), CCA - Cancer Treatment and quality of life, CCA - Imaging and biomarkers, CCA - Cancer biology and immunology, and Hematology
- Subjects
Male ,Time Factors ,DIAGNOSED MULTIPLE-MYELOMA ,Clinical Trial, Phase III ,Administration, Oral ,030204 cardiovascular system & hematology ,Ixazomib ,chemistry.chemical_compound ,0302 clinical medicine ,Autologous stem-cell transplantation ,Maintenance therapy ,Clinical endpoint ,030212 general & internal medicine ,Non-U.S. Gov't ,Boron Compounds/administration & dosage ,IMPROVES SURVIVAL ,INDUCTION ,Research Support, Non-U.S. Gov't ,General Medicine ,CHEMOTHERAPY ,Middle Aged ,Clinical Trial ,DEXAMETHASONE ,Antineoplastic Agents/administration & dosage ,Multicenter Study ,Treatment Outcome ,Administration ,Randomized Controlled Trial ,Disease Progression ,Female ,Multiple Myeloma ,Autologous ,Boron Compounds ,Oral ,medicine.medical_specialty ,Glycine ,Multiple Myeloma/drug therapy ,BORTEZOMIB ,Antineoplastic Agents ,Placebo ,Research Support ,Transplantation, Autologous ,03 medical and health sciences ,Phase III ,Double-Blind Method ,Internal medicine ,medicine ,Journal Article ,Humans ,THALIDOMIDE ,Transplantation ,business.industry ,Clinical trial ,LENALIDOMIDE MAINTENANCE ,Regimen ,chemistry ,autologous stem cell transplantation, multiple myeloma, Ixazomib ,business ,HIGH-DOSE THERAPY ,Glycine/administration & dosage ,Stem Cell Transplantation - Abstract
[Background]: Maintenance therapy following autologous stem cell transplantation (ASCT) can delay disease progression and prolong survival in patients with multiple myeloma. Ixazomib is ideally suited for maintenance therapy given its convenient once-weekly oral dosing and low toxicity profile. In this study, we aimed to determine the safety and efficacy of ixazomib as maintenance therapy following ASCT. [Methods]: The phase 3, double-blind, placebo-controlled TOURMALINE-MM3 study took place in 167 clinical or hospital sites in 30 countries in Europe, the Middle East, Africa, Asia, and North and South America. Eligible participants were adults with a confirmed diagnosis of symptomatic multiple myeloma according to International Myeloma Working Group criteria who had achieved at least a partial response after undergoing standard-of-care induction therapy followed by high-dose melphalan (200 mg/m²) conditioning and single ASCT within 12 months of diagnosis. Patients were randomly assigned in a 3:2 ratio to oral ixazomib or matching placebo on days 1, 8, and 15 in 28-day cycles for 2 years following induction, high-dose therapy, and transplantation. The initial 3 mg dose was increased to 4 mg from cycle 5 if tolerated during cycles 1–4. Randomisation was stratified by induction regimen, pre-induction disease stage, and response post-transplantation. The primary endpoint was progression-free survival (PFS) by intention-to-treat analysis. Safety was assessed in all patients who received at least one dose of ixazomib or placebo, according to treatment actually received. This trial is registered with ClinicalTrials.gov, number NCT02181413, and follow-up is ongoing. [Findings]: Between July 31, 2014, and March 14, 2016, 656 patients were enrolled and randomly assigned to receive ixazomib maintenance therapy (n=395) or placebo (n=261). With a median follow-up of 31 months (IQR 27·3–35·7), we observed a 28% reduction in the risk of progression or death with ixazomib versus placebo (median PFS 26·5 months [95% CI 23·7–33·8] vs 21·3 months [18·0–24·7]; hazard ratio 0·72, 95% CI 0·58–0·89; p=0·0023). No increase in second malignancies was noted with ixazomib therapy (12 [3%] patients) compared with placebo (eight [3%] patients) at the time of this analysis. 108 (27%) of 394 patients in the ixazomib group and 51 (20%) of 259 patients in the placebo group experienced serious adverse events. During the treatment period, one patient died in the ixazomib group and none died in the placebo group. [Interpretation]: Ixazomib maintenance prolongs PFS and represents an additional option for post-transplant maintenance therapy in patients with newly diagnosed multiple myeloma, This study was sponsored by Millennium Pharmaceuticals, a wholly owned subsidiary of Takeda Pharmaceutical Company.
- Published
- 2019
16. The choice of serum-free light chain analysis method could potentially have clinical consequences for myeloma patients.
- Author
-
Veskovski L, Jakobsson I, Andersson PO, Gustafsson T, Sedigh A, Knut-Bojanowska D, Hansson M, Hveding Blimark C, and Mellqvist UH
- Abstract
Multiple myeloma (MM) is a disease, that at times poses diagnostic and monitoring challenges. Over the last decades laboratory methods have been expanded with serum free light chain (FLC) analysis. Alerted by two index cases with clinical impact due to failure of the FLC analysis to indicate a disease progression, we aimed to identify any clinical consequences due to known differences between FLC analysis methods. We applied two FLC analysis methods (Freelite Binding Site [FBS] and N-Latex Siemens [NLS]) on all patients with MM and monoclonal gammopathy of uncertain significance diagnosed/followed up at Södra Älvsborg Hematology Unit, from April to December 2022. From a total of 123 patients with malignant plasma cell disorder, we identified five cases (4.1%) where solely the FBS method, as opposed to NLS, urine and serum electrophoresis, could support diagnosis or detect progression. The consequences of this discrepancy included not only change of diagnosis or delayed therapy but also change of treatment. Our findings indicate that a stronger awareness of the potential weaknesses of different FLC methods is needed, which calls for a closer collaboration between clinical chemists and hematologists., Competing Interests: The authors declare no conflict of interest., (© 2024 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
