165 results on '"Schjesvold F"'
Search Results
52. PB2116 EVOLUTION OF MULTIPLE MYELOMA MANAGEMENT IN NORWAY: EXPERT OPINION 2013–2018
- Author
-
Schjesvold, F. H., primary
- Published
- 2019
- Full Text
- View/download PDF
53. Pembrolizumab plus lenalidomide and dexamethasone for patients with treatment-naive multiple myeloma (KEYNOTE-185): a randomised, open-label, phase 3 trial.
- Author
-
Usmani S.Z., Oriol A., Karlin L., Cavo M., Rifkin R.M., Hayek F., Kirschner E., Bharany N., Overton L., Mannem S., Harroff A., Jain S., Roque T., McIntyre K., Yasencha C.K., Houck W., Schjesvold F., Yimer H.A., LeBlanc R., Takezako N., McCroskey R.D., Lim A.B.M., Suzuki K., Kosugi H., Grigoriadis G., Avivi I., Facon T., Jagannath S., Lonial S., Ghori R.U., Farooqui M.Z.H., Marinello P., San-Miguel J., Lim A., Walker T., Nicol A., Reece D., Elemary M., Boudreault Pedneault J.S., Attal M., Weisel K., Engelhardt M., Mackensen A., Quinn J., Cohen A., Magen-Nativ H., Benyamini N., Larocca A., Matsumoto M., Iida S., Ishikawa T., Kondo Y., Sunami K., Ando K., Teshima T., Chou T., Iwasaki H., Miki H., Matsumura I., Onishi Y., Izutsu K., Kizaki M., George A., Blacklock H., Simpson D., Waage A., Samoilova O., Nikitin E., Chagorova T., McDonald A., Patel M., Oriol Rocafiguera A., San Miguel Izquierdo J., Mateos M., Streetly M., Forsyth P., Jackson G., Jenkins S., Rifkin R., Yimer H., McCroskey R., Martincic D., Tarantolo S., Larson S., Faroun Y., Vaughn J., Baz R., Saylors G., Neppalli A., Raptis A., Fung H., Janosky M., Stevens D., Coleman M., Costa D., Cross S., Fanning S., Berges D.F., Harris T., Zackon I., Atanackovic D., Lee K., Oliff I., Lee W., Bensinger W., Lutzky J., Baron A., Usmani S.Z., Oriol A., Karlin L., Cavo M., Rifkin R.M., Hayek F., Kirschner E., Bharany N., Overton L., Mannem S., Harroff A., Jain S., Roque T., McIntyre K., Yasencha C.K., Houck W., Schjesvold F., Yimer H.A., LeBlanc R., Takezako N., McCroskey R.D., Lim A.B.M., Suzuki K., Kosugi H., Grigoriadis G., Avivi I., Facon T., Jagannath S., Lonial S., Ghori R.U., Farooqui M.Z.H., Marinello P., San-Miguel J., Lim A., Walker T., Nicol A., Reece D., Elemary M., Boudreault Pedneault J.S., Attal M., Weisel K., Engelhardt M., Mackensen A., Quinn J., Cohen A., Magen-Nativ H., Benyamini N., Larocca A., Matsumoto M., Iida S., Ishikawa T., Kondo Y., Sunami K., Ando K., Teshima T., Chou T., Iwasaki H., Miki H., Matsumura I., Onishi Y., Izutsu K., Kizaki M., George A., Blacklock H., Simpson D., Waage A., Samoilova O., Nikitin E., Chagorova T., McDonald A., Patel M., Oriol Rocafiguera A., San Miguel Izquierdo J., Mateos M., Streetly M., Forsyth P., Jackson G., Jenkins S., Rifkin R., Yimer H., McCroskey R., Martincic D., Tarantolo S., Larson S., Faroun Y., Vaughn J., Baz R., Saylors G., Neppalli A., Raptis A., Fung H., Janosky M., Stevens D., Coleman M., Costa D., Cross S., Fanning S., Berges D.F., Harris T., Zackon I., Atanackovic D., Lee K., Oliff I., Lee W., Bensinger W., Lutzky J., and Baron A.
- Abstract
Background: Lenalidomide and dexamethasone has been a standard of care in transplant-ineligible patients with newly diagnosed multiple myeloma. The addition of a third drug to the combination is likely to improve treatment efficacy. KEYNOTE-185 assessed the efficacy and safety of lenalidomide and dexamethasone with and without pembrolizumab in patients with previously untreated multiple myeloma. Here, we present the results of an unplanned interim analysis done to assess the benefit-risk of the combination at the request of the US Food and Drug Administration (FDA). Method(s): KEYNOTE-185 was a randomised, open-label, phase 3 trial done at 95 medical centres across 15 countries (Australia, Canada, France, Germany, Ireland, Israel, Italy, Japan, New Zealand, Norway, Russia, South Africa, Spain, UK, and USA). Transplantation-ineligible patients aged 18 years and older with newly diagnosed multiple myeloma, Eastern Cooperative Oncology Group performance status of 0 or 1, and who were treatment naive were enrolled, and randomly assigned 1:1 to receive either pembrolizumab plus lenalidomide and dexamethasone or lenalidomide and dexamethasone alone using an interactive voice or integrated web response system. Patients received oral lenalidomide 25 mg on days 1-21 and oral dexamethasone 40 mg on days 1, 8, 15, and 22 of repeated 28-day cycles, with or without intravenous pembrolizumab 200 mg every 3 weeks. The primary endpoint was progression-free survival, which was investigator-assessed because of early trial termination. Efficacy was analysed in all randomly assigned patients and safety was analysed in all patients who received at least one dose of study drug. This trial is registered at ClinicalTrials.gov, number NCT02579863, and it is closed for accrual. Finding(s): Between Jan 7, 2016, and June 9, 2017, 301 patients were randomly assigned to the pembrolizumab plus lenalidomide and dexamethasone group (n=151) or the lenalidomide and dexamethasone group (n=150). On Jul
- Published
- 2019
54. OPTIMISMM: PHASE 3 STUDY OF POMALIDOMIDE, BORTEZOMIB, AND LOW DOSE OF DEXAMENTASONE VS. BORTEZOMIB AND LOW DOSE OF DEXAMETHASONE IN PATIENTS EXPOSED TO LENALIDOMIDE AFTER FIRST COLLECTION OF MULTIPLE MYELOMA
- Author
-
Oriol, A Richardson, P Beksac, M Liberati, AM Galli, M Schjesvold, F Lindsay, J Weisel, K White, D Facon, T others
- Subjects
Health Sciences ,Επιστήμες Υγείας - Published
- 2018
55. International myeloma working group recommendations for the diagnosis and management of myeloma-related renal impairment
- Author
-
Dimopoulos, M.A. Sonneveld, P. Leung, N. Merlini, G. Ludwig, H. Kastritis, E. Goldschmidt, H. Joshua, D. Orlowski, R.Z. Powles, R. Vesole, D.H. Garderet, L. Einsele, H. Palumbo, A. Cavo, M. Richardson, P.G. Moreau, P. Miguel, J.S. Vincent Rajkumar, S. Durie, B.G.M. Terpos, E. Abildgaard, N. Abonour, R. Alsina, M. Anderson, K.C. Attal, M. Avet-Loiseau, H. Badros, A. Bahlis, N.J. Barlogie, B. Bataille, R. Beksaç, M. Belch, A. Ben-Yehuda, D. Bensinger, B. Leif Bergsagel, P. Bhutani, M. Bird, J. Bladé, J. Broijl, A. Boccadoro, M. Caers, J. Chanan-Khan, A. Chari, A. Chen, W.M. Chesi, M. Anthony Child, J. Chim, C.S. Chng, W.-J. Comenzo, R. Cook, G. Crowley, J. Crusoe, E. Dalton, W. Lee Moffitt, H. Davies, F. de la Rubia, J. de Souza, C. Delforge, M. Dhodapkar, M. Dispenzieri, A. Drach, J. Drake, M. Du, J. Dytfeld, D. Facon, T. Fantl, D. Fermand, J.-P. Fernández de Larrea, C. Fonseca, R. Gahrton, G. Garćia-Sanz, R. Gasparetto, C. Gertz, M. Ghobrial, I. Gibson, J. Gimsing, P. Giralt, S. Gu, J. Hajek, R. Hardan, I. Hari, P. Hata, H. Hattori, Y. Heffner, T. Hillengass, J. Ho, J. Hoering, A. Hoffman, J.E. Hou, J. Huang, J. Hungria, V. Ida, S. Jagannath, S. Jakubowiak, A.J. Johnsen, H.E. Jurczyszyn, A. Kaiser, M. Kaufman, J. Kawano, M. Korde, N. Kovacs, E. Krishnan, A. Kristinsson, S. Kröger, N. Kumar, S. Kyle, R.A. Kyriacou, C. Lacy, M. Lahuerta, J.J. Landgren, O. Larocca, A. Laubach, J. da Costa, F.L. Lee, J.-H. Leiba, M. Leleu, X. Lentzsch, S. Lokhorst, H. Lonial, S. Lu, J. Mahindra, A. Maiolino, A. Manasanch, E.E. Mark, T. Mateos, M.-V. Mazumder, A. McCarthy, P. Mehta, J. Mellqvist, U.-H. Mikhael, J. Morgan, G. Munshi, N. Nahi, H. Nawarawong, W. Niesvizky, R. Nouel, A. Novis, Y. Ocio, E. O'Dwyer, M. O'Gorman, P. Orfao, A. Otero, P.R. Paiva, B. Pavlovsky, S. Pilarski, L. Pratt, G. Qui, L. Raje, N. Reece, D. Reiman, A. Remaggi, G. Richter, J. Serra, E.R. Morales, A.R. Romeril, K.R. Roodman, D. Rosiñol, L. Rossi, A. Roussel, M. Russell, S. Schjesvold, F. Schots, R. Sevcikova, S. Sezer, O. Shah, J.J. Shimizu, K. Shustik, C. Siegel, D. Singhal, S. Spencer, A. Stadtmauer, E. Stewart, K. Tan, D. Terragna, C. Tosi, P. Tricot, G. Turesson, I. Usmani, S. Van Camp, B. Van de Donk, N. Van Ness, B. Van Riet, I. Broek, I.V. Vanderkerken, K. Vescio, R. Vij, R. Voorhees, P. Waage, A. Wang, M. Weber, D. Weiss, B.M. Westin, J. Wheatley, K. Zamagni, E. Zonder, J. Zweegman, S.
- Abstract
Purpose: The aim of the International Myeloma Working Group was to develop practical recommendations for the diagnosis and management of multiple myeloma–related renal impairment (RI). Methods: Recommendations were based on published data through December 2015, and were developed using the system developed by the Grading of Recommendation, Assessment, Development, and Evaluation Working Group. Recommendations: All patients with myeloma at diagnosis and at disease assessment should have serum creatinine, estimated glomerular filtration rate, and electrolytes measurements as well as free light chain, if available, and urine electrophoresis of a sample from a 24-hour urine collection (grade A). The Chronic Kidney Disease Epidemiology Collaboration, preferably, or the Modification of Diet in Renal Disease formula should be used for the evaluation of estimated glomerular filtration rate in patients with stabilized serum creatinine (grade A). International Myeloma Working Group criteria for renal reversibility should be used (grade B). For the management of RI in patients with multiple myeloma, high fluid intake is indicated along with antimyeloma therapy (grade B). The use of high-cutoff hemodialysis membranes in combination with antimyeloma therapy can be considered (grade B). Bortezomib-based regimens remain the cornerstone of the management of myeloma-related RI (grade A). High-dose dexamethasone should be administered at least for the first month of therapy (grade B). Thalidomide is effective in patients with myeloma with RI, and no dose modifications are needed (grade B). Lenalidomide is effective and safe, mainly in patients with mild to moderate RI (grade B); for patients with severe RI or on dialysis, lena-lidomide should be given with close monitoring for hematologic toxicity (grade B) with dose reduction as needed. High-dose therapy with autologous stem cell transplantation (with melphalan 100 mg/m2 to 140 mg/m2) is feasible in patients with RI (grade C). Carfilzomib can be safely administered to patients with creatinine clearance > 15 mL/min, whereas ixazomib in combination with lenalidomide and dex-amethasone can be safely administered to patients with creatinine clearance > 30 mL/min (grade A). © 2016 by American Society of Clinical Oncology.
- Published
- 2016
56. KEYNOTE-185: A randomized, open-label phase 3 study of pembrolizumab in combination with lenalidomide and low-dose dexamethasone in newly diagnosed and treatment-naive multiple myeloma (MM)
- Author
-
Lonial, S., primary, Ribeiro de Oliveira, M., additional, Yimer, H., additional, Mateos, M.V., additional, Rifkin, R., additional, Schjesvold, F., additional, San-Miguel, J., additional, Ghori, R., additional, Marinello, P., additional, and Jagannath, S., additional
- Published
- 2016
- Full Text
- View/download PDF
57. Oral Apixaban for the Treatment of Acute Venous Thromboembolism
- Author
-
Agnelli, G, Buller, H, Cohen, A, Gallus, A, Raskob, G, Weitz, J, Prins, M, Brandjes, D, Kolbach, D, Limburg, M, Mac Gillavry, M, Otten, Jm, Peters, R, Roos, Y, Segers, A, Slagboom, T, Bounameaux, H, Hirsh, J, Samama, Mm, Wedel, H, Curto, M, Johnson, M, Masiukiewicz, U, Pak, R, Porcari, A, Sanders, P, Sisson, M, Sullivan, B, Thompson, J, Auerbach, J, Cesario, L, Crawford, J, Gordon, M, Noble, M, Pennington, A, Reinhold, P, Simmons, M, Urwin, K, Ceresetto, J, Mcrae, S, Pabinger, I, Pereira, Ah, Spencer, F, Wang, C, Zhang, J, Gorican, K, Husted, Se, Mottier, D, Harenberg, J, Vértes, A, Pinjala, R, Zeltser, D, Prandoni, Paolo, Sandset, M, Torbicki, A, Fijalkowska, A, Alvares, Jp, Kirienko, A, Shvarts, Y, Sala, La, Jacobson, B, Gudz, I, Ortel, T, Spyropoulos, A, Beyer Westendorf, J, Sipos, G, Bredikhin, R, Della Siega, A, Klinke, W, Lawall, H, Zwettler, U, Prasol, V, Cannon, K, Vasylyuk, S, Jin, B, Prandoni, P, Desai, S, Zaichuk, A, Katelnitskiy, I, De Pellegrin, A, Santonastaso, M, Skupyy, O, Pesant, Y, Shvalb, P, Spacek, R, Visonà, A, Alvarez Sala, L, Borja, V, Noori, E, Sereg, M, Braester, A, Falvo, N, Vöhringer, H, Laperna, L, Oliven, A, Skalicka, L, Bolster, D, Haidar, A, Schellong, S, Smith, S, Sergeev, O, Pullman, J, Torp Pedersen, C, Zimlichman, R, Elias, M, Fourie, N, Pernod, G, Panchenko, E, Pendleton, R, van Nieuwenhuizen, E, Vinereanu, D, Becattini, C, Manina, G, Leduc, J, Dunaj, M, Frost, L, Gavish, D, Jakobsen, T, Lishner, M, Morales, L, Chochola, J, Gubka, O, Holaj, R, Hussein, O, Katona, A, Sergeeva, E, Bova, C, Cepeda, J, Cohen, K, Sobkowicz, B, Grzelakowski, P, Husted, S, Lupkovics, G, Dedek, V, Liu, C, Puskas, A, Ritchie, B, Ambrosio, G, Parisi, R, Heuer, H, Livneh, A, Podpera, I, Stanbro, M, Caraco, Y, Fulmer, J, Ghirarduzzi, A, Schmidt Lucke, J, Bergmann, J, Cizek, V, Leyden, M, Stein, R, Abramov, I, Chong, B, Colan, D, Jindal, R, Liu, S, Pereira, A, Porreca, E, Salem, H, Welker, J, Yusen, R, Dhar, A, Podczeck Schweighofer, A, Shtutin, O, Vital Durand, D, Balaji, V, Correa, J, Kline, J, Runyon, M, Laszlo, Z, Martelet, M, Parakh, R, Sandset, Pm, Schmidt, J, Yeo, E, Bhagavan, N, Bura Riviere, A, Ferrer, J, Lacroix, P, Lewczuk, J, Pilger, E, Sokurenko, G, Yu, H, Nikulnikov, P, Pabinger Fasching, I, Sanchez Diaz, C, Schuller, D, Suresh, K, Lobo, S, Lyons, R, Marschang, P, Palla, A, Schulman, S, Spyropoulous, A, Fraiz, J, Gerasymov, V, Lerner, R, Llamas Esperón, G, Manenti, E, Masson, J, Moreira, R, Poy, C, Rodoman, G, Bruckner, I, Gurghean, A, Carrier, M, Freire, A, Gan, E, Gibson, K, Herold, M, Hudcovic, M, Kamath, G, Koslow, A, Meneveau, N, Roos, J, Zahn, R, Balanda, J, Bratsch, H, Dolan, S, Gould, T, Hirschl, M, Hoffmann, U, Kaatz, S, Shah, V, Kadapatti, K, Kræmmer Nielsen, H, Lahav, M, Natarajan, S, Tuxen, C, Tveit, A, Alves, C, Formiga, A, Brudevold, R, Cardozo, M, Lorch, D, Marais, H, Mismetti, P, Panico, M, Pop, C, Quist Paulsen, P, Stevens, D, Tarleton, G, Yoshida, W, Cox, M, Crispin, P, Czekalski, P, Ebrahim, I, Game, M, Ghanima, W, Harrington, D, Jackson, D, Lee, A, Matoska, P, Meade, A, Camargo, Ac, Nishinari, K, Sanchez Llamas, F, Tosetto, A, Vejby Christensen, H, Basson, M, Blombery, P, Fu, G, Jha, V, Keltai, K, Le Jeunne, C, Lodigiani, C, Ma, Y, Nagy, A, Neumeister, A, Shotan, A, Wong, T, Ying, K, Anderson, S, Brenner, B, Carnovali, M, Cerana, S, Cunha, C, Diaz Castañon, J, Graham, M, Kirenko, A, Palareti, G, Rodriguez Cintron, W, Nathanson, A, Rosenthal, S, Sanders, D, Scheinberg, P, Schjesvold, F, Torp, R, van Zyl, L, Venher, I, Xia, G, Brockmyre, A, Chen, Z, Hakki, S, Hanefield, C, Mügge, A, Janczak, D, Karpovych, D, Lancaster, G, Lavigne, C, Lugassy, G, Melaniuk, M, Moran, J, Oliver, M, Schattner, A, Staroverov, I, Timi, J, Vöhringer, F, von