17. Measurable Residual Disease Testing in Multiple Myeloma Routine Clinical Practice: A Modified Delphi Study.
- Author
-
Ramasamy K, Avet-Loiseau H, Hveding Blimark C, Delforge M, Gay F, Manier S, Martinez-Lopez J, Mateos MV, Mohty M, van de Donk NWCJ, and Weisel K
- Abstract
We used a modified Delphi approach to establish areas of consensus and nonconsensus regarding the utility of determining measurable residual disease (MRD) to assess multiple myeloma (MM) treatment response, which may inform disease management and design of future clinical trials. This modified Delphi study incorporated 2 iterative rounds of surveys to evaluate the opinions of an expert panel of 61 practicing hematological oncologists from across 14 countries in Europe concerning the use of MRD testing in MM management. Survey 1 assessed experts' opinions on MRD testing in different clinical situations and associated challenges. Survey 2 focused on the lack of consensus areas identified in survey 1. Consensus to an individual question was defined a priori as 75% agreement or disagreement by the panel. From the 2 rounds of surveys, the experts reached consensus agreement that MRD testing should be performed in newly diagnosed or relapsed patients who achieved complete response (CR) or better after transplantation. In transplant-ineligible patients, experts recommended MRD testing in those who are ≤70 years old and in CR. If a patient was previously positive on positron-emission tomography and computed tomography (PET/CT), both MRD and PET/CT should be assessed at CR. MRD testing should be performed ≤6 months after transplantation and every 6-12 months in continuously treated patients in CR. There was no consensus on making treatment decisions based on MRD status. MRD testing is an important component of clinical management in MM. Additional data will further clarify the role of MRD in guiding treatment decisions., Competing Interests: KR: Has served as an advisor for and received honoraria from Janssen, Adaptive Biotechnologies, Amgen, Takeda, Abbvie, Oncopeptides, Celgene – Bristol Myers Squibb, Pfizer, and GSK. Has received grant funding from Janssen, Amgen, Takeda, GSK and Celgene – Bristol Myers Squibb. HA-L: Has served as an advisor for and received honoraria from Amgen, Bristol Myers Squibb, Celgene, Janssen, Sanofi, GSK, Takeda, and Adaptive Biotechnologies. CHB: Has served as an advisor for and received honoraria from Amgen, Bristol Myers Squibb, Takeda, Sanofi, GSK, and Adaptive Biotechnologies. MD: Has served as an advisor for and received honoraria from Amgen, Celgene – Bristol Myers Squibb, GSK, Janssen, Sanofi, and Takeda. FG: Has served as an advisor for Janssen, Amgen, Celgene – Bristol Myers Squibb, Adaptive Biotechnologies, Roche, Abbvie, GSK, Takeda, Bluebird Bio, Oncopeptides, Pfizer, and Sanofi. Has received honoraria from Janssen, Amgen, Celgene – Bristol Myers Squibb, GSK, Takeda, and Sanofi. SM: Has served as an advisor for Abbvie, Adaptive Biotechnologies, Amgen, Celgene – Bristol Myers Squibb, GSK, Janssen, Oncopeptides, Pfizer, Regeneron, Roche, Sanofi, and Takeda. JM-L: Has served as an advisor for and received honoraria from Amgen, Bristol Myers Squibb, Celgene, Janssen, Sanofi, GSK, Incyte, Roche, Pfizer, Novartis and Adaptive Biotechnologies. MVM: Has served as an advisor for and received honoraria from Janssen, Celgene – Bristol Myers Squibb, Takeda, Amgen, Adaptive Biotechnologies, Oncopeptides, Sanofi, Roche, Regeneron, and Pfizer. MM: Has received honoraria from Amgen, Astellas, Bristol Myers Squibb, Celgene, Gilead, Janssen, Jazz, Takeda, Novartis, Pfizer, Sanofi, and Adaptive Biotechnologies. Has received research funding from Celgene, Janssen, Jazz, and Sanofi. NWCJvdD: Has served as an advisor for Janssen Pharmaceuticals, Amgen, Adaptive Biotechnologies, Celgene, Bristol Myers Squibb, Novartis, Roche, Takeda, Bayer, and Servier. Has received grant funding from Janssen Pharmaceuticals, Amgen, Celgene, Cellectis, and Bristol Myers Squibb. KW: Has served as an advisor for Amgen, Adaptive Biotechnologies, Bristol Myers Squibb, Celgene, GSK, Janssen, Karyopharm, Oncopeptides, Sanofi, and Takeda. Has received honoraria from Amgen, Adaptive Biotechnologies, Bristol Myers Squibb, Celgene, GSK, Janssen, Karyopharm, Oncopeptides, Sanofi, Takeda, and Roche. Has received grant funding from Amgen, Janssen, Celgene, and Sanofi (to the institution)., (Copyright © 2023 the Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the European Hematology Association.)
- Published
- 2023
- Full Text
- View/download PDF
18. Comorbidities in multiple myeloma and implications on survival: A population-based study.
- Author
-
Sverrisdóttir IS, Rögnvaldsson S, Thorsteinsdottir S, Gíslason GK, Aspelund T, Turesson I, Björkholm M, Gregersen H, Hveding Blimark C, Landgren O, and Kristinsson SY
- Subjects
- Adult, Aged, Aged, 80 and over, Comorbidity, Disease-Free Survival, Female, Follow-Up Studies, Humans, Male, Middle Aged, Prevalence, Risk Factors, Survival Rate, Sweden epidemiology, Multiple Myeloma mortality, Registries
- Abstract
High proportion of patients with multiple myeloma suffer from comorbidities which may alter clinical management. Therefore, our aims were to evaluate the prevalence of comorbidities and their impact on survival. We included patients diagnosed with multiple myeloma 1990-2013 in Sweden and all diagnoses from each patient from 1985. A total of 13 656 patients with multiple myeloma were included in the study, thereof 7404 (54%) had comorbidity at diagnosis. The risk of death was increased for those with one comorbidity at diagnosis compared to those without any comorbidity (hazard ratio = 1.19; 95% confidence interval:1.14-1.25); this risk was higher for those with two (1.38; 1.30-1.47) and three or more comorbidities (1.72; 1.62-1.83). Furthermore, the risk of death was increased in patients with prior history of cancer, arrhythmia, heart failure, diabetes mellitus, cerebrovascular disease, chronic lung disease, psychological disease, peptic ulcer, neurological disease, peripheral vascular disease, chronic kidney disease, dementia, and inflammatory bowel disease. This large study shows that over 50% of multiple myeloma patients have a comorbidity at diagnosis and survival decreased with increasing numbers of comorbidities. This emphasizes the importance of comorbidities when evaluating patients and deciding on treatment strategies for individuals with multiple myeloma., (© 2021 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
19. Ixazomib-Thalidomide-low dose dexamethasone induction followed by maintenance therapy with ixazomib or placebo in newly diagnosed multiple myeloma patients not eligible for autologous stem cell transplantation; results from the randomized phase II HOVON-126/NMSG 21.13 trial.
- Author
-
Zweegman S, Stege CAM, Haukas E, Schjesvold FH, Levin MD, Waage A, Leys RBL, Klein SK, Szatkowski D, Axelsson P, Hieu Do T, Knut-Bojanowska D, van der Spek E, Svirskaite A, Klostergaard A, Salomo M, Blimark C, Ypma PF, Mellqvist UH, Poddighe PJ, Stevens-Kroef M, van de Donk NWCJ, Sonneveld P, Hansson M, van der Holt B, and Abildgaard N
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Boron Compounds, Bortezomib therapeutic use, Dexamethasone therapeutic use, Glycine analogs & derivatives, Humans, Stem Cell Transplantation, Thalidomide therapeutic use, Transplantation, Autologous, Treatment Outcome, Hematopoietic Stem Cell Transplantation, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy
- Published
- 2020
- Full Text
- View/download PDF
20. Outcome and characteristics of non-measurable myeloma: A cohort study with population-based data from the Swedish Myeloma Registry.
- Author
-
Wålinder G, Samuelsson J, Näsman P, Hansson M, Juliusson G, Forsberg K, Svensson R, Linder O, Carlson K, Kristinsson SY, Mellqvist UH, Hveding Blimark C, Turesson I, and Nahi H
- Subjects
- Adult, Aged, Aged, 80 and over, Asymptomatic Diseases, Biomarkers, Cohort Studies, Combined Modality Therapy, Female, Humans, Immunoglobulin Light Chains, Male, Middle Aged, Multiple Myeloma diagnosis, Multiple Myeloma metabolism, Multiple Myeloma therapy, Myeloma Proteins, Neoplasm Staging, Patient Outcome Assessment, Population Surveillance, Prognosis, Registries, Sweden epidemiology, Treatment Outcome, Young Adult, Multiple Myeloma epidemiology
- Abstract
Objective: We describe survival in patients with oligo- and non-secretory multiple myeloma (MM). We refer to the whole group as non-measurable MM and compare it with secretory MM., Methods: Oligo-secretory MM was defined as M protein in serum <10 g/L and M protein in urine <200 measured as mg/day, mg/liter or mg/mmol creatinine. If patients had no M protein, they were defined as non-secretory. The groups were also subdivided by Free Light Chains (SFLC) level and ratio., Results: Out of 4325 patients with symptomatic MM in the Swedish Myeloma Registry during 2008-2016 eligible for the study, 389 patients (9%) had non-measurable MM. Out of these, 253 patients (6%) had oligo-secretory and 136 (3%) had non-secretory MM. Median survival for secretory MM was 42.7 months, non-measurable MM 40.2 months, oligo-secretory MM 38.6 months, and non-secretory MM 44.6 months. Difference in overall observed survival was non-significant for all groups when compared with secretory MM. Within non-secretory MM, stem cell transplantation (SCT), 95% being auto-SCT, was significant for superior survival in multivariate analysis (HR 0.048. P = .0015)., Conclusion: In this population-based study, we found no difference in survival between oligo- or non-secretory MM when compared with secretory MM. SCT appears to be important also for patients with non-secretory disease., (© 2019 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
21. Dramatically improved survival in multiple myeloma patients in the recent decade: results from a Swedish population-based study.
- Author
-
Thorsteinsdottir S, Dickman PW, Landgren O, Blimark C, Hultcrantz M, Turesson I, Björkholm M, and Kristinsson SY
- Subjects
- History, 20th Century, History, 21st Century, Humans, Multiple Myeloma epidemiology, Multiple Myeloma history, Population Surveillance, Registries, Survival Rate, Sweden epidemiology, Multiple Myeloma mortality
- Published
- 2018
- Full Text
- View/download PDF
22. Lenalidomide versus lenalidomide + dexamethasone prolonged treatment after second-line lenalidomide + dexamethasone induction in multiple myeloma.
- Author
-
Lund J, Gruber A, Lauri B, Duru AD, Blimark C, Swedin A, Hansson M, Forsberg K, Ahlberg L, Carlsson C, Waage A, Gimsing P, Vangsted AJ, Frølund U, Holmberg E, Gahrton G, Alici E, Hardling M, Mellqvist UH, and Nahi H
- Subjects
- Adult, Aged, Aged, 80 and over, Angiogenesis Inhibitors pharmacology, Anti-Inflammatory Agents pharmacology, Dexamethasone pharmacology, Female, Humans, Lenalidomide pharmacology, Male, Middle Aged, Multiple Myeloma pathology, Angiogenesis Inhibitors therapeutic use, Anti-Inflammatory Agents therapeutic use, Dexamethasone therapeutic use, Lenalidomide therapeutic use, Multiple Myeloma drug therapy
- Abstract
Lenalidomide (Len) plus dexamethasone (Dex) is approved for the treatment of relapsed or refractory multiple myeloma (RRMM). It is possible that single-agent Len may be effective as prolonged treatment regimen in RRMM once patients demonstrate an initial response to Len+Dex induction. Patients with RRMM who responded to first-line Len+Dex in an observational study (NCT01430546) received up to 24 cycles of either Len (25 mg/day) or Len+Dex (25 mg/day and 40 mg/week) as prolonged treatment in a subsequent phase 2 clinical trial (NCT01450215). In the observational study (N = 133), median time to response was 1.7 (range 0.6-9.6) months. A complete response to all treatments received in both studies was observed in 11% of patients; very good partial response and partial response rates were 31% and 38%, respectively. Corresponding response rates in the subgroup of patients who did not enter the phase 2 trial (n = 71) were 3%, 18%, and 39%, respectively. Rates of disease progression at 2 years in the phase 2 trial were 47% versus 31% for Len versus Len+Dex (P = 0.14). After 36 months median follow-up in surviving patients, median time to progression was not reached with Len+Dex and was 24.9 months (95% confidence interval 12.5-not calculable, P < 0.001) with Len. Three-year OS among the total observational study population was 61% (95% CI, 52-69%). The corresponding rate among patients who entered the phase 2 clinical trial was 73% (95% CI, 60-83%) and was significantly lower among those patients who achieved ≥PR but did not proceed into the phase 2 trial (55%; P = 0.01). In the phase 2 trial, OS was 73% in both treatment arms (P = 0.70). Neutropenia and thrombocytopenia were more common with prolonged (phase 2 trial) versus short-term (observational study) Len administration but remained manageable. Prolonged treatment with Len with or without Dex provides sustained, clinically relevant responses and demonstrates an acceptable safety profile., (© 2018 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
23. Pharmacogenetic study of the impact of ABCB1 single-nucleotide polymorphisms on lenalidomide treatment outcomes in patients with multiple myeloma: results from a phase IV observational study and subsequent phase II clinical trial.