Bilderling, P, Warr, T, White, R, Wronski, J, Wu, C, Almeida, C, Blum, A, Bono, J, Durán, M, Erzinger, F, Fu, W, Jagadesan, R, Jurecka, W, Korban, E, Nguyen, D, Raval, M, Willms, D, Zevin, S, Zhu, H, Abdullah, I, Achkar, A, Albuquerque, L, Ali, M, Bai, C, Bloomfield, D, Chen, J, Fajardo Campos, P, Garcia Bragado, F, Kobza, I, Lindhoff Last, E, Lourenço, A, Marchena Yglesias, P, Marshall, P, Siegel, M, Mikhailova, O, Oliva, M, Pottier, P, Pruszczyk, P, Sauer, M, Baloira, A, Cromer, M, D'Angelo, A, Faucher, J, Gutowski, P, Hong, S, Lissauer, M, Lopes, A, Lopes, R, Maholtz, M, Mesquita, E, Miekus, P, Mohan, B, Ng, H, Peterson, M, Piovella, F, Siragusa, S, Srinivas, R, Tiberio, G, Van Bellen, B, Arutyunov, G, Assi, N, Baker, R, Blanc, F, Curnow, J, Fu, C, Gonzalez Porras, J, Guijarro Merino, R, Gunasingam, S, Gupta, P, Laule, M, Liu, Z, Luber, J, Serifilippi, G, Paulson, R, Shevela, A, Simonneau, G, Siu, D, Sosa Liprandi, M, Takács, J, Tay, J, Vora, K, Witkiewicz, W, Zhao, L, Aquilanti, S, Dabbagh, O, Dellas, C, Denaro, C, Doshi, A, Flippo, G, Giumelli, C, Gomez Cerezo, J, Han, D, Harris, L, Hofmann L., Jr, Kamerkar, D, Kaminski, L, Kazimir, M, Kloczko, J, Ko, Y, Koura, F, Lavender, R, Maly, J, Margolis, B, Mos, L, Sanchez Escalante, L, Solvang, A, Soroka, V, Szopinski, P, Thawani, H, Vickars, L, Yip, G, Zangroniz, P., Internal and Cardiovascular Medicine - Stroke Unit (PERUGIA - ICM-SU), Università degli Studi di Perugia (UNIPG), Department of Vascular Medicine (DVM - AMC), Academic Medical Center - Academisch Medisch Centrum [Amsterdam] (AMC), University of Amsterdam [Amsterdam] (UvA)-University of Amsterdam [Amsterdam] (UvA), King's College Hospital (KCH), Department of Haematology (ADELAIDE - Dep Haemato), Flinders Medical Centre-Flinders University, Health Sciences Center (OKLAHOMA - HSC), University of Oklahoma (OU), Thrombosis and Atherosclerosis Research Institute (TARI), McMaster University [Hamilton, Ontario], Centre d'Investigation Clinique (CIC - Brest), Université de Brest (UBO)-Institut National de la Santé et de la Recherche Médicale (INSERM), Groupe d'Etude de la Thrombose de Bretagne Occidentale (GETBO), Université de Brest (UBO)-Institut Brestois Santé Agro Matière (IBSAM), Université de Brest (UBO)-Université de Brest (UBO), ACS - Amsterdam Cardiovascular Sciences, Vascular Medicine, APH - Amsterdam Public Health, Cardiology, ANS - Amsterdam Neuroscience, Neurology, and Other departments
- Subjects
Male ,MESH: Factor Xa ,[SDV]Life Sciences [q-bio] ,Administration, Oral ,Kaplan-Meier Estimate ,030204 cardiovascular system & hematology ,THERAPY ,MESH: Venous Thromboembolism ,chemistry.chemical_compound ,0302 clinical medicine ,Edoxaban ,MESH: Double-Blind Method ,030212 general & internal medicine ,MESH: Warfarin ,MESH: Treatment Outcome ,MESH: Aged ,RISK ,MESH: Middle Aged ,General Medicine ,MESH: Follow-Up Studies ,Venous Thromboembolism ,Middle Aged ,3. Good health ,Pulmonary embolism ,Treatment Outcome ,MESH: Administration, Oral ,Acute Disease ,MESH: Acute Disease ,Apixaban ,Female ,MESH: Hemorrhage ,medicine.drug ,Andexanet alfa ,Adult ,medicine.medical_specialty ,MESH: Enoxaparin ,Pyridones ,PULMONARY-EMBOLISM ,Hemorrhage ,MESH: Anticoagulants ,03 medical and health sciences ,Double-Blind Method ,BINOMIAL TRIALS ,Internal medicine ,MESH: Pyridones ,medicine ,Humans ,Enoxaparin ,MESH: Kaplan-Meier Estimate ,RIVAROXABAN ,Aged ,Rivaroxaban ,MESH: Humans ,business.industry ,Warfarin ,Anticoagulants ,MESH: Adult ,medicine.disease ,Confidence interval ,MESH: Male ,Surgery ,chemistry ,Relative risk ,Pyrazoles ,business ,MESH: Female ,MESH: Pyrazoles ,Factor Xa Inhibitors ,Follow-Up Studies - Abstract
International audience; BACKGROUND: Apixaban, an oral factor Xa inhibitor administered in fixed doses, may simplify the treatment of venous thromboembolism. METHODS: In this randomized, double-blind study, we compared apixaban (at a dose of 10 mg twice daily for 7 days, followed by 5 mg twice daily for 6 months) with conventional therapy (subcutaneous enoxaparin, followed by warfarin) in 5395 patients with acute venous thromboembolism. The primary efficacy outcome was recurrent symptomatic venous thromboembolism or death related to venous thromboembolism. The principal safety outcomes were major bleeding alone and major bleeding plus clinically relevant nonmajor bleeding. RESULTS: The primary efficacy outcome occurred in 59 of 2609 patients (2.3%) in the apixaban group, as compared with 71 of 2635 (2.7%) in the conventional-therapy group (relative risk, 0.84; 95% confidence interval [CI], 0.60 to 1.18; difference in risk [apixaban minus conventional therapy], -0.4 percentage points; 95% CI, -1.3 to 0.4). Apixaban was noninferior to conventional therapy (P
- Published
- 2013
- Full Text
- View/download PDF
58. 51TiP - KEYNOTE-185: A randomized, open-label phase 3 study of pembrolizumab in combination with lenalidomide and low-dose dexamethasone in newly diagnosed and treatment-naive multiple myeloma (MM)
- Author
-
Lonial, S., Ribeiro de Oliveira, M., Yimer, H., Mateos, M.V., Rifkin, R., Schjesvold, F., San-Miguel, J., Ghori, R., Marinello, P., and Jagannath, S.
- Published
- 2016
- Full Text
- View/download PDF
59. Autologous bone marrow Th cells can support multiple myeloma cell proliferation in vitroand in xenografted mice
- Author
-
Wang, D, Fløisand, Y, Myklebust, C V, Bürgler, S, Parente-Ribes, A, Hofgaard, P O, Bogen, B, Taskén, K, Tjønnfjord, G E, Schjesvold, F, Dalgaard, J, Tveita, A, and Munthe, L A
- Abstract
Multiple myeloma (MM) is a plasma cell malignancy where MM cell growth is supported by the bone marrow (BM) microenvironment with poorly defined cellular and molecular mechanisms. MM cells express CD40, a receptor known to activate autocrine secretion of cytokines and elicit proliferation. Activated T helper (Th) cells express CD40 ligand (CD40L) and BM Th cells are significantly increased in MM patients. We hypothesized that activated BM Th cells could support MM cell growth. We here found that activated autologous BM Th cells supported MM cell growth in a contact- and CD40L-dependent manner in vitro.MM cells had retained the ability to activate Th cells that reciprocated and stimulated MM cell proliferation. Autologous BM Th cells supported MM cell growth in xenografted mice and were found in close contact with MM cells. MM cells secreted chemokines that attracted Th cells, secretion was augmented by CD40-stimulation. Within 14 days of culture of whole BM aspirates in autologous serum, MM cells and Th cells mutually stimulated each other, and MM cells required Th cells for further expansion in vitroand in mice. The results suggest that Th cells may support the expansion of MM cells in patients.
- Published
- 2017
- Full Text
- View/download PDF
60. Neoplastic plasma cells with Auer rod‐like inclusions
- Author
-
Nordberg Nørgaard Jakob, Bugge Askeland Frida, Tjønnfjord Geir, and Schjesvold Fredrik
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
61. Minimal Residual Disease (MRD) follow up of MM patients obtaining stringent complete remission (sCR) in the NMSG/EMN02 clinical trial – defining sCR numbers and prediction of disease progression by multiparametric flow cytometry (MFC)
- Author
-
Hans Erik Johnsen, Alexander Schmitz, Julie Støve Bødker, Sonneveld, P., Hofste Op Bruinink, D., Holt, B., Velden, V. H. J., Abildgaard, N., Ahlberg, L., Andersen, N., Andreasson, B., Brandefors, L., Christiansen, H. B., Faxoe, S., Fransson, C., Friestad, M., Gregersen, H., Hammerlund, Y., Hansson, M., Haukaas, E., Hector, M., Helleberg, C., Holst, M., Jönsson, A. K., Karlsson, V., Klostergaard, A., Kjær, S., Lauri, B., Lenhoff, S., Lonn, I., Myhr, E., Pedersen, B. H., Renna, R., Sjøgren, T. V., Asta Svirskaite, Szatkowski, D. L., Vangsted, S., Waage, A., Schjesvold, F. H., U-H, Mellqvist, Helle Høholt, Louise Hvilshøj Madsen, Henning Sand Christensen, Rasmus Brøndum, Martin Bøgsted, and Karen Dybkaer
62. Treatment of multiple myeloma-related bone disease
- Author
-
Michele Cavo, Ioannis Ntanasis-Stathopoulos, Charalampia Kyriakou, Brian G.M. Durie, Jesús F. San-Miguel, Fredrik Schjesvold, Philippe Moreau, Sonja Zweegman, Noopur Raje, Evangelos Terpos, Nikhil C. Munshi, Matthew T. Drake, Meletios A. Dimopoulos, Elena Zamagni, Suzanne Lentzsch, Niels Abildgaard, Jens Hillengass, Javier de la Rubia, Ramón García-Sanz, Terpos E., Zamagni E., Lentzsch S., Drake M.T., Garcia-Sanz R., Abildgaard N., Ntanasis-Stathopoulos I., Schjesvold F., de la Rubia J., Kyriakou C., Hillengass J., Zweegman S., Cavo M., Moreau P., San-Miguel J., Dimopoulos M.A., Munshi N., Durie B.G.M., and Raje N.
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Bone Density Conservation Agent ,Bone disease ,03 medical and health sciences ,0302 clinical medicine ,Spinal cord compression ,medicine ,Humans ,Multiple myeloma ,Bone Density Conservation Agents ,business.industry ,medicine.disease ,Surgery ,Discontinuation ,Transplantation ,030104 developmental biology ,Zoledronic acid ,Denosumab ,Oncology ,030220 oncology & carcinogenesis ,Practice Guidelines as Topic ,Bone Diseases ,business ,Bone Disease ,Multiple Myeloma ,medicine.drug ,Human - Abstract
In this Policy Review, the Bone Working Group of the International Myeloma Working Group updates its clinical practice recommendations for the management of multiple myeloma-related bone disease. After assessing the available literature and grading recommendations using the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) method, experts from the working group recommend zoledronic acid as the preferred bone-targeted agent for patients with newly diagnosed multiple myeloma, with or without multiple myeloma-related bone disease. Once patients achieve a very good partial response or better, after receiving monthly zoledronic acid for at least 12 months, the treating physician can consider decreasing the frequency of or discontinuing zoledronic acid treatment. Denosumab can also be considered for the treatment of multiple myeloma-related bone disease, particularly in patients with renal impairment. Denosumab might prolong progression-free survival in patients with newly diagnosed multiple myeloma who have multiple myeloma-related bone disease and who are eligible for autologous stem-cell transplantation. Denosumab discontinuation is challenging due to the rebound effect. The Bone Working Group of the International Myeloma Working Group also found cement augmentation to be effective for painful vertebral compression fractures. Radiotherapy is recommended for uncontrolled pain, impeding or symptomatic spinal cord compression, or pathological fractures. Surgery should be used for the prevention and restoration of long-bone pathological fractures, vertebral column instability, and spinal cord compression with bone fragments within the spinal route.
- Published
- 2021
- Full Text
- View/download PDF
63. COVID-19 vaccination in patients with multiple myeloma: a consensus of the European Myeloma Network
- Author
-
Heinz Ludwig, Pieter Sonneveld, Thierry Facon, Jesus San-Miguel, Hervé Avet-Loiseau, Mohamad Mohty, Maria-Victoria Mateos, Philippe Moreau, Michele Cavo, Charlotte Pawlyn, Sonja Zweegman, Monika Engelhardt, Christoph Driessen, Gordon Cook, Melitios A Dimopoulos, Francesca Gay, Hermann Einsele, Michel Delforge, Jo Caers, Katja Weisel, Graham Jackson, Laurent Garderet, Niels van de Donk, Xavier Leleu, Hartmut Goldschmidt, Meral Beksac, Inger Nijhof, Martin Schreder, Niels Abildgaard, Roman Hajek, Niklas Zojer, Efstathios Kastritis, Annemiek Broijl, Fredrik Schjesvold, Mario Boccadoro, Evangelos Terpos, Hematology, Ludwig H., Sonneveld P., Facon T., San-Miguel J., Avet-Loiseau H., Mohty M., Mateos M.-V., Moreau P., Cavo M., Pawlyn C., Zweegman S., Engelhardt M., Driessen C., Cook G., Dimopoulos M.A., Gay F., Einsele H., Delforge M., Caers J., Weisel K., Jackson G., Garderet L., van de Donk N., Leleu X., Goldschmidt H., Beksac M., Nijhof I., Schreder M., Abildgaard N., Hajek R., Zojer N., Kastritis E., Broijl A., Schjesvold F., Boccadoro M., Terpos E., and Austrian Forum Against Cancer
- Subjects
Oncology ,medicine.medical_specialty ,COVID-19 Vaccines ,Consensus ,medicine.drug_class ,medicine.medical_treatment ,COVID-19 Vaccine ,Consensu ,Review ,Disease ,Monoclonal antibody ,Immune system ,SDG 3 - Good Health and Well-being ,Internal medicine ,Concomitant Therapy ,medicine ,Humans ,Multiple myeloma ,business.industry ,SARS-CoV-2 ,Vaccination ,COVID-19 ,Immunosuppression ,Hematology ,medicine.disease ,Discontinuation ,Practice Guidelines as Topic ,business ,Multiple Myeloma ,Human - Abstract
Patients with multiple myeloma frequently present with substantial immune impairment and an increased risk for infections and infection-related mortality. The risk for infection with SARS-CoV-2 virus and resulting mortality is also increased, emphasising the importance of protecting patients by vaccination. Available data in patients with multiple myeloma suggest a suboptimal anti-SARS-CoV-2 immune response, meaning a proportion of patients are unprotected. Factors associated with poor response are uncontrolled disease, immunosuppression, concomitant therapy, more lines of therapy, and CD38 antibody-directed and B-cell maturation antigen-directed therapy. These facts suggest that monitoring the immune response to vaccination in patients with multiple myeloma might provide guidance for clinical management, such as administration of additional doses of the same or another vaccine, or even temporary treatment discontinuation, if possible. In those who do not exhibit a good response, prophylactic treatment with neutralising monoclonal antibody cocktails might be considered. In patients deficient of a SARS-CoV-2 immune response, adherence to measures for infection risk reduction is particularly recommended. This consensus was generated by members of the European Multiple Myeloma Network and some external experts. The panel members convened in virtual meetings and conducted an extensive literature research and evaluated recently published data and work presented at meetings, as well as findings from their own studies. The outcome of the discussions on establishing consensus recommendations for COVID-19, This study has been funded by the Austrian Forum against Cancer, which covered in part expenses for interaction between authors and for secretarial support.
- Published
- 2021
- Full Text
- View/download PDF
64. Treatment of relapsed and refractory multiple myeloma: recommendations from the International Myeloma Working Group
- Author
-
Mario Boccadoro, Peter Leif Bergsagel, Noopur Raje, Xavier Leleu, Cristina João, Joseph R. Mikhael, Monika Engelhardt, Brian G.M. Durie, Sarah A. Holstein, Heinz Ludwig, Parameswaran Hari, Gordon Cook, Anders Waage, Maria-Victoria Mateos, Ulf-Henrik Mellqvist, Hareth Nahi, Faith E. Davies, Jian Hou, Angelo Maiolino, Saad Z. Usmani, Nizar J. Bahlis, Wee Joo Chng, Sigurdur Y. Kristinsson, Fernando Leal da Costa, Hartmut Goldschmidt, Evangelos Terpos, Pieter Sonneveld, Dominik Dytfeld, Artur Jurczyszyn, Michele Cavo, Fredrik Schjesvold, Meletios A. Dimopoulos, Jacob P. Laubach, Jesus San Miguel, Kwee Yong, Elena Zamagni, Orhan Sezer, Martin Kaiser, Surbhi Sidana, Vania Hungria, Meral Beksac, Enrique M. Ocio, Nikhil C. Munshi, Kenneth C. Anderson, Katja Weisel, Thierry Facon, Annette Juul Vangsted, Christoph Driessen, Hermann Einsele, Luciano J. Costa, Shaji Kumar, Paul G. Richardson, Philippe Moreau, Jesus G. Berdeja, Roman Hájek, Thomas G. Martin, Sagar Lonial, David H. Vesole, Rafael Fonseca, Suzanne Lentzsch, Eloisa Riva, Simon J. Harrison, Hang Quach, Rachid Baz, S. Vincent Rajkumar, Niels W.C.J. van de Donk, Joan Bladé, Jean-Luc Harousseau, Sonja Zweegman, Moreau P., Kumar S.K., San Miguel J., Davies F., Zamagni E., Bahlis N., Ludwig H., Mikhael J., Terpos E., Schjesvold F., Martin T., Yong K., Durie B.G.M., Facon T., Jurczyszyn A., Sidana S., Raje N., van de Donk N., Lonial S., Cavo M., Kristinsson S.Y., Lentzsch S., Hajek R., Anderson K.C., Joao C., Einsele H., Sonneveld P., Engelhardt M., Fonseca R., Vangsted A., Weisel K., Baz R., Hungria V., Berdeja J.G., Leal da Costa F., Maiolino A., Waage A., Vesole D.H., Ocio E.M., Quach H., Driessen C., Blade J., Leleu X., Riva E., Bergsagel P.L., Hou J., Chng W.J., Mellqvist U.-H., Dytfeld D., Harousseau J.-L., Goldschmidt H., Laubach J., Munshi N.C., Gay F., Beksac M., Costa L.J., Kaiser M., Hari P., Boccadoro M., Usmani S.Z., Zweegman S., Holstein S., Sezer O., Harrison S., Nahi H., Cook G., Mateos M.-V., Rajkumar S.V., Dimopoulos M.A., Richardson P.G., and Hematology
- Subjects
medicine.medical_specialty ,Line of therapy ,Drug Resistance ,Salvage therapy ,Antineoplastic Agents ,030204 cardiovascular system & hematology ,Antineoplastic Agent ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Intensive care medicine ,Multiple myeloma ,Salvage Therapy ,Hematology ,business.industry ,Cancer ,Refractory Multiple Myeloma ,medicine.disease ,Drug access ,Clinical research ,Neoplasm Recurrence ,Oncology ,Local ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Practice Guidelines as Topic ,Neoplasm ,Multiple Myeloma ,Neoplasm Recurrence, Local ,business ,Human - Abstract
This Policy Review presents the International Myeloma Working Group's clinical practice recommendations for the treatment of relapsed and refractory multiple myeloma. Based on the results of phase 2 and phase 3 trials, these recommendations are proposed for the treatment of patients with relapsed and refractory disease who have received one previous line of therapy, and for patients with relapsed and refractory multiple myeloma who have received two or more previous lines of therapy. These recommendations integrate the issue of drug access in both low-income and middle-income countries and in high-income countries to help guide real-world practice and thus improve patient outcomes.
- Published
- 2021
- Full Text
- View/download PDF
65. Pembrolizumab plus lenalidomide and dexamethasone for patients with treatment-naive multiple myeloma (KEYNOTE-185): a randomised, open-label, phase 3 trial
- Author
-
Saad Zafar Usmani, Fredrik Schjesvold, Albert Oriol, Lionel Karlin, Michele Cavo, Robert M Rifkin, Habte Aragaw Yimer, Richard LeBlanc, Naoki Takezako, Robert Donald McCroskey, Andrew Boon Ming Lim, Kenshi Suzuki, Hiroshi Kosugi, George Grigoriadis, Irit Avivi, Thierry Facon, Sundar Jagannath, Sagar Lonial, Razi Uddin Ghori, Mohammed Z H Farooqui, Patricia Marinello, Jesus San-Miguel, Andrew Lim, Trish Walker, Andrew Nicol, Donna Reece, Mohamed Elemary, Jean Samuel Boudreault Pedneault, Michel Attal, Katja Weisel, Monika Engelhardt, Andreas Mackensen, John Quinn, Amos Cohen, Hila Magen-Nativ, Noam Benyamini, Alessandra Larocca, Morio Matsumoto, Shinsuke Iida, Takayuki Ishikawa, Yukio Kondo, Kazutaka Sunami, Kiyoshi Ando, Takanori Teshima, Takaaki Chou, Hiromi Iwasaki, Hirokazu Miki, Itaru Matsumura, Yasushi Onishi, Koji Izutsu, Masahiro Kizaki, Anupkumar George, Hillary Blacklock, David Simpson, Anders Waage, Olga Samoilova, Evgeniy Nikitin, Tatiana Chagorova, Andrew McDonald, Moosa Patel, Albert Oriol Rocafiguera, Jesus San Miguel Izquierdo, Maria Mateos, Matthew Streetly, Peter Forsyth, Graham Jackson, Stephen Jenkins, Robert Rifkin, Habte Yimer, Robert McCroskey, Danko Martincic, Stefano Tarantolo, Sarah Larson, Yacoub Faroun, Jennifer Vaughn, Rachid Baz, Gene Saylors, Amarendra Neppalli, Anastasios Raptis, Henry Fung, Maxwell Janosky, Don Stevens, Morton Coleman, Dennis Costa, Scott Cross, Suzanne Fanning, Daniel Farray Berges, Thomas Harris, Ira Zackon, Djordje Atanackovic, Kelvin Lee, Ira Oliff, Wes Lee, William Bensinger, Jose Lutzky, Ari Baron, Fadi Hayek, Eli Kirschner, Neeraj Bharany, Lindsay Overton, Siva Mannem, Allyson Harroff, Sharad Jain, Tammy Roque, Kristi McIntyre, Christopher K Yasencha, William Houck, Usmani SZ1, Schjesvold F2, Oriol A3, Karlin L4, Cavo M5, Rifkin RM6, Yimer HA7, LeBlanc R8, Takezako N9, McCroskey RD10, Lim ABM11, Suzuki K12, Kosugi H13, Grigoriadis G14, Avivi I15, Facon T16, Jagannath S17, Lonial S18, Ghori RU19, Farooqui MZH19, Marinello P19, San-Miguel J20, and KEYNOTE-185 Investigators. Lim A, Grigoriadis G, Walker T, Nicol A, LeBlanc R, Reece D, Elemary M, Boudreault Pedneault JS, Karlin L, Facon T, Attal M, Weisel K, Engelhardt M, Mackensen A, Quinn J, Avivi I, Cohen A, Magen-Nativ H, Benyamini N, Cavo M, Larocca A, Takezako N, Suzuki K, Kosugi H, Matsumoto M, Iida S, Ishikawa T, Kondo Y, Sunami K, Ando K, Teshima T, Chou T, Iwasaki H, Miki H, Matsumura I, Onishi Y, Izutsu K, Kizaki M, George A, Blacklock H, Simpson D, Schjesvold F, Waage A, Samoilova O, Nikitin E, Chagorova T, McDonald A, Patel M, Oriol Rocafiguera A, San Miguel Izquierdo J, Mateos M, Streetly M, Forsyth P, Jackson G, Jenkins S, Rifkin R, Yimer H, McCroskey R, Martincic D, Tarantolo S, Larson S, Faroun Y, Vaughn J, Baz R, Saylors G, Neppalli A, Raptis A, Fung H, Janosky M, Stevens D, Coleman M, Costa D, Cross S, Fanning S, Berges DF, Harris T, Zackon I, Atanackovic D, Lee K, Oliff I, Lee W, Bensinger W, Lutzky J, Baron A, Hayek F, Kirschner E, Bharany N, Overton L, Mannem S, Harroff A, Jain S, Roque T, McIntyre K, Yasencha CK, Houck W.
- Subjects
Male ,medicine.medical_specialty ,Pembrolizumab ,Antibodies, Monoclonal, Humanized ,Dexamethasone ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Progression-free survival ,Lenalidomide ,Survival rate ,Multiple myeloma ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,business.industry ,Pneumonia ,Hematology ,Interim analysis ,medicine.disease ,Progression-Free Survival ,Survival Rate ,Treatment Outcome ,030220 oncology & carcinogenesis ,Female ,Multiple Myeloma ,business ,Pembrolizumab, lenalidomide, dexamethasone ,030215 immunology ,medicine.drug - Abstract
Summary Background Lenalidomide and dexamethasone has been a standard of care in transplant-ineligible patients with newly diagnosed multiple myeloma. The addition of a third drug to the combination is likely to improve treatment efficacy. KEYNOTE-185 assessed the efficacy and safety of lenalidomide and dexamethasone with and without pembrolizumab in patients with previously untreated multiple myeloma. Here, we present the results of an unplanned interim analysis done to assess the benefit–risk of the combination at the request of the US Food and Drug Administration (FDA). Methods KEYNOTE-185 was a randomised, open-label, phase 3 trial done at 95 medical centres across 15 countries (Australia, Canada, France, Germany, Ireland, Israel, Italy, Japan, New Zealand, Norway, Russia, South Africa, Spain, UK, and USA). Transplantation-ineligible patients aged 18 years and older with newly diagnosed multiple myeloma, Eastern Cooperative Oncology Group performance status of 0 or 1, and who were treatment naive were enrolled, and randomly assigned 1:1 to receive either pembrolizumab plus lenalidomide and dexamethasone or lenalidomide and dexamethasone alone using an interactive voice or integrated web response system. Patients received oral lenalidomide 25 mg on days 1–21 and oral dexamethasone 40 mg on days 1, 8, 15, and 22 of repeated 28-day cycles, with or without intravenous pembrolizumab 200 mg every 3 weeks. The primary endpoint was progression-free survival, which was investigator-assessed because of early trial termination. Efficacy was analysed in all randomly assigned patients and safety was analysed in all patients who received at least one dose of study drug. This trial is registered at ClinicalTrials.gov , number NCT02579863 , and it is closed for accrual. Findings Between Jan 7, 2016, and June 9, 2017, 301 patients were randomly assigned to the pembrolizumab plus lenalidomide and dexamethasone group (n=151) or the lenalidomide and dexamethasone group (n=150). On July 3, 2017, the FDA decided to halt the study because of the imbalance in the proportion of death between groups. At database cutoff (June 2, 2017), with a median follow-up of 6·6 months (IQR 3·4–9·6), 149 patients in the pembrolizumab plus lenalidomide and dexamethasone group and 145 in the lenalidomide and dexamethasone group had received their assigned study drug. Median progression-free survival was not reached in either group; progression-free survival estimates at 6-months were 82·0% (95% CI 73·2–88·1) versus 85·0% (76·8–90·5; hazard ratio [HR] 1·22; 95% CI 0·67–2·22; p=0·75). Serious adverse events were reported in 81 (54%) patients in the pembrolizumab plus lenalidomide and dexamethasone group versus 57 (39%) patients in the lenalidomide and dexamethasone group; the most common serious adverse events were pneumonia (nine [6%]) and pyrexia (seven [5%]) in the pembrolizumab plus lenalidomide and dexamethasone group and pneumonia (eight [6%]) and sepsis (two [1%]) in the lenalidomide and dexamethasone group. Six (4%) treatment-related deaths occurred in the pembrolizumab plus lenalidomide and dexamethasone group (cardiac arrest, cardiac failure, myocarditis, large intestine perforation, pneumonia, and pulmonary embolism) and two (1%) in the lenalidomide and dexamethasone group (upper gastrointestinal haemorrhage and respiratory failure). Interpretation The results from this unplanned, FDA-requested, interim analysis showed that the benefit–risk profile of pembrolizumab plus lenalidomide and dexamethasone is unfavourable for patients with newly diagnosed, previously untreated multiple myeloma. Long-term safety and survival follow-up is ongoing. Funding Merck Sharp & Dohme, a subsidiary of Merck & Co, Inc (Kenilworth, NJ, USA).
- Published
- 2019
66. 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
- Full Text
- View/download PDF
67. International myeloma working group immunotherapy committee recommendation on sequencing immunotherapy for treatment of multiple myeloma.
- Author
-
Costa LJ, Banerjee R, Mian H, Weisel K, Bal S, Derman BA, Htut MM, Nagarajan C, Rodriguez C, Richter J, Frigault MJ, Ye JC, van de Donk NWCJ, Voorhees PM, Puliafito B, Bahlis N, Popat R, Chng WJ, Ho PJ, Kaur G, Kapoor P, Du J, Schjesvold F, Berdeja J, Einsele H, Cohen AD, Mikhael J, Biru Y, Rajkumar SV, Lin Y, Martin TG, and Chari A
- Abstract
T-cell redirecting therapy (TCRT), specifically chimeric antigen receptor T-cell therapy (CAR T-cells) and bispecific T-cell engagers (TCEs) represent a remarkable advance in the treatment of multiple myeloma (MM). There are several products available around the world and several more in development targeting primarily B-cell maturation antigen (BCMA) and G protein-coupled receptor class C group 5 member D (GRPC5D). The relatively rapid availability of multiple immunotherapies brings the necessity to understand how a certain agent may affect the safety and efficacy of a subsequent immunotherapy so MM physicians and patients can aim at optimal sequential use of these therapies. The International Myeloma Working Group conveyed panel of experts to review patient and disease-related factors affecting efficacy and safety of immunotherapy, summarize existing information on sequencing therapy and provide a series of core recommendations., Competing Interests: Competing interests: LJC: Honoraria (Amgen, Bristol-Myers-Squibb, AbbVie, Pfizer, Johnson & Johnson, Adaptive Biotechnologies, Sanofi, Genentech/Roche), Research Funding (Amgen, Bristol-Myers-Squibb, Johnson & Johnson, AbbVie, Caribou, Gracell, Genentech/Roche); RB: Honoraria (Bristol-Myers-Squibb, Pfizer, Adaptive Biotechnologies, Caribou, Genentech/Roche, GSK, Karyopharm, Legend Biotech, Johnson & Johnson, SparkCures), Research Funding (AbbVie, Bristol-Myers-Squibb, Johnson & Johnson, Novartis, Pack Health, Prothena, Sanofi); HM: Honoraria (Bristol-Myers-Squibb, Pfizer, GSK, Karyopharm, AbbVie) Research Funding (Pfizer); SB: Honoraria (AbbVie, Adaptive Biotechnologies, Bristol-Myers-Squibb, Johnson & Johnson, MJH Lifesciences); BD: Honoraria (Sanofi, Johnson & Johnson, Canopy, COTA, Bristol-Myers-Squibb) Research Funding (Amgen, GSK); CN: Honoraria (Johnson & Johnson); JR: Honoraria (Janssen, Bristol-Myers-Squibb, Pfizer, Karyopharm, Sanofi, Takeda, Genentech, AbbVie, Regeneron, Forus, Menarini, Adaptive Biotechnologies); KW:Honoraria (Abbvie, Amgen, Adaptive Biotech, Astra Zeneca, Beigene, BMS, Celgene, Janssen, GSK, Karyopharm, Menarini, Novartis, Oncopeptides, Pfizer, Roche, Sanofi, Stemline, Takeda) Research Funding (Amgen, BMS/Celgene, Janssen, Sanofi; GSK, Abbvie) Consultancy (Abbvie, Amgen, Adaptive Biotech, Beigene, BMS, Celgene, Janssen, GSK, Karyopharm, Menarini, Novartis, Oncopeptides, Pfizer, Regeneron, Roche, Sanofi, Takeda); MJF: Honoraria (Kite/Gilead, CMS, Novartis, Janssen, Legend, Cytoagents, SITC); JCY: Honoraria (Janssen, Sanofi, Bristol-Myers-Squibb, Regeneron, GSK, Pfizer, Menarini); NWCJvdD: Honoraria (Janssen, Amgen, Celgene, Novartis, Cellectis, Bristol-Myers-Squibb/Celgene, Sanofi, Takeda, Roche, Novartis, Bayer, Adaptive, Pfizer, AbbVie, Servier) Research Funding (Janssen, Amgen, Bristol-Myers-Squibb/Celgene, Novartis, Cellectis); NB: Honoraria (AbbVie, Amgen, Bristol-Myers-Squibb, Forus, Genentech, GlaxoSmithKline, Janssen, Pfizer, Sanofi, Takeda) Research Funding (Pfizer); RP: Honoraria (GSK, Pfizer, Janssen, Bristol-Myers-Squibb, Abbvie, Roche) Research Funding (GSK, Pfizer); W-JC: Honoraria (AbbVie, Amgen, Pfizer, Sanofi, Regeneron, GSK, Novartis) Research Funding (Bristol-Myers-Squibb, Janssen, Novartis); PJH: Advisory Board without honoraria (Antengene, Gilead, iTeos Therapeutics, GSK, Janssen, Pfizer), Research Funding: Novartis; GK: Honoraria (Janssen, Bristol-Myers-Squibb, Sanofi, Kite, Arcellx, Pfizer, Kedrion, Cellectar); PK: Research Funding (Takeda Pharmaceuticals, Celgene, Sanofi, AbbVie, Karyopharm Therapeutics, Sorrento Therapeutics, Ichnos Sciences, Amgen); FS: Honoraria (AbbVie, GSK, Celgene, Takeda, Janssen, Oncopeptides, Sanofi, Bristol-Myers-Squibb, Novartis, SkyliteDX, Pfizer, Daiki-Sankyo,); JB: Honoraria (AstraZeneca, Bristol-Myers-Squibb, Caribou Biosciences, Galapagos, Janssen, K36 Therapeutics, Kite Pharma, Legend Biotech, Pfizer, Regeneron, Roche, Sanofi, Sebia, Takeda) Research Funding (2 Seventy Bio, Abbvie, Amgen, Bristol-Myers-Squibb, C4 Therapeutics, Caribou Biosciences, CARsgen, Cartesian Therapeutics, Celularity, CRISPR Therapeutics, Fate Therapeutics, Genentech, GSK, Ichnos Sciences, Incyte, Janssen, Juno Therapeutics, K36 Therapeutics. Karyopharm, Lilly, Novartis, Poseida, Roche, Takeda); HE: Honoraria (Bristol-Myers-Squibb, Celgene, Janssen, Amgen, Takeda, Sanofi, GSK, Novartis, Roche), Research Funding (BMS/Celgene, Janssen, Sanofi; GSK, Amgen) Consultancy (Amgen, BMS, Celgene, Janssen, GSK, Novartis, Roche, Sanofi, Takeda); ADC: Honoraria (GSK; Bristol-Myers-Squibb, Janssen, AbbVie, Pfizer, iTeos, Ichnos, Arcellx, Legend; Genentech/Roche, Novartis, AstraZeneca, Kite); Research Funding (GSK, Genentech, Janssen, BMS); Patent (Novartis); JM: Honoraria (Amgen, Sanofi, Bristol-Myers-Squibb, Janssen, Takeda); YL: Honoraria (Janssen, Sanofi, NexImmune, Caribou, Bristol-Myers-Squibb, Pfizer, Regeneron, Genentech; Sanofi, Regeneron) Research Funding (Janssen, Bristol-Myers-Squibb); TGM: Honoraria (GSK, Sanofi, Amgen, Janssen); AC: Honoraria (AbbVie, Adaptive biotechnologies, Amgen, Angengene, Bristol-Myers-Squibb, Forus, Genentech/Roche, GSK, Janssen, Karyopharm, Millenium/Takeda, Sanofi). The remaining authors declare no conflict of interest. The work presented in this manuscript did not receive any financial or material support., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
68. A plain language summary of the ICARIA study, comparing isatuximab-pomalidomide- dexamethasone with pomalidomide- dexamethasone in people with multiple myeloma.