- Author
-
Jakobsen Falk I, Lund J, Gréen H, Gruber A, Alici E, Lauri B, Blimark C, Mellqvist UH, Swedin A, Forsberg K, Carlsson C, Hardling M, Ahlberg L, Lotfi K, and Nahi H
- Subjects
- ATP Binding Cassette Transporter, Subfamily B genetics, Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Disease Progression, Female, Humans, Lenalidomide adverse effects, Male, Middle Aged, Prospective Studies, Skin Diseases chemically induced, Treatment Outcome, Antineoplastic Agents therapeutic use, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Pharmacogenomic Testing, Polymorphism, Single Nucleotide
- Abstract
Purpose: Despite therapeutic advances, patients with multiple myeloma (MM) continue to experience disease relapse and treatment resistance. The gene ABCB1 encodes the drug transporter P-glycoprotein, which confers resistance through drug extrusion across the cell membrane. Lenalidomide (Len) is excreted mainly via the kidneys, and, given the expression of P-gp in the renal tubuli, single-nucleotide polymorphisms (SNPs) in the ABCB1 gene may influence Len plasma concentrations and, subsequently, the outcome of treatment. We, therefore, investigated the influence of ABCB1 genetic variants on Len treatment outcomes and adverse events (AEs)., Methods: Ninety patients with relapsed or refractory MM, who received the second-line Len plus dexamethasone in the Rev II trial, were genotyped for the ABCB1 SNPs 1199G>A (Ser400Asn, rs2229109), 1236C>T (silent, rs1128503), 2677G>T/A (Ala893Ser, rs2032582), and 3435C>T (silent, rs1045642) using pyrosequencing, and correlations to response parameters, outcomes, and AEs were investigated., Results: No significant associations were found between genotype and either best response rates or hematological AEs, and 1236C>T, 2677G>T or 3435C>T genotypes had no impact on survival. There was a trend towards increased time to progression (TTP) in patients carrying the 1199A variant, and a significant difference in TTP between genotypes in patients with standard-risk cytogenetics., Conclusions: Our findings show a limited influence of ABCB1 genotype on lenalidomide treatment efficacy and safety. The results suggest that 1199G>A may be a marker of TTP following Len treatment in standard-risk patients; however, larger studies are needed to validate and clarify the relationship.
- Published
- 2018
- Full Text
- View/download PDF
24. Incidence, characteristics, and outcome of solitary plasmacytoma and plasma cell leukemia. Population-based data from the Swedish Myeloma Register.
- Author
-
Nahi H, Genell A, Wålinder G, Uttervall K, Juliusson G, Karin F, Hansson M, Svensson R, Linder O, Carlson K, Björkstrand B, Kristinsson SY, Mellqvist UH, Blimark C, and Turesson I
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Female, Humans, Incidence, Leukemia, Plasma Cell diagnosis, Leukemia, Plasma Cell mortality, Male, Middle Aged, Patient Outcome Assessment, Plasmacytoma diagnosis, Plasmacytoma mortality, Population Surveillance, Registries, Survival Analysis, Sweden epidemiology, Leukemia, Plasma Cell epidemiology, Plasmacytoma epidemiology
- Abstract
Solitary plasmacytoma (SP) and plasma cell leukemia (PCL) are uncommon (3-6%) types of plasma cell disease. The risk of progression to symptomatic multiple myeloma (MM) is probably important for the outcome of SP. PCL is rare and has a dismal outcome. In this study, we report on incidence and survival in PCL/SP, and progression to MM in SP, using the prospective observational Swedish Multiple Myeloma Register designed to document all newly diagnosed plasma cell diseases in Sweden since 2008. Both solitary bone plasmacytoma (SBP) (n=124) and extramedullary plasmacytoma (EMP) (n=67) have better overall survival (OS) than MM (n=3549). Progression to MM was higher in SBP than in EMP (35% and 7% at 2 years, respectively), but this did not translate into better survival in EMP. In spite of treatment developments, the OS of primary PCL is still dismal (median of 11 months, 0% at 5 years). Hence, there is a great need for diagnostic and treatment guidelines as well as prospective studies addressing the role for alternative treatment options, such as allogeneic stem cell transplantation and monoclonal antibodies in the treatment of PCL., (© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
25. Survival in multiple myeloma patients who develop second malignancies: a population-based cohort study.
- Author
-
Jonsdottir G, Lund SH, Björkholm M, Turesson I, Wahlin A, Mailankody S, Blimark C, Hultcrantz M, Porwit A, Landgren O, and Kristinsson SY
- Subjects
- Aged, Antineoplastic Agents therapeutic use, Cohort Studies, Female, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute pathology, Male, Multiple Myeloma complications, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes etiology, Myelodysplastic Syndromes pathology, Neoplasms, Second Primary drug therapy, Neoplasms, Second Primary etiology, Neoplasms, Second Primary pathology, Risk, Survival Analysis, Sweden, Leukemia, Myeloid, Acute mortality, Multiple Myeloma mortality, Myelodysplastic Syndromes mortality, Neoplasms, Second Primary mortality, Registries