- Author
-
Richardson PG, Perrot A, San-Miguel J, Beksac M, Spicka I, Leleu X, Schjesvold F, Moreau P, Dimopoulos MA, Huang SY, Minarik J, Cavo M, Prince HM, and Anderson KC
- Published
- 2025
- Full Text
- View/download PDF
69. Combined Proteasome and Autophagy Inhibition in Relapsed/Refractory Multiple Myeloma-A Phase I Trial of Hydroxychloroquine, Carfilzomib, and Dexamethasone.
- Author
-
Slørdahl TS, Askeland FB, Hanssen MSS, Hov H, Sundt-Hansen SM, Lindahl S, Vethe NT, Hjorth-Hansen H, Fenstad MH, Waage A, Hjertner Ø, Schjesvold F, and Sundan A
- Abstract
Multiple myeloma is characterized by malignant cells which produce high amounts of monoclonal immunoglobulin. Myeloma cells are, therefore, dependent on effective protein degradation. Proteasomal protein degradation is targeted by proteasome inhibitors in routine care. Autophagic protein degradation is currently not targeted in myeloma treatment. This Phase I trial showed that the combination of the proteasome inhibitor carfilzomib and the autophagy inhibitor hydroxychloroquine was well tolerated in patients with relapsed/refractory multiple myeloma. Adverse events were mostly Grades 1 and 2. An overall response rate of 44% indicates a meaningful clinical efficacy of this combination. Trial Registration : The study was registered at clinicaltrials.gov # NCT04163107., Competing Interests: Tobias S. Slørdahl has received honoraria from Takeda, Celgene, Amgen, Janssen, AbbVie, Pfizer; consultancy for Bristol Myers Squibb, GSK, Pfizer, Menarini Group, Sanofi; advisory board consultancy: Bristol Myers Squibb, Sanofi, Amgen, Celgene, GSK, Janssen. Frida Bugge Askeland has received consulting fees from Janssen, Sanofi, and Amgen, honoraria/payment from Janssen and Sanofi, participated in advisory boards for Janssen and Sanofi, and received research support from Sanofi. Fredrik Schjesvold has received grants from Targovax, consulting fees from GSK, BMS, Oncopeptides, XNK Therapeutics, Takeda, Janssen, Sanofi, and Galapagos, honoraria/payment from Amgen, BMS, Takeda, Sanofi, Menarini, AbbVie, Janssen, Oncopeptides, and GSK, and participated in advisory boards for AbbVie, Janssen, Regeneron, and Sanofi. The other authors declare no conflicts of interest., (© 2025 The Author(s). eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2025
- Full Text
- View/download PDF
70. Pomalidomide, Bortezomib, and Dexamethasone Versus Bortezomib and Dexamethasone in Relapsed or Refractory Multiple Myeloma: Final Survival and Subgroup Analyses From the OPTIMISMM Trial.
- Author
-
Richardson P, Beksaç M, Oriol A, Lindsay J, Schjesvold F, Galli M, Yağcı M, Larocca A, Weisel K, Yu X, Donahue C, Acosta J, Peluso T, and Dimopoulos M
- Abstract
Introduction: In the OPTIMISMM trial, pomalidomide/bortezomib/dexamethasone (PVd) significantly prolonged median progression-free survival (PFS) versus bortezomib/dexamethasone (Vd) in lenalidomide-exposed relapsed and refractory multiple myeloma (RRMM). We report final overall survival (OS) and updated efficacy analyses., Methods: Adults with RRMM who had 1-3 prior regimens, including lenalidomide (≥ 2 cycles), were assigned (1:1) to PVd or Vd., Primary Endpoint: PFS. Prespecified secondary endpoint: OS. Prespecified exploratory endpoints: PFS2 and subgroup efficacy analyses., Results: With an overall event rate of 70.0%, OS data were mature in the intent-to-treat population (N = 559). After median follow-up of 64.5 months (data cutoff: May 13, 2022), median OS was 35.6 months with PVd versus 31.6 months with Vd (HR 0.94, 95% CI 0.77-1.15, p = 0.571); adjusting for subsequent therapies, OS improved with PVd versus Vd (HR 0.76, 95% CI 0.619-0.931, p = 0.008). Median PFS2 was 22.1 versus 16.9 months, respectively (HR 0.77, 95% CI 0.64-0.94, nominal p = 0.008). Treatment-emergent adverse events led to study drug discontinuation in 92 (33.1%) and 53 (19.6%) patients in PVd and Vd arm, respectively., Conclusions: Findings showed a nonsignificant trend towards improved OS with PVd versus Vd. PFS2 favored PVd, supporting its use in RRMM., (© 2025 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2025
- Full Text
- View/download PDF
71. Daratumumab or Active Monitoring for High-Risk Smoldering Multiple Myeloma.
- Author
-
Dimopoulos MA, Voorhees PM, Schjesvold F, Cohen YC, Hungria V, Sandhu I, Lindsay J, Baker RI, Suzuki K, Kosugi H, Levin MD, Beksac M, Stockerl-Goldstein K, Oriol A, Mikala G, Garate G, Theunissen K, Spicka I, Mylin AK, Bringhen S, Uttervall K, Pula B, Medvedova E, Cowan AJ, Moreau P, Mateos MV, Goldschmidt H, Ahmadi T, Sha L, Cortoos A, Katz EG, Rousseau E, Li L, Dennis RM, Carson R, and Rajkumar SV
- Abstract
Background: Daratumumab, an anti-CD38 monoclonal antibody, has been approved for the treatment of multiple myeloma. Data are needed regarding the use of daratumumab for high-risk smoldering multiple myeloma, a precursor disease of active multiple myeloma for which no treatments have been approved., Methods: In this phase 3 trial, we randomly assigned patients with high-risk smoldering multiple myeloma to receive either subcutaneous daratumumab monotherapy or active monitoring. Treatment was continued for 39 cycles, for 36 months, or until confirmation of disease progression, whichever occurred first. The primary end point was progression-free survival; progression to active multiple myeloma was assessed by an independent review committee in accordance with International Myeloma Working Group diagnostic criteria., Results: Among the 390 enrolled patients, 194 were assigned to the daratumumab group and 196 to the active-monitoring group. With a median follow-up of 65.2 months, the risk of disease progression or death was 51% lower with daratumumab than with active monitoring (hazard ratio, 0.49; 95% confidence interval [CI], 0.36 to 0.67; P<0.001). Progression-free survival at 5 years was 63.1% with daratumumab and 40.8% with active monitoring. A total of 15 patients (7.7%) in the daratumumab group and 26 patients (13.3%) in the active-monitoring group died (hazard ratio, 0.52; 95% CI, 0.27 to 0.98). Overall survival at 5 years was 93.0% with daratumumab and 86.9% with active monitoring. The most common grade 3 or 4 adverse event was hypertension, which occurred in 5.7% and 4.6% of the patients in the daratumumab group and the active-monitoring group, respectively. Adverse events led to treatment discontinuation in 5.7% of the patients in the daratumumab group, and no new safety concerns were identified., Conclusions: Among patients with high-risk smoldering multiple myeloma, subcutaneous daratumumab monotherapy was associated with a significantly lower risk of progression to active multiple myeloma or death and with higher overall survival than active monitoring. No unexpected safety concerns were identified. (Funded by Janssen Research and Development; AQUILA ClinicalTrials.gov number, NCT03301220.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
72. Health-Related Quality of Life During Carfilzomib-Lenalidomide-Dexamethasone Consolidation: Findings From the Multiple Myeloma CONPET Study.
- Author
-
Rosenberg T, Möller S, Abildgaard N, Nørgaard JN, Lysén A, Tsykonova G, Joao C, Vangsted A, Schjesvold F, and Nielsen LK
- Abstract
Background: In the CONPET study, multiple myeloma patients with abnormal 18FDG positron emission/computed tomography scan after upfront autologous stem cell transplantation were treated with four cycles of carfilzomib-lenalidomide-dexamethasone (KRd). Side effect registrations show that carfilzomib might cause dyspnea, cough, respiratory tract infections, and heart failure. The aims were to investigate patient-reported shortness of breath and dyspnea during KRd consolidation., Methods: To assess shortness of breath, patients completed the Functional Assessment of Cancer Therapy-Pulmonary Symptom Index (FACT-PSI) and the EORTC QLQ-C30 to assess dyspnea. Shortness of breath was defined as decrease in FACT-PSI score or starting/increasing diuretic drugs. Mixed effect logistic regression was used for the effect analysis. Linear mixed model and clinical relevance were used to investigate dyspnea., Results: A total of 50 patients were included, median age 62 years (interquartile range 54-67). 17% reported shortness of breath at Day 15 Cycles 1-4 versus 11% at Day 1 Cycles 2-4, Cycle 4 Day 29, and 1 month posttreatment (p-value 0.048). Compared with baseline, patients reported significant, and clinically relevant worsening in dyspnea during consolidation., Conclusion: Our study confirmed earlier findings of carfilzomib causing shortness of breath during KRd administration and revealed dyspnea during consolidation compared to baseline., Trial Registration: Clinicaltrials.gov: NCT03314636, EudraCT: 2017-000586-72., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
73. Monoclonal Insulin Autoimmune Syndrome Successfully Treated With Plasma Cell Directed Therapy.
- Author
-
Askeland FB, Frøen HM, Bolstad N, Thorsby PM, Schjesvold F, Wammer ACP, Følling I, and Tjønnfjord GE
- Abstract
Background: Monoclonal insulin autoimmune syndrome (IAS) is a very rare disease characterized by severe attacks of hypoglycemia caused by circulating anti-insulin antibodies produced by a B-cell clone, usually clonal plasma cells., Method: We present 2 female Norwegian patients with monoclonal IAS. The anti-insulin antibodies were quantified by immune precipitation and characterized using a 3-step manual in-house assay. Both patients received plasma cell directed therapy., Result: The first patient received plasma cell directed therapy for a time-limited period and achieved a sustained clinical remission without detectable anti-insulin antibodies. The second patient receives continuous plasma cell directed therapy and is in clinical remission with low values of detectable anti-insulin antibodies., Conclusion: Plasma cell directed therapy was effective and safe in our 2 cases of monoclonal IAS. We recommend considering plasma cell directed therapy for these patients., Competing Interests: Disclosure Frida Bugge Askeland has received consulting fees from Janssen, Sanofi and Amgen, honoraria/payment from Janssen and Sanofi and participated in advisory boards for Janssen and Sanofi. Hege M. Frøen has no conflict of interest Nils Bolstad has no conflict of interest Per Medbøe Thorsby has received consulting fees from Antidoping Norway. Fredrik Schjesvold has received grants from Targovax, consulting fees from GSK, BMS, Oncopeptides, XNK Therapeutics, Takeda, Janssen, Sanofi and Galapagos, honoraria/payment from Amgen, BMS, Takeda, Sanofi, Menarini, AbbVie, Janssen, Oncopeptides and GSK and participated in advisory boards for AbbVie, Janssen, Regeneron and Sanofi. Anne Cathrine Parelius Wammer has received consulting fees from Novo Nordisk, honoraria/payment from Novo Nordisk, Sanofi and Buehringer and participated in advisory boards for Novo Nordisk. Ivar Følling has no conflict of interest. Geir E. Tjønnfjord has no conflict of interest., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
74. Advances and challenges in anti-cancer vaccines for multiple myeloma.
- Author
-
Abdollahi P, Norseth HM, and Schjesvold F
- Subjects
- Humans, Animals, Antigens, Neoplasm immunology, Immunotherapy methods, Multiple Myeloma immunology, Multiple Myeloma therapy, Cancer Vaccines immunology, Cancer Vaccines therapeutic use
- Abstract
Multiple myeloma (MM) is a hematological cancer marked by plasma cell accumulation in the bone marrow. Despite treatment advancements, MM remains incurable in most patients. MM-associated immune dysregulation fosters disease progression, prompting research into immunotherapy to combat the disease. An area of immunotherapy investigation is the design of myeloma vaccine therapy to reverse tumor-associated immune suppression and elicit tumor-specific immune responses to effectively target MM cells. This article reviews vaccine immunotherapy for MM, categorizing findings by antigen type and delivery method. Antigens include idiotype (Id), tumor-associated (TAA), tumor-specific (TSA), and whole tumor lysate. Myeloma vaccination has so far shown limited clinical efficacy. However, further studies are essential to optimize various aspects, including antigen and patient selection, vaccine timing and sequencing, and rational combinations with emerging MM treatments., Competing Interests: HN: Honoraria: Janssen, BMS and Sanofi. FS: Honoraria: Amgen, BMS, Takeda, Abbvie, Janssen, Novartis, SkyliteDX, Oncopeptides, Sanofi, Schain, Pfizer, Daiki-Sankyo; Consultancy: GSK, Celgene, Takeda, Janssen, Oncopeptides, Sanofi. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest., (Copyright © 2024 Abdollahi, Norseth and Schjesvold.)
- Published
- 2024
- Full Text
- View/download PDF
75. Consensus guidelines and recommendations for the management and response assessment of chimeric antigen receptor T-cell therapy in clinical practice for relapsed and refractory multiple myeloma: a report from the International Myeloma Working Group Immunotherapy Committee.
- Author
-
Lin Y, Qiu L, Usmani S, Joo CW, Costa L, Derman B, Du J, Einsele H, Fernandez de Larrea C, Hajek R, Ho PJ, Kastritis E, Martinez-Lopez J, Mateos MV, Mikhael J, Moreau P, Nagarajan C, Nooka A, O'Dwyer M, Schjesvold F, Sidana S, van de Donk NW, Weisel K, Zweegman S, Raje N, Otero PR, Anderson LD Jr, Kumar S, and Martin T
- Subjects
- Humans, Receptors, Antigen, T-Cell therapeutic use, Receptors, Antigen, T-Cell immunology, Treatment Outcome, Consensus, Immunotherapy, Adoptive adverse effects, Multiple Myeloma therapy, Multiple Myeloma immunology, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen therapeutic use
- Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown promise in patients with late-line refractory multiple myeloma, with response rates ranging from 73 to 98%. To date, three products have been approved: Idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel), which are approved by the US Food and Drug Administration, the European Medicines Agency, Health Canada (ide-cel only), and Brazil ANVISA (cilta-cel only); and equecabtagene autoleucel (eque-cel), which was approved by the Chinese National Medical Products Administration. CAR T-cell therapy is different from previous anti-myeloma therapeutics with unique toxic effects that require distinct mitigation strategies. Thus, a panel of experts from the International Myeloma Working Group was assembled to provide guidance for clinical use of CAR T-cell therapy in myeloma. This consensus opinion is from experts in the field of haematopoietic cell transplantation, cell therapy, and multiple myeloma therapeutics., Competing Interests: Declaration of interests YL received consultancy fees from Janssen, Sanofi, NexImmune, Caribou, Bristol Myers Squibb (BMS), Pfizer, Regeneron, and Genentech; research fees from Janssen and BMS; serves on advisory boards for Janssen, Sanofi, BMS, Regeneron, and Genentech; serves on scientific advisory boards for NexImmune and Caribou; and serves on the data safety monitor board for Pfizer. LQ received consultancy fees from Beigene, Xi'an, Janssen, Pfizer, Sanofi, and AstraZeneca; and is on the speaker board for Beigene, Xi'an, Janssen, Pfizer, Sanofi, AstraZeneca, and Roche. SU receives consultancy fees from AbbVie, Amgen, BMS, EdoPharma, Genentech, Gilead, GlaxoSmithKline (GSK), Janssen, Karyopharm Therapeutics, Merck, Oncopeptides, Sanofi, Seagen, Secura Bio, SkylineDx, and Takeda; and received research funding from AbbVie, Amgen, Array Biopharma, BMS, EdoPharma, Genentech, Gilead, GSK, Janssen, Merck, Moderna, Pharmacyclics, Seagen, Sanofi, SkylineDx, Takeda, and TeneoBio. CWJ received honoraria from AbbVie, Amgen, BMS, Pfizer, Sanofi, Regeneron, GSK, and Janssen; and research funding from Novartis, Janssen, and BMS Celgene. LC received research grants from Amgen, Janssen, BMS, Genentech, Caribou, and AbbVie; and honoraria from Amgen, Janssen, AbbVie, Pfizer, Sanofi, and Adaptive Biotechnologies. BD received advisory board and consulting fees from Janssen and COTA; research fees from GSK and Amgen; honoraria from the Multiple Myeloma Research Foundation and Plexus Communications; and is an independent reviewer for BMS. HE has an advisory role for and received consulting fees from BMS Celgene, Janssen, Amgen, GSK, and Sanofi; and received research funding from BMS Celgene, Janssen, Amgen, GSK, and Sanofi. CFdL received institutional grants from BMS, Janssen, and Amgen; honoraria from Amgen, Jassen, BMS, GSK, and Sanofi; support for attending meetings or travel from Janssen, BMS, GSK, and Amgen; is on data safety monitoring or advisory boards for Janssen, BMS, Amgen, Pfizer, and Sanofi; and received funding from the Spanish Institute of Health, the Asociación Española Contra el Cancer (AECC), the “La Caixa” Foundation, and AGAUR. RH received grants from Janssen, Amgen, Celgene, BMS, Novartis, and Takeda; consulting fees from Janssen, Amgen, Celgene, BMS, Novartis, Takeda, AbbVie, PharmaMar, Oncopeptides, Sanofi, and GSK; and honoraria from Janssen, Amgen, Celgene, BMS, PharmaMar, and Takeda. PJH serves on advisory boards (honoraria not accepted) for Antengene, Gilead, iTeos Therapeutics, Janssen, Novartis, and Pfizer. EK received honoraria from Janssen, Pfizer, GSK, and Prothena; and research support to the institution from GSK, Janssen, and Pfizer. JM-L received consultancy fees from Janssen, BMS, Sanofi, GSK, Novartis, Menarini, Incity, Roche, Gilead, Pfizer, and Karyopharm; and research funding from BMS, Janssen, Incity, Amgen, and Pfizer. M-VM received honoraria for serving on advisory boards and received fees for lectures from Janssen, BMS Celgene, Novartis, GSK, Sanofi, Amgen, Pfizer, AbbVie, and Regeneron. JM received consultancy fees from Amgen, BMS, Janssen, Sanofi, and Takeda; and research funding from BMS Clinical Trial. PM serves on advisory boards and received honoraria from Janssen and BMS Celgene. CN received consultancy fees from Janssen, BMS, Sanofi, GSK, Pfizer, Amgen, AstraZeneca, and DKSH; and research funding from Janssen and Amgen. AN serves on advisory boards and received honoraria from Adaptive Biotechnologies, Amgen, BMS, Cellectar Biosciences, GSK, Janssen, K36 therapeutics, ONK therapeutics, Pfizer, Sanofi, and Takeda; received research grants from Aduro Biotech, Amgen, Arch Oncology, BMS, Cellectis, Genentech, GSK, Janssen, Karyopharm, Kite Pharma, Merck, Pfizer, and Takeda; and received grants for investigator initiated studies from Amgen, GSK, Janssen, Merck, and Takeda. FS received consultancy fees from AbbVie, GSK, Celgene, Takeda, Janssen, Oncopeptides, Sanofi, and BMS; honoraria from Amgen, BMS, Takeda, AbbVie, Janssen, Novartis, SkyliteDX, Oncopeptides, Sanofi, Pfizer, Daiki-Sankyo, and GSK; and research grants from Celgene, Janssen, Oncopeptides, Sanofi, GSK, and Targovax. SS received research grants from Magenta Therapeutics, BMS, Allogene, Jansse, Novartis, AbbVie, Sanofi, Oncopeptides, Takeda, Kite, and Regeneron; and consultancy fees from Magenta Therapeutics, BMS, Janssen, AbbVie, Sanofi, Oncopeptides, Takeda, Kite, and Regeneron. NWCJvdD received research grants from Janssen, Amgen, Celgene, Novartis, Cellectis, and BMS; and serves on advisory boards for Janssen, Amgen, Celgene, BMS, Sanofi, Takeda, Roche, Novartis, Bayer, Adaptive, Pfizer, AbbVie, and Servier (all paid to institution). KW received research grants from AbbVie, Amgen, BMS Celgene, Janssen, GSK, Sanofi, and Takeda; honoraria from AbbVie, Amgen, Adaptive Biotech, Astra Zeneca, BMS Celgene, BeiGene, Janssen, GSK, Karyopharm, Novartis, Oncopeptides, Pfizer, Roche Pharma, Sanofi, Stemline, Takeda, and Menarini; and consulting fees from AbbVie, Amgen, Adaptive Biotech, BMS Celgene, BeiGene, Janssen, GSK, Karyopharm, Oncopeptides, Pfizer, Roche Pharma, Sanofi, Takeda, and Menarini. SZ serves on advisory boards for Janssen, BMS, Takeda, Sanofi, Oncopeptides, and Amgen (no personal fees). PRO received honoraria for consulting or advisory board from BMS Celgene, Janssen, Roche, AbbVie, Pfizer, GSK, Sanofi, H3Biomedicine; travel grants from Pfizer; serves on steering committees for BMS Celgene, Regeneron, and Janssen; and is on the speakers bureau for Janssen, BMS Celgene, GSK, Sanofi, and AbbVie. LDA received consulting fees and serves on advisory boards for Janssen, Celgene, BMS, Amgen, GSK, AbbVie, Beigene, Cellectar, Sanofi, and Prothena. SK received consultancy fees from Oncopeptides, AbbVie, Celgene, Janssen, Takeda, Adaptive, KITE, and MedImmune AstraZeneca; and research grants from AbbVie, Celgene, Janssen, Takeda, Adaptive, KITE, MedImmune AstraZeneca, Merck, Novartis, Roche, and Sanofi. TM received research funding from Sanofi. All other authors declare no competing interests. IMWG are listed in the appendix (pp 9–21)., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
76. Belantamab mafodotin: an important treatment option for vulnerable patients with triple class exposed relapsed and/or refractory multiple myeloma.
- Author
-
Mateos MV, Weisel K, Terpos E, Delimpasi S, Kastritis E, Zamagni E, Delforge M, Ocio E, Katodritou E, Gay F, Larocca A, Leleu X, Otero PR, Schjesvold F, Cavo M, and Dimopoulos MA
- Subjects
- Humans, Antibodies, Monoclonal, Humanized therapeutic use, Drug Resistance, Neoplasm drug effects, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Recurrence, Male, Antineoplastic Agents therapeutic use, Female, Multiple Myeloma drug therapy, Multiple Myeloma pathology
- Published
- 2024
- Full Text
- View/download PDF
77. Isatuximab-pomalidomide-dexamethasone versus pomalidomide-dexamethasone in patients with relapsed and refractory multiple myeloma: final overall survival analysis.
- Author
-
Richardson PG, Perrot A, Miguel JS, Beksac M, Spicka I, Leleu X, Schjesvold F, Moreau P, Dimopoulos MA, Huang SY, Minarik J, Cavo M, Prince HM, Macé S, Zhang R, Dubin F, Morisse MC, and Anderson KC
- Subjects
- Humans, Male, Aged, Female, Middle Aged, Aged, 80 and over, Adult, Drug Resistance, Neoplasm, Treatment Outcome, Recurrence, Survival Analysis, Multiple Myeloma drug therapy, Multiple Myeloma mortality, Multiple Myeloma pathology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone administration & dosage, Thalidomide analogs & derivatives, Thalidomide administration & dosage, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized therapeutic use
- Abstract
The primary and prespecified updated analyses of ICARIA-MM (clinicaltrial gov. Identifier: NCT02990338) demonstrated improved progression-free survival (PFS) and a benefit in overall survival (OS) was reported with the addition of isatuximab, an anti-CD38 monoclonal antibody, to pomalidomide-dexamethasone (Pd) in patients with relapsed/refractory multiple myeloma. Here, we report the final OS analysis. This multicenter, randomized, open-label, phase III study included patients who had received and failed ≥2 previous therapies, including lenalidomide and a proteasome inhibitor. Between January 10, 2017, and February 2, 2018, 307 patients were randomized (1:1) to isatuximab-pomalidomide-dexamethasone (Isa-Pd; N=154) or Pd (N=153), stratified based on age (<75 vs. ≥75 years) and number of previous lines of therapy (2-3 vs. >3). At data cutoff for the final OS analysis after 220 OS events (January 27, 2022), median follow-up duration was 52.4 months. Median OS was 24.6 months (95% confidence interval [CI]: 20.3-31.3) with Isa-Pd and 17.7 months (95% CI: 14.4- 26.2) with Pd (hazard ratio=0.78; 95% CI: 0.59-1.02; 1-sided P=0.0319). Despite subsequent daratumumab use in the Pd group and its potential benefit on PFS in the first subsequent therapy line, median PFS2 was significantly longer with Isa-Pd versus Pd (17.5 vs. 12.9 months; log-rank 1-sided P=0.0091). In this analysis, Isa-Pd continued to be efficacious and well tolerated after follow-up of approximately 52 months, contributing to a clinically meaningful, 6.9-month improvement in median OS in patients with relapsed/refractory multiple myeloma.
- Published
- 2024
- Full Text
- View/download PDF
78. International Myeloma Working Group immunotherapy committee consensus guidelines and recommendations for optimal use of T-cell-engaging bispecific antibodies in multiple myeloma.
- Author
-
Rodriguez-Otero P, Usmani S, Cohen AD, van de Donk NWCJ, Leleu X, Gállego Pérez-Larraya J, Manier S, Nooka AK, Mateos MV, Einsele H, Minnema M, Cavo M, Derman BA, Puig N, Gay F, Ho PJ, Chng WJ, Kastritis E, Gahrton G, Weisel K, Nagarajan C, Schjesvold F, Mikhael J, Costa L, Raje NS, Zamagni E, Hájek R, Weinhold N, Yong K, Ye JC, Sidhana S, Merlini G, Martin T, Lin Y, Chari A, Popat R, and Kaufman JL
- Subjects
- Humans, Immunotherapy methods, Immunotherapy standards, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Agents, Immunological adverse effects, Antibodies, Bispecific therapeutic use, Multiple Myeloma immunology, Multiple Myeloma therapy, Multiple Myeloma drug therapy, Consensus, T-Lymphocytes immunology, T-Lymphocytes drug effects
- Abstract
Multiple myeloma remains an incurable disease, despite the development of numerous drug classes and combinations that have contributed to improved overall survival. Immunotherapies directed against cancer cell-surface antigens, such as chimeric antigen receptor (CAR) T-cell therapy and T-cell-redirecting bispecific antibodies, have recently received regulatory approvals and shown unprecedented efficacy. However, these immunotherapies have unique mechanisms of action and toxicities that are different to previous treatments for myeloma, so experiences from clinical trials and early access programmes are essential for providing specific recommendations for management of patients, especially as these agents become available across many parts of the world. Here, we provide expert consensus clinical practice guidelines for the use of bispecific antibodies for the treatment of myeloma. The International Myeloma Working Group is also involved in the collection of prospective real-time data of patients treated with such immunotherapies, with the aim of learning continuously and adapting clinical practices to optimise the management of patients receiving immunotherapies., Competing Interests: Declaration of interests PR-O reports personal fees derived from consulting or advisory board roles from Celgene-BMS, Janssen, Roche, AbbVie, Pfizer, GSK, Sanofi, and H3Biomedicine; steering committee membership from Celgene-BMS, Regeneron, and Janssen; speaker's bureau fess from Janssen, Celgene-BMS, GSK, Sanofi, and AbbVie; and a travel grant from Pfizer. SU reports grants and personal fees from AbbVie, Amgen, BMS, EdoPharma, Genentech, Gilead, GSK, Merck, Sanofi, and Seagen; speakers’ bureau fees, consulting and advisory board participation, and steering committee membership from Janssen; participation on advisory boards from Karyopharm Therapeutics, Oncopeptides, Secura Bio, SkylineDx, and Takeda; and grants from Moderna, TeneoBio, and Pharmacyclics, outside the submitted work. ADC reports personal fees and participation on advisory boards from GSK; personal fees from Bristol Myers Squibb, Janssen, AbbVie, Pfizer, iTeos, Ichnos, Arcellx, and Legend; personal fees and participation on advisory boards from Genentech/Roche; participation on advisory boards from Novartis, outside the submitted work; and has a patent licensed for Novartis. NWCJvdD reports personal fees from Janssen Pharmaceuticals, Amgen, Celgene, Novartis, Cellectis, Bristol Myers Squibb/Celgene, Sanofi, Takeda, Roche, Novartis, Bayer, Adaptive, Pfizer, AbbVie, and Servier, outside the submitted work. JGP-L reports personal fees from Janssen and GSK, outside the submitted work. MVM reports personal fees from Janssen, Celgene/Bristol Myers Squibb, Novartis, GSK, Sanofi, Amgen, Pfizer, AbbVie, and Regeneron, outside the submitted work. HE reports personal fees and research support from Bristol Myers Squibb/Celgene, Janssen, Sanofi, and GSK; and personal fees from Amgen, Takeda, and Novartis; outside the submitted work. MM reports personal fees from Janssen, Bristol Myers Squibb, WebMD global, GSK, and CDR-Life; and research funding from Siemens and BeiGene; outside the submitted work. BAD reports advisory board participation and consulting fees from COTA and Janssen; personal fees from Multiple Myeloma Research Foundation; honoraria for CME-related activities from Plexus Communications; research funding from Amgen and GSK; and involvement as an independent reviewer of a clinical trial for BMS; outside the submitted work. NP reports research funding and accomodation expenses from Amgen, Bristol Myers Squibb, Janssen, Takeda, and The Binding Site; research funding from Sanofi, and personal fees from Pfizer, outside the submitted work. PJH reports personal fees from Antengene, Gilead, iTeos Therapeutics, Janssen, Novartis, and Pfizer, outside the submitted work. W-JC reports personal fees from AbbVie, Amgen, Pfizer, Sanofi, Regeneron, GSK, and Novartis; and grants and personal fees from Bristol Myers Squibb, Janssen, and Novartis, outside the submitted work. GG reports personal fees from and is a shareholder of XNK Therapeutics Sweden, and personal fees from Fujimoto Pharmaceutical corporation, outside the submitted work. FS reports personal fees from AbbVie, GSK, Celgene, Takeda, Janssen, Oncopeptides, Sanofi, BMS, Novartis, SkyliteDX, Pfizer, and Daiki-Sankyo, outside the submitted work. JCY reports personal fees from Janssen, Sanofi, BMS, Regeneron, GSK, Pfizer, Menarini, outside the submitted work. EZ reports personal fees from Janssen, Bristol Myers Squibb, Amgen, and Takeda, outside the submitted work. RP reports personal fees and travel support from GSK and Janssen; and personal fees from Pfizer, AbbVie, Bristol Myers Squibb, and Sanofi, outside the submitted work. CN reports personal fees and participation on advisory boards from Janssen; personal fees from Bristol Myers Squibb, Sanofi, GSK, Pfizer, AstraZeneca, and DKSH; and personal fees and participation on advisory boards from Amgen, outside the submitted work. YL reports grants and personal fees from Janssen; personal fees from Sanofi, NexImmune, Caribou, Bristol Myers Squibb, Regeneron, and Genentech; and personal fees and participation on data safety monitoring boards from Pfizer, outside the submitted work. AC reports personal fees from AbbVie, Adaptive, Amgen, Antengene, Bristol Myers Squibb, Forus, Genetech/Roche, GSK, Janssen, Karyopharm, Millenium/Takeda, and Sanofi/Genzyme, outside the submitted work. JM reports personal fees from Amgen, Sanofi, Bristol Myers Squibb, Janssen, and Takeda, outside the submitted work. MC reports personal fees from Janssen, Celgene/Bristol Myers Squibb, GSK, Sanofi, Amgen, Menarini-Stemline, AbbVie, and Pfizer, outside the submitted work. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
79. In multiple myeloma, monthly treatment with zoledronic acid beyond two years offers sustained protection against progressive bone disease.
- Author
-
Lund T, Gundesen MT, Juul Vangsted A, Helleberg C, Haukås E, Silkjær T, Asmussen JT, Manuela Teodorescu E, Amdi Jensen B, Schmidt Slørdahl T, Nahi H, Waage A, Abildgaard N, and Schjesvold F
- Subjects
- Humans, Zoledronic Acid therapeutic use, Diphosphonates therapeutic use, Bone and Bones, Multiple Myeloma drug therapy, Bone Diseases drug therapy, Bone Diseases etiology, Bone Diseases prevention & control, Bone Neoplasms, Bone Density Conservation Agents therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
80. VRD versus VCD as induction therapy before autologous stem cell transplantation in multiple myeloma: a nationwide population-based study.