- Published
- 2016
- Full Text
- View/download PDF
26. Multiple myeloma and infections: a population-based study on 9253 multiple myeloma patients.
- Author
-
Blimark C, Holmberg E, Mellqvist UH, Landgren O, Björkholm M, Hultcrantz M, Kjellander C, Turesson I, and Kristinsson SY
- Subjects
- Adult, Aged, Aged, 80 and over, Bacterial Infections etiology, Bacterial Infections mortality, Case-Control Studies, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multiple Myeloma mortality, Multiple Myeloma therapy, Neoplasm Staging, Prognosis, Registries, Survival Rate, Sweden epidemiology, Virus Diseases etiology, Virus Diseases mortality, Bacterial Infections epidemiology, Combined Modality Therapy adverse effects, Multiple Myeloma complications, Virus Diseases epidemiology
- Abstract
Infections are a major cause of morbidity and mortality in patients with multiple myeloma. To estimate the risk of bacterial and viral infections in multiple myeloma patients, we used population-based data from Sweden to identify all multiple myeloma patients (n=9253) diagnosed from 1988 to 2004 with follow up to 2007 and 34,931 matched controls. Cox proportional hazard models were used to estimate the risk of infections. Overall, multiple myeloma patients had a 7-fold (hazard ratio =7.1; 95% confidence interval = 6.8-7.4) risk of developing any infection compared to matched controls. The increased risk of developing a bacterial infection was 7-fold (7.1; 6.8-7.4), and for viral infections 10-fold (10.0; 8.9-11.4). Multiple myeloma patients diagnosed in the more recent calendar periods had significantly higher risk of infections compared to controls (P<0.001). At one year of follow up, infection was the underlying cause in 22% of deaths in multiple myeloma patients. Mortality due to infections remained constant during the study period. Our findings confirm that infections represent a major threat to multiple myeloma patients. The effect on infectious complications due to novel drugs introduced in the treatment of multiple myeloma needs to be established and trials on prophylactic measures are needed., (Copyright© Ferrata Storti Foundation.)
- Published
- 2015
- Full Text
- View/download PDF
27. Improved survival in myeloma patients: starting to close in on the gap between elderly patients and a matched normal population.
- Author
-
Liwing J, Uttervall K, Lund J, Aldrin A, Blimark C, Carlson K, Enestig J, Flogegård M, Forsberg K, Gruber A, Haglöf Kviele H, Johansson P, Lauri B, Mellqvist UH, Swedin A, Svensson M, Näsman P, Alici E, Gahrton G, Aschan J, and Nahi H
- Subjects
- Adult, Age Distribution, Age Factors, Aged, Angiogenesis Inhibitors administration & dosage, Angiogenesis Inhibitors therapeutic use, Antineoplastic Agents administration & dosage, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Boronic Acids administration & dosage, Boronic Acids therapeutic use, Bortezomib, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Humans, Lenalidomide, Male, Middle Aged, Multiple Myeloma drug therapy, Pyrazines administration & dosage, Pyrazines therapeutic use, Registries, Survival Analysis, Sweden epidemiology, Thalidomide administration & dosage, Thalidomide analogs & derivatives, Thalidomide therapeutic use, Treatment Outcome, Multiple Myeloma mortality
- Abstract
The outcome for multiple myeloma patients has improved since the introduction of bortezomib, thalidomide and lenalidomide. However, studies comparing new and conventional treatment include selected patient groups. We investigated consecutive patients (n = 1638) diagnosed in a defined period and compared survival with a gender- and age-matched cohort Swedish population (n = 9 340 682). Median overall survival for non-high-dose treated patients was 2·8 years. The use of bortezomib, thalidomide or lenalidomide in first line therapy predicted a significantly longer overall survival (median 4·9 years) compared to conventional treatment (2·3 years). Among non-high-dose treated patients receiving at least 2 lines with bortezomib, thalidomide or lenalidomide, 69% and 63% have survived at 3 and 5 years as compared to 48% and 22% with conventional drugs and 88% and 79% in the matched cohort populations, respectively. The median overall survival in high-dose treated patients was 6·9 years. Of these patients, 84% survived at 3 years and 70% at 5 years as compared to 98% and 95% in the matched cohort population. Overall survival in the best non-high-dose treated outcome group is closing the gap with the matched cohort. Upfront use of new drugs is clearly better than waiting until later lines of treatment., (© 2013 John Wiley & Sons Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
28. Treatment for high-risk smoldering myeloma.
- Author
-
Kristinsson SY, Holmberg E, and Blimark C
- Subjects
- Female, Humans, Male, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Thalidomide analogs & derivatives
- Published
- 2013
- Full Text
- View/download PDF
29. Bortezomib consolidation after autologous stem cell transplantation in multiple myeloma: a Nordic Myeloma Study Group randomized phase 3 trial.
- Author
-
Mellqvist UH, Gimsing P, Hjertner O, Lenhoff S, Laane E, Remes K, Steingrimsdottir H, Abildgaard N, Ahlberg L, Blimark C, Dahl IM, Forsberg K, Gedde-Dahl T, Gregersen H, Gruber A, Guldbrandsen N, Haukås E, Carlson K, Kvam AK, Nahi H, Lindås R, Andersen NF, Turesson I, Waage A, and Westin J
- Subjects
- Bortezomib, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multiple Myeloma mortality, Multiple Myeloma therapy, Neoplasm Recurrence, Local mortality, Neoplasm Recurrence, Local therapy, Prognosis, Quality of Life, Survival Rate, Transplantation, Autologous, Antineoplastic Agents therapeutic use, Boronic Acids therapeutic use, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy, Pyrazines therapeutic use, Stem Cell Transplantation
- Abstract
The Nordic Myeloma Study Group conducted an open randomized trial to compare bortezomib as consolidation therapy given after high-dose therapy and autologous stem cell transplantation (ASCT) with no consolidation in bortezomib-naive patients with newly diagnosed multiple myeloma. Overall, 370 patients were centrally randomly assigned 3 months after ASCT to receive 20 doses of bortezomib given during 21 weeks or no consolidation. The hypothesis was that consolidation therapy would prolong progression-free survival (PFS). The PFS after randomization was 27 months for the bortezomib group compared with 20 months for the control group (P = .05). Fifty-one of 90 patients in the treatment group compared with 32 of 90 controls improved their response after randomization (P = .007). No difference in overall survival was seen. Fatigue was reported more commonly by the bortezomib-treated patients in self-reported quality-of-life (QOL) questionnaires, whereas no other major differences in QOL were recorded between the groups. Consolidation therapy seemed to be beneficial for patients not achieving at least a very good partial response (VGPR) but not for patients in the ≥ VGPR category at randomization. Consolidation with bortezomib after ASCT in bortezomib-naive patients improves PFS without interfering with QOL. This trial was registered at www.clinicaltrials.gov as #NCT00417911.