- Author
-
Nørgaard JN, Moore KLF, Slørdahl TS, Vik A, Tvedt THA, and Schjesvold F
- Subjects
- Humans, Induction Chemotherapy, Transplantation, Autologous, Bortezomib therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Stem Cell Transplantation, Multiple Myeloma drug therapy, Hematopoietic Stem Cell Transplantation
- Published
- 2024
- Full Text
- View/download PDF
81. Pomalidomide, Bortezomib, and Dexamethasone in Lenalidomide-Pretreated Multiple Myeloma: A Subanalysis of OPTIMISMM by Frailty and Bortezomib Dose Adjustment.
- Author
-
Oriol A, Dimopoulos M, Schjesvold F, Beksac M, Facon T, Dhanasiri S, Guo S, Mu Y, Hong K, Gentili C, Galli M, Yagci M, Larocca A, Richardson P, and Weisel K
- Subjects
- Humans, Bortezomib pharmacology, Bortezomib therapeutic use, Lenalidomide therapeutic use, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone therapeutic use, Multiple Myeloma, Frailty, Thalidomide analogs & derivatives
- Abstract
Introduction: A proportion of patients with multiple myeloma (MM) are older and/or have comorbidities, requiring dose adjustments. Data from OPTIMISMM (NCT01734928) supported the use of pomalidomide, bortezomib, and dexamethasone (PVd) for treating relapsed/refractory MM. This subanalysis of OPTIMISMM assessed outcome by frailty and/or bortezomib dose adjustment., Methods: Patient frailty (nonfrail vs. frail) was classified using age, Charlson Comorbidity Index, and Eastern Cooperative Oncology Group performance status. Data from patients requiring a bortezomib dose reduction, interruption, and/or withdrawal during PVd treatment were assessed., Results: Among 559 patients, 93 of 281 (33.1%) and 93 of 278 (33.5%) patients who received PVd and bortezomib and dexamethasone (Vd), respectively, were frail. Overall response rate (ORR) and median progression-free survival (PFS) were higher in nonfrail vs. frail with PVd treatment (ORR, 82.8% vs. 79.6%; PFS, 14.7 vs. 9.7 months); significantly higher than with Vd regardless of frailty. Grade ≥ 3 treatment-emergent adverse events (TEAEs) were higher with PVd vs. Vd, regardless of frailty. Discontinuations of PVd were lower in nonfrail vs. frail patients (19.2% vs. 30.1%); the median duration of treatment was similar (DoT; 8.8 vs. 8.9 months, respectively). Patients who received PVd with a bortezomib dose adjustment (n = 240) had a longer median DoT (9.3 vs. 4.5 months) and PFS (12.1 vs. 8.4 months) vs. those without., Conclusion: Frail patients treated with PVd demonstrated a higher ORR and a longer PFS and DoT vs. Vd, despite a higher frequency of grade ≥ 3 TEAEs leading to pomalidomide, bortezomib, and/or dexamethasone discontinuation. Therefore, PVd treatment may improve patient outcomes, regardless of frailty., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
82. Ixazomib, Lenalidomide, and Dexamethasone (IRD) Treatment with Cytogenetic Risk-Based Maintenance in Transplant-Eligible Myeloma: A Phase 2 Multicenter Study by the Nordic Myeloma Study Group.
- Author
-
Partanen A, Waage A, Peceliunas V, Schjesvold F, Anttila P, Säily M, Uttervall K, Putkonen M, Carlson K, Haukas E, Sankelo M, Szatkowski D, Hansson M, Marttila A, Svensson R, Axelsson P, Lauri B, Mikkola M, Karlsson C, Abelsson J, Ahlstrand E, Sikiö A, Klimkowska M, Matuzeviciene R, Fenstad MH, Ilveskero S, Pelliniemi TT, Nahi H, and Silvennoinen R
- Abstract
Scarce data exist on double maintenance in transplant-eligible high-risk (HR) newly diagnosed multiple myeloma (NDMM) patients. This prospective phase 2 study enrolled 120 transplant-eligible NDMM patients. The treatment consisted of four cycles of ixazomib-lenalidomide-dexamethasone (IRD) induction plus autologous stem cell transplantation followed by IRD consolidation and cytogenetic risk-based maintenance therapy with lenalidomide + ixazomib (IR) for HR patients and lenalidomide (R) alone for NHR patients. The main endpoint of the study was undetectable minimal residual disease (MRD) with sensitivity of <10
-5 by flow cytometry at any time, and other endpoints were progression-free survival (PFS) and overall survival (OS). We present the preplanned analysis after the last patient has been two years on maintenance. At any time during protocol treatment, 28% (34/120) had MRD < 10-5 at least once. At two years on maintenance, 66% of the patients in the HR group and 76% in the NHR group were progression-free ( p = 0.395) and 36% (43/120) were CR or better, of which 42% (18/43) had undetectable flow MRD <10-5 . Altogether 95% of the patients with sustained MRD <10-5 , 82% of the patients who turned MRD-positive, and 61% of those with positive MRD had no disease progression at two years on maintenance ( p < 0.001). To conclude, prolonged maintenance with all-oral ixazomib plus lenalidomide might improve PFS in HR patients.- Published
- 2024
- Full Text
- View/download PDF
83. Health-related quality of life and quality-adjusted progression free survival for carfilzomib and dexamethasone maintenance following salvage autologous stem-cell transplantation in patients with multiple myeloma: a randomized phase 2 trial by the Nordic Myeloma Study Group.
- Author
-
Nielsen LK, Schjesvold F, Möller S, Guldbrandsen N, Hansson M, Remes K, Peceliunas V, Abildgaard N, Gregersen H, and King MT
- Subjects
- Humans, Progression-Free Survival, Quality of Life, Transplantation, Autologous, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Hematopoietic Stem Cell Transplantation, Oligopeptides
- Abstract
Background: Decisions regarding maintenance therapy in patients with multiple myeloma should be based on both treatment efficacy and health-related quality of life (HRQL) consequences. In the CARFI trial, patients with first relapse of multiple myeloma underwent salvage autologous stem cell transplantation (salvage ASCT) before randomization to carfilzomib-dexamethasone maintenance therapy (Kd) or observation. The primary clinical endpoint was time to progression, which was extended by 8 months by Kd. The aim of this paper is to present the all HRQL endpoints of the CARFI trial including the HRQL effect of Kd maintenance therapy relative to observation. The primary HRQL endpoint was assessed by EORTC QLQ-C30 Summary score (QLQ-C30-sum) at 8 months follow-up. A key secondary HRQL endpoint was quality-adjusted progression-free-survival (QAPFS)., Methods: HRQL was assessed with EORTC QLQ-C30, EORTC QLQ-MY20 and FACT/GOG-Ntx at randomization and every second month during follow-up. HRQL data were analyzed with linear mixed effect models until 8 months follow-up. QAPFS per individual was calculated by multiplying progression-free survival (PFS) by two quality-adjustment metrics, the QLQ-C30-sum and EORTC Quality of Life Utility Measure-Core 10 dimensions (QLU-C10D). The QAPFS per treatment group was estimated with the Kaplan-Meier method. P < 0.05 was used for statistical significance, and a between-group minimal important difference of 10 points was interpreted as clinically relevant for the QLQ-C30-sum., Results: 168 patients were randomized. HRQL questionnaire compliance was 93%. For the QLQ-C30-sum, the difference of 4.62 points (95% confidence interval (CI) -8.9: -0.4, p = 0.032) was not clinically relevant. PFS was 19.3 months for the Kd maintenance group and 16.8 months for the observation group; difference = 2.5 months (95% CI 0.5; 4.5). QAPFS based on the QLQ-C30-sum for the Kd maintenance group was 18.0 months (95% CI 16.4; 19.6) and for the observation group 15.0 months (95% CI 13.5; 16.5); difference = 3.0 months (95% CI 0.8-5.3). QAPFS based on the QLU-C10D for the Kd maintenance group was 17.5 months (95% CI 15.9; 19.2) and 14.0 months (95% CI 12.4; 15.5) for the observation group; difference = 3.5 months (95% CI 1.1-5.9)., Conclusions: Kd maintenance therapy after salvage ASCT did not adversely affect overall HRQL, but adjustment for HRQL reduced the PFS compared to unadjusted PFS. PFS of maintenance therapy should be quality-adjusted to balance the benefits and HRQL impact., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
84. Daratumumab, Bortezomib, Lenalidomide, and Dexamethasone for Multiple Myeloma.
- Author
-
Sonneveld P, Dimopoulos MA, Boccadoro M, Quach H, Ho PJ, Beksac M, Hulin C, Antonioli E, Leleu X, Mangiacavalli S, Perrot A, Cavo M, Belotti A, Broijl A, Gay F, Mina R, Nijhof IS, van de Donk NWCJ, Katodritou E, Schjesvold F, Sureda Balari A, Rosiñol L, Delforge M, Roeloffzen W, Silzle T, Vangsted A, Einsele H, Spencer A, Hajek R, Jurczyszyn A, Lonergan S, Ahmadi T, Liu Y, Wang J, Vieyra D, van Brummelen EMJ, Vanquickelberghe V, Sitthi-Amorn A, de Boer CJ, Carson R, Rodriguez-Otero P, Bladé J, and Moreau P
- Subjects
- Humans, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Bortezomib administration & dosage, Bortezomib adverse effects, Dexamethasone administration & dosage, Dexamethasone adverse effects, Disease Progression, Lenalidomide administration & dosage, Lenalidomide adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Multiple Myeloma drug therapy
- Abstract
Background: Daratumumab, a monoclonal antibody targeting CD38, has been approved for use with standard myeloma regimens. An evaluation of subcutaneous daratumumab combined with bortezomib, lenalidomide, and dexamethasone (VRd) for the treatment of transplantation-eligible patients with newly diagnosed multiple myeloma is needed., Methods: In this phase 3 trial, we randomly assigned 709 transplantation-eligible patients with newly diagnosed multiple myeloma to receive either subcutaneous daratumumab combined with VRd induction and consolidation therapy and with lenalidomide maintenance therapy (D-VRd group) or VRd induction and consolidation therapy and lenalidomide maintenance therapy alone (VRd group). The primary end point was progression-free survival. Key secondary end points were a complete response or better and minimal residual disease (MRD)-negative status., Results: At a median follow-up of 47.5 months, the risk of disease progression or death in the D-VRd group was lower than the risk in the VRd group. The estimated percentage of patients with progression-free survival at 48 months was 84.3% in the D-VRd group and 67.7% in the VRd group (hazard ratio for disease progression or death, 0.42; 95% confidence interval, 0.30 to 0.59; P<0.001); the P value crossed the prespecified stopping boundary (P = 0.0126). The percentage of patients with a complete response or better was higher in the D-VRd group than in the VRd group (87.9% vs. 70.1%, P<0.001), as was the percentage of patients with MRD-negative status (75.2% vs. 47.5%, P<0.001). Death occurred in 34 patients in the D-VRd group and 44 patients in the VRd group. Grade 3 or 4 adverse events occurred in most patients in both groups; the most common were neutropenia (62.1% with D-VRd and 51.0% with VRd) and thrombocytopenia (29.1% and 17.3%, respectively). Serious adverse events occurred in 57.0% of the patients in the D-VRd group and 49.3% of those in the VRd group., Conclusions: The addition of subcutaneous daratumumab to VRd induction and consolidation therapy and to lenalidomide maintenance therapy conferred a significant benefit with respect to progression-free survival among transplantation-eligible patients with newly diagnosed multiple myeloma. (Funded by the European Myeloma Network in collaboration with Janssen Research and Development; PERSEUS ClinicalTrials.gov number, NCT03710603; EudraCT number, 2018-002992-16.)., (Copyright © 2023 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
85. A plain language summary of the PERSEUS study of daratumumab plus bortezomib, lenalidomide, and dexamethasone for treating newly diagnosed multiple myeloma.
- Author
-
Sonneveld P, Dimopoulos MA, Boccadoro M, Quach H, Ho PJ, Beksac M, Hulin C, Antonioli E, Leleu X, Mangiacavalli S, Perrot A, Cavo M, Belotti A, Broijl A, Gay F, Mina R, van de Donk NWCJ, Katodritou E, Schjesvold F, Balari AS, Rosiñol L, Delforge M, Roeloffzen W, Silzle T, Vangsted A, Einsele H, Spencer A, Hajek R, Jurczyszyn A, Lonergan S, Ahmadi T, Liu Y, Wang J, Vieyra D, van Brummelen EMJ, Vanquickelberghe V, Sitthi-Amorn A, de Boer CJ, Carson R, Rodriguez-Otero P, Bladé J, and Moreau P
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Treatment Outcome, Randomized Controlled Trials as Topic, Clinical Trials, Phase II as Topic, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib administration & dosage, Bortezomib therapeutic use, Dexamethasone administration & dosage, Dexamethasone therapeutic use, Lenalidomide administration & dosage, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Multiple Myeloma diagnosis
- Abstract
What Is This Summary About?: This summary describes the first analysis of the PERSEUS study, which looked at adults with multiple myeloma that had never been treated before, also called newly diagnosed multiple myeloma. Multiple myeloma is a type of cancer in the blood, specifically in plasma cells within the soft, spongy tissue in the center of most bones, called the bone marrow. Researchers wanted to see if adding daratumumab (D) to a standard treatment of three other medicines called VRd, which stands for bortezomib (V), lenalidomide (R), and dexamethasone (d), could stop the multiple myeloma from getting worse and help participants live longer without multiple myeloma.Half of the participants were assigned to the treatment plan with daratumumab; they received D-VRd during initial treatment phases (induction and consolidation), followed by daratumumab as well as lenalidomide (D-R) in the maintenance phase. The other half of participants received treatment without daratumumab; they received VRd induction and consolidation followed by lenalidomide alone (R) maintenance. In addition, all participants were able to receive an autologous stem cell transplant, a procedure used to further help reduce multiple myeloma., What Were the Results?: At the time of this analysis of PERSEUS, about 4 years after participants started the study, participants who received D-VRd treatment followed by D-R maintenance had a better response to treatment (as measured by specific markers of multiple myeloma) and were more likely to be alive and free from their multiple myeloma getting worse in comparison to participants who received VRd followed by R maintenance. Side effects (unwanted or undesirable effects of treatment) in both treatment groups were in line with the known side effects of daratumumab and VRd., What Do the Results Mean?: The results of the PERSEUS study showed that including daratumumab in D-VRd induction/consolidation and D-R maintenance was better for treating multiple myeloma than the current standard VRd treatment followed by R maintenance alone in adults with a new diagnosis of multiple myeloma who were also able to receive an autologous stem cell transplant. Of importance, there were no unexpected side effects in either group. Clinical Trial Registration: NCT02874742 (GRIFFIN) (ClinicalTrials.gov).
- Published
- 2024
- Full Text
- View/download PDF
86. Treatment of primary plasma cell leukaemia with carfilzomib and lenalidomide-based therapy (EMN12/HOVON-129): final analysis of a non-randomised, multicentre, phase 2 study.