- Published
- 2013
- Full Text
- View/download PDF
30. Monoclonal gammopathy of undetermined significance and risk of infections: a population-based study.
- Author
-
Kristinsson SY, Tang M, Pfeiffer RM, Björkholm M, Goldin LR, Blimark C, Mellqvist UH, Wahlin A, Turesson I, and Landgren O
- Subjects
- Adult, Aged, Aged, 80 and over, Bacterial Infections blood, Bacterial Infections complications, Bacterial Infections immunology, Case-Control Studies, Female, Humans, Immunoglobulins blood, Immunoglobulins immunology, Longitudinal Studies, Male, Middle Aged, Monoclonal Gammopathy of Undetermined Significance blood, Monoclonal Gammopathy of Undetermined Significance complications, Monoclonal Gammopathy of Undetermined Significance immunology, Risk, Sweden epidemiology, Virus Diseases blood, Virus Diseases complications, Virus Diseases immunology, Bacterial Infections epidemiology, Monoclonal Gammopathy of Undetermined Significance epidemiology, Virus Diseases epidemiology
- Abstract
No comprehensive evaluation has been made to assess the risk of viral and bacterial infections among patients with monoclonal gammopathy of undetermined significance. Using population-based data from Sweden, we estimated risk of infections among 5,326 monoclonal gammopathy of undetermined significance patients compared to 20,161 matched controls. Patients with monoclonal gammopathy of undetermined significance had a 2-fold increased risk (P<0.05) of developing any infection at 5- and 10-year follow up. More specifically, patients with monoclonal gammopathy of undetermined significance had an increased risk (P<0.05) of bacterial (pneumonia, osteomyelitis, septicemia, pyelonephritis, cellulitis, endocarditis, and meningitis), and viral (influenza and herpes zoster) infections. Patients with monoclonal gammopathy of undetermined significance with M-protein concentrations over 2.5 g/dL at diagnosis had highest risks of infections. However, the risk was also increased (P<0.05) among those with concentrations below 0.5 g/dL. Patients with monoclonal gammopathy of undetermined significance who developed infections had no excess risk of developing multiple myeloma, Waldenström macroglobulinemia or related malignancy. Our findings provide novel insights into the mechanisms behind infections in patients with plasma cell dyscrasias, and may have clinical implications.
- Published
- 2012
- Full Text
- View/download PDF
31. Personal and family history of immune-related conditions increase the risk of plasma cell disorders: a population-based study.
- Author
-
Lindqvist EK, Goldin LR, Landgren O, Blimark C, Mellqvist UH, Turesson I, Wahlin A, Björkholm M, and Kristinsson SY
- Subjects
- Adult, Aged, Aged, 80 and over, Autoimmune Diseases physiopathology, Case-Control Studies, Cohort Studies, Disease Susceptibility immunology, Family Health, Female, Humans, Infections physiopathology, Inflammation physiopathology, Male, Middle Aged, Monoclonal Gammopathy of Undetermined Significance epidemiology, Monoclonal Gammopathy of Undetermined Significance etiology, Multiple Myeloma epidemiology, Multiple Myeloma etiology, Multiple Myeloma immunology, Registries, Risk, Sweden epidemiology, Young Adult, Autoimmune Diseases immunology, Immune System Diseases physiopathology, Monoclonal Gammopathy of Undetermined Significance immunology
- Abstract
The associations between immune-related conditions and multiple myeloma (MM) and monoclonal gammopathy of undetermined significance (MGUS) have previously been investigated with inconsistent results. In a large population-based study, we identified 19 112 patients with MM, 5403 patients with MGUS, 96 617 matched control subjects, and 262 931 first-degree relatives. We calculated odds ratios (ORs) and 95% confidence intervals (CIs) for the association of MM and MGUS with immune-related conditions by use of logistic regression. A personal history of all infections combined was associated with a significantly increased risk of MM (OR = 1.2; 95% CI, 1.1-1.3), and a personal history of all conditions in the categories infections (OR = 1.6; 95% CI, 1.5-1.7), inflammatory conditions (OR = 1.4; 95% CI, 1.2-1.5), and autoimmune diseases (OR = 2.1; 95% CI, 1.9-2.4) was associated with a significantly increased risk of MGUS. Several specific immune-related conditions elevated the risk of MM and/or MGUS. A family history of autoimmune disease was associated with a significantly increased risk of MGUS (OR = 1.1; 95% CI, 1.00-1.2), but not MM. Our findings suggest that immune-related conditions and/or their treatment are of importance in the etiology of MGUS and possibly MM. The association of both personal and family history of autoimmune disease with MGUS indicates the potential for shared susceptibility for these conditions.
- Published
- 2011
- Full Text
- View/download PDF
32. Melphalan 100 mg/m2 with stem cell support as first relapse treatment is safe and effective for myeloma patients with long remission after autologous stem cell transplantation.
- Author
-
Blimark C, Veskovski L, Westin J, Rödjer S, Brune M, Hjorth M, Holmberg E, Andersson PO, and Mellqvist UH
- Subjects
- Adult, Aged, Antineoplastic Agents, Alkylating adverse effects, Combined Modality Therapy, Disease-Free Survival, Female, Humans, Male, Melphalan adverse effects, Middle Aged, Recurrence, Retrospective Studies, Time Factors, Transplantation, Autologous, Treatment Outcome, Antineoplastic Agents, Alkylating administration & dosage, Melphalan administration & dosage, Multiple Myeloma drug therapy, Multiple Myeloma therapy, Stem Cell Transplantation
- Abstract
Introduction: Today, a number of therapeutic options are available as the patient with myeloma relapses from initial treatment with high-dose melphalan and autologous stem cell transplantation (ASCT). For patients who experience a durable response to primary ASCT, retreatment with high-dose melphalan is recommended by many current guidelines. Yet, toxicity is an important aspect in the choice of relapse treatment, and a second ASCT in this setting could be associated with enhanced toxicity. As the goal for the treatment for relapsed myeloma should be disease control while maintaining quality of life, lower doses of melphalan might be preferable., Methods and Objectives: In this retrospective study, we account for the outcome of 66 patients with myeloma in first systemic relapse after ASCT, who were treated with intermediate-dose melphalan, 100 mg/m2, and stem cell support (MEL 100). The aim was to evaluate this treatment in relation to prior response duration after initial ASCT and with respect to response rate, toxicity and survival., Results: The overall response rate was 62%. There was limited, mostly haematological, toxicity, and no treatment-related mortality was observed. The median progression-free survival (PFS) was 8.5 months, and the median overall survival was 24 months. Patients with time to progression of 34 months or more (n=17; ≥75th percentile) after initial ASCT had a median PFS of 12.5 months after MEL 100., Conclusion: For patients with a long-lasting response after ASCT, MEL 100 could be a therapeutic option with low toxicity and with efficacy comparable to newer immunomodulatory drugs., (© 2011 John Wiley & Sons A/S.)
- Published
- 2011
- Full Text
- View/download PDF
33. Monoclonal gammopathy of undetermined significance and risk of skeletal fractures: a population-based study.
- Author
-
Kristinsson SY, Tang M, Pfeiffer RM, Björkholm M, Blimark C, Mellqvist UH, Wahlin A, Turesson I, and Landgren O
- Subjects
- Adult, Aged, Aged, 80 and over, Case-Control Studies, Confidence Intervals, Female, Follow-Up Studies, Humans, Immunoglobulin Isotypes, Male, Middle Aged, Monoclonal Gammopathy of Undetermined Significance immunology, Multiple Myeloma complications, Proportional Hazards Models, Risk Factors, Sweden, Young Adult, Fractures, Bone etiology, Monoclonal Gammopathy of Undetermined Significance complications
- Abstract
Patients with multiple myeloma (MM) have an increased risk of fractures. On the basis of small numbers, patients with monoclonal gammopathy of undetermined significance (MGUS) have been reported to have an increased fracture risk. Using population-based data from Sweden, we assessed the risks of fractures in 5326 MGUS patients diagnosed from 1958 to 2006, compared with 20 161 matched controls. MGUS patients had an increased risk of any fracture at 5 (hazard ratio [HR] = 1.74; 95% confidence interval [CI], 1.58-1.92) and 10 (HR = 1.61; 95% CI, 1.49-1.74) years. The risk was significantly higher for axial (skull, vertebral/pelvis, and sternum/costae) compared with distal (arm and leg) fractures (P < .001). On the basis of 10 years of follow-up, there was an increased risk of vertebral/pelvic (HR = 2.37; 95% CI, 2.02-2.78), sternal/costae (HR = 1.93; 95% CI, 1.5-2.48), arm (HR = 1.23; 95% CI, 1.06-1.43), leg (HR = 1.40; 95% CI, 1.26-1.56), and other/multiple fractures (HR = 4.25; 95% CI, 3.29-5.51). Risks for fractures did not differ by isotype or M protein concentration at diagnosis. MGUS patients with (versus without) fractures had no excess risk of MM or Waldenström macroglobulinemia. Our results suggest that bone alterations are present in early myelomagenesis. Our findings may have implications for the development of better prophylaxis for bone disease in MGUS, and they provide novel clues on pathogenesis of MM bone disease.
- Published
- 2010
- Full Text
- View/download PDF
34. Arterial and venous thrombosis in monoclonal gammopathy of undetermined significance and multiple myeloma: a population-based study.
- Author
-
Kristinsson SY, Pfeiffer RM, Björkholm M, Goldin LR, Schulman S, Blimark C, Mellqvist UH, Wahlin A, Turesson I, and Landgren O
- Subjects
- Adult, Aged, Aged, 80 and over, Arteries, Databases, Factual, Female, Humans, Immunoglobulin A blood, Immunoglobulin G blood, Male, Middle Aged, Multiple Myeloma blood, Paraproteinemias blood, Predictive Value of Tests, Proportional Hazards Models, Registries, Risk Factors, Survival Analysis, Sweden epidemiology, Veins, Venous Thrombosis blood, Venous Thrombosis prevention & control, Waldenstrom Macroglobulinemia blood, Waldenstrom Macroglobulinemia mortality, Multiple Myeloma mortality, Paraproteinemias mortality, Venous Thrombosis mortality
- Abstract
Patients with multiple myeloma (MM) have an increased risk of venous thrombosis. Interestingly, excess risk of venous thromboembolism has been observed among patients with monoclonal gammopathy of undetermined significance (MGUS). Using population-based data from Sweden, we assessed the risks of venous and arterial thrombosis in 18,627 MM and 5326 MGUS patients diagnosed from 1958 to 2006, compared with 70,991 and 20,161 matched controls, respectively. At 1, 5, and 10 years after MM diagnosis, there was an increased risk of venous thrombosis: hazard ratios (95% confidence intervals) were 7.5 (6.4-8.9), 4.6 (4.1-5.1), and 4.1 (3.8-4.5), respectively. The corresponding results for arterial thrombosis were 1.9 (1.8-2.1), 1.5 (1.4-1.6), and 1.5 (1.4-1.5). At 1, 5, and 10 years after MGUS diagnosis, hazard ratios were 3.4 (2.5-4.6), 2.1 (1.7-2.5), and 2.1 (1.8-2.4) for venous thrombosis. The corresponding risks for arterial thrombosis were 1.7 (1.5-1.9), 1.3 (1.2-1.4), and 1.3 (1.3-1.4). IgG/IgA (but not IgM) MGUS patients had increased risks for venous and arterial thrombosis. Risks for thrombosis did not vary by M-protein concentration (> 10.0 g/L or < 10.0 g/L) at diagnosis. MGUS patients with (vs without) thrombosis had no excess risk of MM or Waldenström macroglobulinemia. Our findings are of relevance for future studies and for improvement of thrombosis prophylaxis strategies.
- Published
- 2010
- Full Text
- View/download PDF
35. Patterns of hematologic malignancies and solid tumors among 37,838 first-degree relatives of 13,896 patients with multiple myeloma in Sweden.