- Author
-
van de Donk NWCJ, Minnema MC, van der Holt B, Schjesvold F, Wu KL, Broijl A, Roeloffzen WWH, Gadisseur A, Pietrantuono G, Pour L, van der Velden VHJ, Lund T, Offidani M, Grasso M, Giaccone L, Razawy W, Tacchetti P, Mancuso K, Silkjaer T, Caers J, Zweegman S, Hájek R, Benjamin R, Vangsted AJ, Boccadoro M, Gay F, Sonneveld P, and Musto P
- Abstract
Background: Primary plasma cell leukaemia is a rare and aggressive plasma cell disorder with a poor prognosis. The aim of the EMN12/HOVON-129 study was to improve the outcomes of patients with primary plasma cell leukaemia by incorporating carfilzomib and lenalidomide in induction, consolidation, and maintenance therapy., Methods: The EMN12/HOVON-129 study is a non-randomised, phase 2, multicentre study conducted at 19 academic centres and hospitals in seven European countries (Belgium, Czech Republic, Denmark, Italy, Norway, The Netherlands, and the UK) for previously untreated patients with primary plasma cell leukaemia aged 18 years or older. Inclusion criteria were newly diagnosed primary plasma cell leukaemia (defined as >2 ×10
9 cells per L circulating monoclonal plasma cells or plasmacytosis >20% of the differential white cell count) and WHO performance status 0-3. Patients aged 18-65 years (younger patients) and 66 years or older (older patients) were treated in age-specific cohorts and were analysed separately. Younger patients were treated with four 28-day cycles of carfilzomib (36 mg/m2 intravenously on days 1, 2, 8, 9, 15, and 16), lenalidomide (25 mg orally on days 1-21), and dexamethasone (20 mg orally on days 1, 2, 8, 9, 15, 16, 22, and 23). Carfilzomib-lenalidomide-dexamethasone (KRd) induction was followed by double autologous haematopoietic stem-cell transplantation (HSCT), four cycles of KRd consolidation, and then maintenance with carfilzomib (27 mg/m2 intravenously on days 1, 2, 15, and 16 for the first 12 28-day cycles, and then 56 mg/m2 on days 1 and 15 in all subsequent cycles) and lenalidomide (10 mg orally on days 1-21) until progression. Patients who were eligible for allogeneic HSCT, could also receive a single autologous HSCT followed by reduced-intensity conditioning allogeneic HSCT and then carfilzomib-lenalidomide maintenance. Older patients received eight cycles of KRd induction followed by maintenance therapy with carfilzomib and lenalidomide until progression. The primary endpoint was progression-free survival. The primary analysis population was the intention-to-treat population, irrespective of the actual treatment received. Data from all participants who received any study drug were included in the safety analyses. The trial was registered at www.trialregister.nl (until June 2022) and https://trialsearch.who.int/ as NTR5350; recruitment is complete and this is the final analysis., Findings: Between Oct 23, 2015, and Aug 5, 2021, 61 patients were enrolled and received KRd induction treatment (36 patients aged 18-65 years [20 (56%) were male and 16 (44%) female], and 25 aged ≥66 years [12 (48%) were male and 13 (52%) female]). With a median follow-up of 43·5 months (IQR 27·7-67·8), the median progression-free survival was 15·5 months (95% CI 9·4-38·4) for younger patients. For older patients, median follow-up was 32·0 months (IQR 24·7-34·6), and median progression-free survival was 13·8 months (95% CI 9·2-35·5). Adverse events were most frequently observed directly after treatment initiation, with infections (two of 36 (6%) younger patients and eight of 25 (32%) older patients) and respiratory events (two of 36 [6%] younger patients and four of 25 [16%] older patients) being the most common grade 3 or greater events during the first four KRd cycles. Treatment-related serious adverse events were reported in 26 (72%) of 36 younger patients and in 19 (76%) of 25 older patients, with infections being the most common. Treatment-related deaths were reported in none of the younger patients and three (12%) of the older patients (two infections and one unknown cause of death)., Interpretation: Carfilzomib and lenalidomide-based therapy provides improved progression-free survival compared with previously published data. However, results remain inferior in primary plasma cell leukaemia compared with multiple myeloma, highlighting the need for new studies incorporating novel immunotherapies., Funding: Dutch Cancer Society, Celgene (a BMS company), and AMGEN., Competing Interests: Declaration of interests NWCJvdD reports research support from Janssen Pharmaceuticals, AMGEN, Celgene, Novartis, Cellectis and BMS, and advisory board role with Janssen Pharmaceuticals, AMGEN, Celgene, BMS, Takeda, Roche, Novartis, Bayer, Pfizer, Abbvie, Adaptive, and Servier, all paid to employer. MCM reports research support from Beigene; consultancy or advisory roles with Janssen, GSK, and CDR-life; honoraria from BMS, Pfizer, Janssen, and Medscape; and data safety monitoring board role for BMS, all paid to employer. BvdH reports honoraria for data safety monitoring board membership from the Intergroupe Francophone du Myélome. FS reports research support from Celgene, Janssen, Oncopeptides, Sanofi, GSK, and Targovax; honoraria from Amgen, BMS, Takeda, Abbvie, Janssen, Novartis, SkyliteDX, Oncopeptides, Sanofi, Pfizer, Daiki-Sankyo, and GSK; and participated on a data safety monitoring board or advisory board for Abbvie, GSK, Celgene, Takeda, Janssen, Oncopeptides, Sanofi, and BMS. KLW has served in advisory boards for Janssen and Pfizer. AB received honoraria and served in advisory boards from BMS, Janssen, Amgen, and Sanofi. WWHR reports honoraria from Amgen and SANOFI, all paid to employer; support for travel from Takeda and Abbvie; and advisory board participations for BMS/Celgene, and Janssen, all paid to employer. VHJvdV reports a Laboratory Services Agreement with BD Biosciences, Cytognos, and Agilent, all paid to employer. TL has received research grants from Takeda, AMGEN, and Janssen, honoraria from AMGEN, Janssen, BMS, and Pfizer; traveling support from Pfizer and Janssen; and equipment from Takeda and Pfizer. MO has received honoraria and served in advisory board for AbbVie, Amgen, BMS, GSK, Janssen, Sanofi, and Takeda. PT has received honoraria from Janssen, BMS/Celgene, GlaxoSmithKline, Takeda, Amgen, Sanofi, Pfizer, and AbbVie and support for traveling from Janssen, BMS/Celgene, Sanofi, and AMGEN. KM has received honoraria from GSK, Janssen, Pfizer, and Sanofi; support for traveling from Sanofi; and participates on an advisory board for Pfizer. JC has received research funding from Janssen, honoraria from AMGEN and Janssen, and support for travel from GILEAD. SZ has received research funding from Takeda and Janssen and serves in advisory boards for Janssen, Takeda, and BMS, all paid to institution. RH has received research funding from Janssen, AMGEN, Celgene/BMS, Novartis, and Takeda, all paid to employer; consulting fees from Janssen, AMGEN, Celgene, Abbvie, BMS, Novartis, Pharmamar, and Takeda; honoraria from Janssen, AMGEN, Celgene, BMS, Pharmamar, and Takeda; support for attending meetings from AMGEN, Celgene, Takeda, and Janssen; and has participated on advisory boards from BMS, Takeda, AMGEN, Oncopeptides, Sanofi, Janssen, and GSK. RB reports educational grant from BMS/Celgene. AJV reports honoraria from BMS. MB reports research funding from Sanofi, Celgene, Amgen, Janssen, Novartis, BMS, and Mundipharma; honoraria from Sanofi, Celgene, Amgen, Janssen, Novartis, BMS, and Abbvie; and advisory board role for Janssen. FG reports honoraria from Amgen, Celgene, Janssen, Takeda, Bristol Myers Squibb, AbbVie, GlaxoSmithKline, data safety monitoring board or advisory board participation for Amgen, Celgene, Janssen, Takeda, Bristol Myers Squibb, AbbVie, GlaxoSmithKline, Roche, Adaptive Biotechnologies, Oncopeptides, bluebird bio, and Pfizer. PS reports advisory board roles with Amgen, BMS, Celgene, Janssen, Karyopharm, and Pfizer and research funding from Amgen, BMS, Celgene, Janssen, Karyopharm, and Pfizer. PM reports honoraria from or Advisory Board role with AMGEN and BMS. All other authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
87. Intensifying treatment in PET-positive multiple myeloma patients after upfront autologous stem cell transplantation.
- Author
-
Nørgaard JN, Abildgaard N, Lysén A, Tsykunova G, Vangsted AJ, João C, Remen N, Nielsen LK, Osnes L, Stokke C, Connelly JP, Revheim MR, and Schjesvold F
- Subjects
- Humans, Treatment Outcome, Transplantation, Autologous, Neoplasm, Residual diagnosis, Positron-Emission Tomography, Dexamethasone therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Stem Cell Transplantation, Multiple Myeloma diagnostic imaging, Multiple Myeloma therapy, Hematopoietic Stem Cell Transplantation methods
- Abstract
18 F-Fluorodeoxyglucose positron emission tomography/computed tomography (PET) positivity after first-line treatment with autologous stem cell transplantation (ASCT) in multiple myeloma is strongly correlated with reduced progression-free and overall survival. However, PET-positive patients who achieve PET negativity after treatment seem to have comparable outcomes to patients who were PET negative at diagnosis. Hence, giving PET-positive patients additional treatment may improve their outcome. In this phase II study, we screened first-line patients with very good partial response (VGPR) or better after ASCT with PET. PET-positive patients received four 28-day cycles of carfilzomib-lenalidomide-dexamethasone (KRd). Flow cytometry-based minimal residual disease (MRD) analysis was performed before and after treatment for correlation with PET. Overall, 159 patients were screened with PET. A total of 53 patients (33%) were PET positive and 57% of PET-positive patients were MRD negative, demonstrating that these response assessments are complementary. KRd consolidation converted 33% of PET-positive patients into PET negativity. MRD-negative patients were more likely to convert than MRD-positive patients. In summary, PET after ASCT detected residual disease in a substantial proportion of patients in VGPR or better, even in patients who were MRD negative, and KRd consolidation treatment changed PET status in 33% of patients., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
88. Difficult-to-treat patients with relapsed/refractory multiple myeloma: A review of clinical trial results.
- Author
-
Raab MS, Zamagni E, Manier S, Rodriguez-Otero P, Schjesvold F, and Broijl A
- Abstract
Overall outcomes for multiple myeloma have improved due to the availability of new therapies, but patients with relapsed/refractory multiple myeloma harbouring certain factors continue to pose a therapeutic challenge. These challenging features include high-risk cytogenetics, renal impairment, patient characteristics such as age and frailty, and extramedullary disease. Prior refractory status and number of prior lines add further complexity to the treatment of these patients. While newer regimens are available and have suggested efficacy in these patient populations through subgroup analyses, differences in trial definitions and cut-offs make meaningful comparisons difficult. This review aims to examine the available clinical trial data for patients with high-risk cytogenetics, renal impairment, age and frailty and extramedullary disease., Competing Interests: M‐SR: honoraria – BMS, Janssen; advisory role – Amgen, BMS, Janssen, Novartis, Sanofi, Takeda; consultant – Amgen, BMS, Janssen, Novartis, Sanofi, Takeda; research support – BMS, Janssen, Novartis, Sanofi. EZ: honoraria – Amgen, BMS, GSK, Janssen, Pfizer, Roche, Sanofi, Takeda; advisory role – Amgen, BMS, GSK, Janssen, Pfizer, Roche, Sanofi, Takeda. SM: advisory role – AbbVie, Adaptive Biotechnology, Amgen, Celgene/BMS, GlaxoSmithKline, Janssen, Novartis, Oncopeptide, Regeneron, Roche, Sanofi, Takeda; research support – AbbVie, Adaptive Biotechnology, Amgen, Celgene/BMS, GlaxoSmithKline, Janssen, Novartis, Oncopeptide, Regeneron, Roche, Sanofi, Takeda. PR‐O: honoraria – Amgen, BMS, GSK, Janssen, Regeneron, Sanofi, Takeda; advisory role – Abbvie, BMS, GSK, Janssen, Kite Pharma, Oncopeptides, Pfizer, Sanofi, Takeda; consultant – Celgene‐BMS, GSK, Pfizer. FS: honoraria – Abbvie, Amgen, BMS, Daiichi‐Sankyo, GSK, Janssen, Novartis, Oncopeptides, Pfizer, Sanofi, SkyliteDX, Takeda; advisory role – Abbvie, Celgene, GSK, Janssen, Oncopeptides, Sanofi, Takeda; research support – Celgene, GSK, Janssen, Oncopeptides, Sanofi, Targovax. AB: honoraria – Amgen, BMS, Janssen, Sanofi, Takeda; advisory role – Amgen, BMS, Janssen, Sanofi, Takeda., (© 2023 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
89. Potential value of pre-planned imaging of bone disease in multiple myeloma.
- Author
-
Gundesen MT, Asmussen JT, Schjesvold F, Vangsted AJ, Helleberg C, Haukås E, Silkjær T, Teodorescu EM, Jensen BA, Slørdahl TS, Nahi H, Waage A, Abildgaard N, and Lund T
- Subjects
- Humans, Diagnostic Imaging, Bone Marrow, Multiple Myeloma complications, Multiple Myeloma diagnostic imaging, Bone Diseases diagnostic imaging, Bone Diseases etiology
- Published
- 2023
- Full Text
- View/download PDF
90. Prevention and management of adverse events during treatment with bispecific antibodies and CAR T cells in multiple myeloma: a consensus report of the European Myeloma Network.
- Author
-
Ludwig H, Terpos E, van de Donk N, Mateos MV, Moreau P, Dimopoulos MA, Delforge M, Rodriguez-Otero P, San-Miguel J, Yong K, Gay F, Einsele H, Mina R, Caers J, Driessen C, Musto P, Zweegman S, Engelhardt M, Cook G, Weisel K, Broijl A, Beksac M, Bila J, Schjesvold F, Cavo M, Hajek R, Touzeau C, Boccadoro M, and Sonneveld P
- Subjects
- Humans, Cytokine Release Syndrome etiology, Cytokine Release Syndrome prevention & control, Cytokine Release Syndrome drug therapy, Interleukin 1 Receptor Antagonist Protein therapeutic use, Consensus, Immunotherapy, Adoptive adverse effects, T-Lymphocytes, Multiple Myeloma drug therapy, Antibodies, Bispecific adverse effects
- Abstract
T-cell redirecting bispecific antibodies (BsAbs) and chimeric antigen receptor T cells (CAR T cells) have revolutionised multiple myeloma therapy, but adverse events such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome (ICANS), cytopenias, hypogammaglobulinaemia, and infections are common. This Policy Review presents a consensus from the European Myeloma Network on the prevention and management of these adverse events. Recommended measures include premedication, frequent assessing for symptoms and severity of cytokine release syndrome, step-up dosing for several BsAbs and some CAR T-cell therapies; corticosteroids; and tocilizumab in the case of cytokine release syndrome. Other anti-IL-6 drugs, high-dose corticosteroids, and anakinra might be considered in refractory cases. ICANS often arises concomitantly with cytokine release syndrome. Glucocorticosteroids in increasing doses are recommended if needed, as well as anakinra if the response is inadequate, and anticonvulsants if convulsions occur. Preventive measures against infections include antiviral and antibacterial drugs and administration of immunoglobulins. Treatment of infections and other complications is also addressed., Competing Interests: Declaration of interests HL reports honoraria for lectures and for advisory boards from Celgene-BMS, Janssen-Cilag, Takeda, Amgen, Sanofi, AbbVie, and Pfizer; and research funding from Amgen and Sanofi. ET reports grants from Amgen, Janssen, GSK, Takeda, and Sanofi; payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Amgen, ASTRA, BMS, GSK, Integris, Pfizer, Sanofi, and Takeda; support for attending meetings or travel from Amgen, Takeda, and Eusa Pharma; and participation on a data safety monitoring board or advisory board from BMS, GSK, Sanofi, Takeda, Eusa Pharma, Janssen, ASTRA, and Amgen. NvdD reports research support from Janssen, Amgen, Celgene, Novartis, Cellectis, and BMS; and participation on a data safety monitoring board or advisory board from Janssen, Amgen, Celgene, BMS, Takeda, Roche, Novartis, and Adaptive. M-VM reports honoraria derived from lectures and participation in advisory boards from Janssen, BMS-Celgene, Takeda, Amgen, GSK, AbbVie, Pfizer, Regeneron, Roche, Sanofi, and Oncopeptides. PMo reports honoraria for advisory boards and symposia from Celgene, Janssen, Pfizer, and Abbvie; and payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Janssen, Celgene, Takeda, Amgen, AbbVie, Sanofi, and GSK. M-AD reports participation in advisory boards for Amgen, Takeda, BMS, Janssen, Beigene, and Sanofi. MD reports grants from Janssen and BMS; consulting fees from Amgen, BMS, Janssen, Sanofi, Stemline, and Takeda; payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from BMS and Janssen; and participation on a data safety monitoring board or advisory board for BMS and Takeda. PR-O reports honoraria for lectures from BMS, Janssen, Sanofi, GSK, Amgen, Regeneron, and Takeda; and participation in advisory boards for BMS, Janssen, Sanofi, Kite Pharma, AbbVie, Oncopeptides, Takeda, Pfizer, and GSK. JS-M reports consulting fees from Takeda, Janssen, Celgene, BMS, Amgen, Sanofi, and GSK; honoraria from Takeda, Janssen, Celgene, Amgen, BMS, Sanofi, and GSK; and participation in scientific advisory boards for Takeda, Janssen, Celgene, Novartis, Amgen, BMS, Sanofi, GSK Roche, and Regeneron. KY reports consulting fees from Sanofi, and Janssen; speaker bureaus from Sanofi, Amgen, Takeda, and Pfizer; honoraria from Sanofi, Janssen, Amgen, Takeda, and Pfizer; and research funding from Sanofi, Janssen, Amgen, Takeda, and BMS. FG reports payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Janssen, Amgen, Sanofi, BMS, Takeda, GSK, Roche, and AbbVie; and participation on a data safety monitoring board or advisory board from Janssen, Amgen, Sanofi, BMS, Takeda, GSK, Roche, AbbVie, Oncopeptides, Pfizer, and Adaptive. HE reports consulting fees, payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events; and support for attending meetings or travel from Janssen, BMS–Celgene, Amgen, GSK, Sanofi, Takeda, and Novartis. RM reports consulting fees from Janssen, Takeda, and Sanofi; payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Janssen, Celgene, Takeda, Amgen; and participation on an advisory board for Janssen, Celgene, Takeda, BMS, and Amgen. JC reports grants from Janssen; consulting fees from Janssen and Sanofi; payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Amgen; support for attending meetings or travel from Gilead; and a patent planned, issued, or pending: 20175032.0: anti-CD38 single-domain antibodies in disease monitoring and treatment. ME reports payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Janssen, BMS, Takeda, Amgen, GSK, and Sanofi; and support for attending meetings or travel from Janssen, BMS, Takeda, and Amgen. GC reports research funding from Takeda and BMS; consulting fees from Takeda, BMS, AbbVie, Roche, Pfizer, Janssen, Karyopharm, and Amgen; and speaker bureau for Janssen, Takeda, AbbVie, and Amgen. KW reports honoraria and participation on advisory boards from AbbVie, Adaptive, Amgen, Bristol Myers Squibb, Celgene, Janssen, GSK, Karyopharm, Novartis, Oncopeptides, Pfizer, Roche Pharma, Takeda, and Sanofi; and reports research funding from Amgen, Celgene, Janssen, and Sanofi. AB reports payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from BMS, Sanofi, Janssen, and Amgen. MBe reports participation on advisory boards for Amgen, Bristol-Myers Squibb, Janssen, Sanofi, Takeda, and Oncopeptides. JB reports payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Janssen, Takeda, and Amgen; payment for expert testimony from Janssen and Amgen; support for attending meetings or travel from Janssen and Takeda; and a leadership or fiduciary role in other board, society, committee, or advocacy group, paid or unpaid, in the Serbian Myeloma Group and the Balkan Myeloma Study Group. FS reports grants or contracts from Celgene, Janssen, Oncopeptides, Sanofi, GSK, and Targovax; payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Amgen, BMS, Takeda, AbbVie, Janssen, Novartis, SkyliteDX, Oncopeptides, Sanofi, Pfizer, Daiki-Sankyo, and GSK; and participation on a data safety monitoring board or advisory board for AbbVie, GSK, Celgene, Takeda, Janssen, Oncopeptides, Sanofi, and BMS. MC reports consulting fees from Glaxo Smith Kline, Janssen, Sanofi, Roche, Amgen, Takeda, AbbVie, Bristol Myers Squibb, Celgene, and Pfizer; payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Janssen, Sanofi, and Bristol Myers Squibb; and participation on a data safety monitoring board or advisory board from Glaxo Smith Kline, Janssen, Sanofi, Roche, Amgen, Takeda, AbbVie, Bristol Myers Squibb, Celgene, and Pfizer. RH reports grants or contracts paid to institution from Janssen, Amgen, Celgene, BMS, Novartis, and Takeda; consulting fees from Janssen, Amgen, Celgene, AbbVie, BMS, Novartis, PharmaMar, and Takeda; payment or honoraria for lectures, presentations, speaker bureaus, manuscript writing, or educational events from Janssen, Amgen, Celgene, BMS, PharmaMar, and Takeda; support for attending meetings or travel from Amgen, Celgene, Takeda, and Janssen; and participation in a data safety monitoring board or advisory board for BMS, Takeda, Amgen, Oncopeptides, Sanofi, Janssen, and GSK. CT reports consulting fees from AbbVie, Janssen, and Pfizer; payment for lectures from Janssen and BMS; support for attending meetings or travel from Janssen; and participation in advisory boards for Janssen, Pfizer, and BMS. MBo reports honoraria from Sanofi, Celgene, Amgen, Janssen, Novartis, Bristol Myers Squibb, and AbbVie; participation in an advisory board for Janssen and GSK; and research funding from Sanofi, Celgene, Amgen, Janssen, Novartis, BMS, and Mundipharma. PS reports participation in an advisory board for and research funding from Amgen, BMS, Celgene, Janssen, Karyopharm, and Pfizer. All other authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
91. Management of patients with multiple myeloma and COVID-19 in the post pandemic era: a consensus paper from the European Myeloma Network (EMN).