- Author
-
Kristinsson SY, Björkholm M, Goldin LR, Blimark C, Mellqvist UH, Wahlin A, Turesson I, and Landgren O
- Subjects
- Adult, Aged, Aged, 80 and over, Case-Control Studies, Female, Hematologic Neoplasms epidemiology, Humans, Male, Middle Aged, Neoplasms epidemiology, Paraproteinemias epidemiology, Paraproteinemias genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Sweden epidemiology, Urinary Bladder Neoplasms genetics, Family, Hematologic Neoplasms genetics, Multiple Myeloma genetics, Neoplasms genetics
- Abstract
There are emerging data to suggest a role for genetic factors in the pathogenesis of multiple myeloma (MM). Based on small numbers, certain solid tumors have been reported to occur more frequently among blood relatives of patients with MM. Using population-based data, we assessed risks for hematologic malignancies, monoclonal gammopathy of undetermined significance (MGUS), and solid tumors among first-degree relatives of patients with MM. We included 13,896 patients with MM and 54,365 matched controls. Also we identified first-degree relatives of patients with MM (n = 37,838) and controls (n = 151,068). Using a marginal survival model, we estimated relative risks (RRs) and 95% confidence intervals (CIs) for hematologic and solid tumors among family members of patients with MM and controls as measures of familial aggregation. Compared with relatives of controls, relatives of patients with MM had an increased risk of developing MM (RR = 2.1; 95% CI 1.6-2.9), MGUS (2.1; 1.5-3.1), acute lymphoblastic leukemia (ALL) (2.1; 1.0-4.2), any solid tumor (1.1; 1.0-1.1) and bladder cancer (1.3; 1.0-1.5). No significantly increased risk was found for other hematologic or solid malignancies. Our findings support a role for a shared susceptibility (genetic, environmental or both) that predisposes to MM, MGUS, ALL and bladder cancer., ((c) 2009 UICC.)
- Published
- 2009
- Full Text
- View/download PDF
36. Risk of plasma cell and lymphoproliferative disorders among 14621 first-degree relatives of 4458 patients with monoclonal gammopathy of undetermined significance in Sweden.
- Author
-
Landgren O, Kristinsson SY, Goldin LR, Caporaso NE, Blimark C, Mellqvist UH, Wahlin A, Bjorkholm M, and Turesson I
- Subjects
- Adult, Aged, Aged, 80 and over, Case-Control Studies, Female, Humans, Lymphoproliferative Disorders epidemiology, Male, Middle Aged, Neoplasms, Plasma Cell epidemiology, Risk Factors, Sweden epidemiology, Family, Lymphoproliferative Disorders etiology, Neoplasms, Plasma Cell etiology, Paraproteinemias epidemiology
- Abstract
Familial clustering of the precursor condition, monoclonal gammopathy of undetermined significance (MGUS) has been observed in case reports and in smaller studies. Using population-based data from Sweden, we identified 4458 MGUS patients, 17505 population-based controls, and first-degree relatives of patients (n = 14621) and controls (n = 58387) with the aim to assess risk of MGUS and lymphoproliferative malignancies among first-degree relatives of MGUS patients. Compared with relatives of controls, relatives of MGUS patients had increased risk of MGUS (relative risk [RR] = 2.8; 1.4-5.6), multiple myeloma (MM; RR = 2.9; 1.9-4.3), lymphoplasmacytic lymphoma/Waldenström macroglobulinemia (LPL/WM; RR = 4.0; 1.5-11), and chronic lymphocytic leukemia (CLL; RR = 2.0; 1.2-2.3). Relatives of patients with IgG/IgA MGUS had a 4.0-fold (1.7-9.2), 2.9-fold (1.7-4.9), and 20-fold (2.3-170) elevated risk of developing MGUS, MM, and LPL/WM, respectively. Relatives of IgM MGUS patients had 5.0-fold (1.1-23) increased CLL risk and nonsignificant excess MM and LPL/WM risks. The results were very similar when we assessed risk by type of first-degree relative, age at MGUS (above/below 65 years), or sex. Risk of non-Hodgkin lymphoma or Hodgkin lymphoma was not increased among MGUS relatives. Among first-degree relatives of a nationwide MGUS cohort, we found elevated risks of MGUS, MM, LPL/WM, and CLL, supporting a role for germline susceptibility genes, shared environmental influences, or an interaction between both.
- Published
- 2009
- Full Text
- View/download PDF
37. Percutaneous vertebroplasty at C2: case report of a patient with multiple myeloma and a literature review.
- Author
-
Rodriguez-Catarino M, Blimark C, Willén J, Mellqvist UH, and Rödjer S
- Subjects
- Axis, Cervical Vertebra diagnostic imaging, Axis, Cervical Vertebra pathology, Bone Cements therapeutic use, Female, Fluoroscopy, Humans, Magnetic Resonance Imaging, Melphalan therapeutic use, Middle Aged, Monitoring, Intraoperative, Multiple Myeloma drug therapy, Multiple Myeloma radiotherapy, Myeloablative Agonists therapeutic use, Neck Pain diagnostic imaging, Neck Pain etiology, Neck Pain pathology, Radiotherapy, Spinal Fractures diagnosis, Spinal Neoplasms drug therapy, Spinal Neoplasms radiotherapy, Tomography, X-Ray Computed, Treatment Outcome, Vertebroplasty instrumentation, Axis, Cervical Vertebra surgery, Multiple Myeloma complications, Spinal Fractures etiology, Spinal Fractures surgery, Spinal Neoplasms complications, Vertebroplasty methods
- Abstract
Percutaneous vertebroplasty (PVP) of the axis is a challenging procedure which may be performed by a percutaneous or a transoral approach. There are few reports of PVP at the C2 level. We report a case of unstable C2 fracture treated with the percutaneous approach. The fracture was the first manifestation of multiple myeloma in a previously healthy 47-year-old woman. After local radiotherapy and chemotherapy, the fracture was still unstable and the patient had been continuously wearing a stiff cervical collar for 9 months. Complication-free PVP resulted in pain relief and stabilization and use of the cervical collar could be discontinued. At 18 months follow-up the patient remained free from pain, the fracture was stable and she had returned to work. The purpose of this article is to present the technical facts and to highlight the benefits and potential complications of the procedure. The technical characteristics of the procedure, the indication and results of the present case are discussed together with previously reported cases of PVP treatment at C2.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.