- Author
-
Terpos E, Musto P, Engelhardt M, Delforge M, Cook G, Gay F, van de Donk NWCJ, Ntanasis-Stathopoulos I, Vangsted AJ, Driessen C, Schjesvold F, Cerchione C, Zweegman S, Hajek R, Moreau P, Einsele H, San-Miguel J, Boccadoro M, Dimopoulos MA, Sonneveld P, and Ludwig H
- Subjects
- Humans, SARS-CoV-2, COVID-19 Serotherapy, Consensus, Pandemics, Antibodies, Neutralizing, COVID-19 epidemiology, Multiple Myeloma therapy
- Abstract
In the post-pandemic COVID-19 period, human activities have returned to normal and COVID-19 cases are usually mild. However, patients with multiple myeloma (MM) present an increased risk for breakthrough infections and severe COVID-19 outcomes, including hospitalization and death. The European Myeloma Network has provided an expert consensus to guide patient management in this era. Vaccination with variant-specific booster vaccines, such as the bivalent vaccine for the ancestral Wuhan strain and the Omicron BA.4/5 strains, is essential as novel strains emerge and become dominant in the community. Boosters should be administered every 6-12 months after the last vaccine shot or documented COVID-19 infection (hybrid immunity). Booster shots seem to overcome the negative effect of anti-CD38 monoclonal antibodies on humoral responses; however, anti-BCMA treatment remains an adverse predictive factor for humoral immune response. Evaluation of the immune response after vaccination may identify a particularly vulnerable subset of patients who may need additional boosters, prophylactic therapies and prevention measures. Pre-exposure prophylaxis with tixagevimab/cilgavimab is not effective against the new dominant variants and thus is no longer recommended. Oral antivirals (nirmatrelvir/ritonavir and molnupiravir) and remdesivir are effective against Omicron subvariants BA.2.12.1, BA.4, BA.5, BQ.1.1 and/or XBB.1.5 and should be administered in MM patients at the time of a positive COVID-19 test or within 5 days post symptoms onset. Convalescent plasma seems to have low value in the post-pandemic era. Prevention measures during SARS-CoV-2 outbreaks, including mask wearing and avoiding crowded places, seem prudent to continue for MM patients., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
92. Preclinical discovery and initial clinical data of WVT078, a BCMA × CD3 bispecific antibody.
- Author
-
Raab MS, Cohen YC, Schjesvold F, Aardalen K, Oka A, Spencer A, Wermke M, Souza AD, Kaufman JL, Cafro AM, Ocio EM, Doki N, Henson K, Trabucco G, Carrion A, Bender FC, Juif PE, Fessehatsion A, Fan L, Stonehouse JP, Blankenship JW, Granda B, De Vita S, and Lu H
- Subjects
- Animals, Humans, Macaca fascicularis metabolism, B-Cell Maturation Antigen, Immunotherapy, Adoptive, Multiple Myeloma pathology, Immunoconjugates therapeutic use, Antibodies, Bispecific therapeutic use
- Abstract
B-cell maturation antigen (BCMA) is an ideal target in multiple myeloma (MM) due to highly specific expression in malignant plasma cells. BCMA-directed therapies including antibody drug conjugates, chimeric antigen receptor-T cells and bispecific antibodies (BsAbs) have shown high response rates in MM. WVT078 is an anti-BCMA× anti-CD3 BsAb that binds to BCMA with subnanomolar-affinity. It was selected based on potent T cell activation and anti-MM activity in preclinical models with favorable tolerability in cynomolgus monkey. In the ongoing first-in-human phase I dose-escalation study (NCT04123418), 33 patients received intravenous WVT078 once weekly at escalated dosing. At the active doses of 48-250 µg/kg tested to date (n = 26), the overall response rate (ORR) was 38.5% (90% CI: 22.6-56.4%) and the complete response rate (CRR, stringent complete response + complete response) was 11.5%, (90% CI: 3.2-27.2%). At the highest dose level tested, the ORR was 75% (3 of 4 patients). 26 (78.8%) patients reported at least one Grade ≥3 AE and 16 of these AEs were suspected to be drug related. 20 patients (60.6%) experienced cytokine release syndrome. WVT078 has an acceptable safety profile and shows preliminary evidence of clinical activity at doses tested to date., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
93. Phenotypic deconvolution in heterogeneous cancer cell populations using drug-screening data.
- Author
-
Köhn-Luque A, Myklebust EM, Tadele DS, Giliberto M, Schmiester L, Noory J, Harivel E, Arsenteva P, Mumenthaler SM, Schjesvold F, Taskén K, Enserink JM, Leder K, Frigessi A, and Foo J
- Subjects
- Humans, Early Detection of Cancer, Cell Line, Genomics, Neoplasms drug therapy, Antineoplastic Agents pharmacology
- Abstract
Tumor heterogeneity is an important driver of treatment failure in cancer since therapies often select for drug-tolerant or drug-resistant cellular subpopulations that drive tumor growth and recurrence. Profiling the drug-response heterogeneity of tumor samples using traditional genomic deconvolution methods has yielded limited results, due in part to the imperfect mapping between genomic variation and functional characteristics. Here, we leverage mechanistic population modeling to develop a statistical framework for profiling phenotypic heterogeneity from standard drug-screen data on bulk tumor samples. This method, called PhenoPop, reliably identifies tumor subpopulations exhibiting differential drug responses and estimates their drug sensitivities and frequencies within the bulk population. We apply PhenoPop to synthetically generated cell populations, mixed cell-line experiments, and multiple myeloma patient samples and demonstrate how it can provide individualized predictions of tumor growth under candidate therapies. This methodology can also be applied to deconvolution problems in a variety of biological settings beyond cancer drug response., Competing Interests: The authors declare no competing interests., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
94. Phase I Study of Safety and Pharmacokinetics of RO7297089, an Anti-BCMA/CD16a Bispecific Antibody, in Patients with Relapsed, Refractory Multiple Myeloma.
- Author
-
Plesner T, Harrison SJ, Quach H, Lee C, Bryant A, Vangsted A, Estell J, Delforge M, Offner F, Twomey P, Choeurng V, Li J, Hendricks R, Ruppert SM, Sumiyoshi T, Miller K, Cho E, and Schjesvold F
- Abstract
Introduction: This phase 1 trial assessed the safety, pharmacokinetics, and preliminary antitumor activity of RO7297089, an anti-BCMA/CD16a bispecific antibody., Methods: RO7297089 was administered weekly by intravenous infusion to patients with relapsed/refractory multiple myeloma. The starting dose was 60 mg in this dose-escalation study utilizing a modified continual reassessment method with overdose control model., Results: Overall, 27 patients were treated at doses between 60 and 1850 mg. The maximally administered dose was 1850 mg due to excipients in the formulation that did not allow for higher doses to be used. The maximum tolerated dose was not reached. The most common adverse events irrespective of grade and relationship to the drug were anemia, infusion-related reaction, and thrombocytopenia. Most common treatment-related grade ≥ 3 toxicities were ALT/AST increase and reduced lymphocyte count. Pharmacokinetic studies suggested non-linear pharmacokinetics and target-mediated drug disposition, with a trend of approaching linear pharmacokinetics at doses of 1080 mg and higher. Partial response was observed in two patients (7%), minimal response in two patients (7%), and stable disease in 14 patients (52%)., Conclusions: RO7297089 was well tolerated at doses up to 1850 mg, and the efficacy data supported activity of RO7297089 in multiple myeloma. Combination with other agents may further enhance its potential as an innate immune cell engager in multiple myeloma., Trial Registration: ClinicalTrials.gov: NCT04434469; Registered June 16, 2020; https://www., Clinicaltrials: gov/ct2/show/NCT04434469 ., (© 2023. Genentech, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
95. Cobimetinib Alone and Plus Venetoclax With/Without Atezolizumab in Patients With Relapsed/Refractory Multiple Myeloma.
- Author
-
Schjesvold F, Paiva B, Ribrag V, Rodriguez-Otero P, San-Miguel JF, Robak P, Hansson M, Onishi M, Hamidi H, Malhi V, Dail M, Javery A, Ku G, and Raab MS
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Proto-Oncogene Proteins c-bcl-2, Multiple Myeloma drug therapy, Multiple Myeloma etiology
- Abstract
Introduction: Mitogen-activated protein kinase pathway mutations are present in >50% of patients with relapsed/refractory (R/R) multiple myeloma (MM). MEK inhibitors show limited single-agent activity in R/R MM; combination with B-cell lymphoma-2 (BCL-2) and programmed death-ligand 1 inhibition may improve efficacy. This phase Ib/II trial (NCT03312530) evaluated safety and efficacy of cobimetinib (cobi) alone and in combination with venetoclax (ven) with/without atezolizumab (atezo) in patients with R/R MM., Patients and Methods: Forty-nine patients were randomized 1:2:2 to cobi 60 mg/day on days 1-21 (n = 6), cobi 40 mg/day on days 1-21 + ven 800 mg/day on days 1-28 with/without atezo 840 mg on days 1 and 15 of 28-day cycles (cobi-ven, n = 22; cobi-ven-atezo, n = 21). Safety run-in cohorts evaluated cobi-ven and cobi-ven-atezo dose levels., Results: Any-grade common adverse events (AEs) with cobi, cobi-ven, and cobi-ven-atezo, respectively, included diarrhea (33.3%, 81.8%, 90.5%) and nausea (16.7%, 50.0%, 66.7%); common grade ≥3 AEs included anemia (0%, 22.7%, 23.8%), neutropenia (0%, 13.6%, 38.1%), and thrombocytopenia (0%, 18.2%, 23.8%). The overall response rate for all-comers was 0% (cobi), 27.3% (cobi-ven), and 28.6% (cobi-ven-atezo), and 0%, 50.0%, and 100%, respectively, in patients with t(11;14)+. Biomarker analysis demonstrated non-t(11;14) patient selection with NRAS/KRAS/BRAF mutation or high BCL-2/BCL-2-L1 ratio (>52% of the study population) could enrich for responders to the cobi-ven combination., Conclusions: Cobi-ven and cobi-ven-atezo demonstrated manageable safety with moderate activity in all-comers, and higher activity in patients with t(11;14)+ MM, supporting a biomarker-driven approach for ven in MM., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
96. What matters most to patients with multiple myeloma? A Pan-European patient preference study.
- Author
-
Janssens R, Lang T, Vallejo A, Galinsky J, Morgan K, Plate A, De Ronne C, Verschueren M, Schoefs E, Vanhellemont A, Delforge M, Schjesvold F, Cabezudo E, Vandebroek M, Stevens H, Simoens S, and Huys I
- Abstract
Introduction: Given the rapid increase in novel treatments for patients with multiple myeloma (MM), this patient preference study aimed to establish which treatment attributes matter most to MM patients and evaluate discrete choice experiment (DCE) and swing weighting (SW) as two elicitation methods for quantifying patients' preferences., Methods: A survey incorporating DCE and SW was disseminated among European MM patients. The survey included attributes and levels informed by a previous qualitative study with 24 MM patients. Latent class and mixed logit models were used to estimate the DCE attribute weights and descriptive analyses were performed to derive SW weights. MM patients and patient organisations provided extensive feedback during survey development., Results: 393 MM patients across 21 countries completed the survey ( M
years since diagnosis =6; Mprevious therapies =3). Significant differences (p<.01) between participants' attribute weights were revealed depending on participants' prior therapy experience, and their experience with side-effects and symptoms. Multivariate analyses showed that participants across the three MM patient classes identified via the latent class model differed regarding their past number of therapies ( F =4.772, p =.009). Patients with the most treatments (class 1) and those with the least treatments (class 3) attached more value to life expectancy versus quality of life-related attributes such as pain, mobility and thinking problems. Conversely, patients with intermediary treatment experience (class 2) attached more value to quality of life-related attributes versus life expectancy. Participants highlighted the difficulty of trading-off between life expectancy and quality of life and between physical and mental health. Participants expressed a need for greater psychological support to cope with their symptoms, treatment side-effects, and uncertainties. With respect to patients' preferences for the DCE or SW questions, 42% had no preference, 32% preferred DCE, and 25% preferred SW., Conclusions: Quality of life-related attributes affecting MM patients' physical, mental and psychological health such as pain, mobility and thinking problems were considered very important to MM patients, next to life expectancy. This underscores a need to include such attributes in decision-making by healthcare stakeholders involved in MM drug development, evidence generation, evaluation, and clinical practice. This study highlights DCE as the preferred methodology for understanding relative attribute weights from a patient's perspective., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Janssens, Lang, Vallejo, Galinsky, Morgan, Plate, De Ronne, Verschueren, Schoefs, Vanhellemont, Delforge, Schjesvold, Cabezudo, Vandebroek, Stevens, Simoens and Huys.)- Published
- 2022
- Full Text
- View/download PDF
97. Anti-CD38 antibody therapy for patients with relapsed/refractory multiple myeloma: differential mechanisms of action and recent clinical trial outcomes.
- Author
-
Leleu X, Martin T, Weisel K, Schjesvold F, Iida S, Malavasi F, Manier S, Chang-Ki Min, Ocio EM, Pawlyn C, Perrot A, Quach H, Richter J, Spicka I, Yong K, and Richardson PG
- Subjects
- ADP-ribosyl Cyclase 1, Clinical Trials as Topic, Humans, Tumor Microenvironment, Antineoplastic Agents, Immunological therapeutic use, Multiple Myeloma therapy
- Abstract
CD38 is a transmembrane glycoprotein that functions both as a receptor and an ectoenzyme, playing key roles in the regulation of calcium signaling and migration of immune cells to tumor microenvironments. High expression on multiple myeloma (MM) cells and limited expression on normal cells makes CD38 an ideal target for the treatment of MM patients. Two monoclonal antibodies directed at CD38, isatuximab and daratumumab, are available for use in patients with relapsed and/or refractory MM (RRMM); daratumumab is also approved in newly diagnosed MM and light-chain amyloidosis. Clinical experience has shown that anti-CD38 antibody therapy is transforming treatment of MM owing to its anti-myeloma efficacy and manageable safety profile. Isatuximab and daratumumab possess similarities and differences in their mechanisms of action, likely imparted by their binding to distinct, non-overlapping epitopes on the CD38 molecule. In this review, we present the mechanistic properties of these two antibodies and outline available evidence on their abilities to induce adaptive immune responses and modulate the bone marrow niche in MM. Further, we discuss differences in regulatory labeling between these two agents and analyze recent key clinical trial results, including evidence in patients with underlying renal impairment and other poor prognostic factors. Finally, we describe the limited existing evidence for the use of isatuximab or daratumumab after disease progression on prior anti-CD38 mono- or combination therapy, highlighting the need for additional clinical evaluations to define optimal anti-CD38 antibody therapy selection and sequencing in RRMM., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
98. Comparison of [ 18 F]fluciclovine and [ 18 F]FDG PET/CT in Newly Diagnosed Multiple Myeloma Patients.
- Author
-
Stokke C, Nørgaard JN, Feiring Phillips H, Sherwani A, Nuruddin S, Connelly J, Schjesvold F, and Revheim ME
- Subjects
- Humans, Pilot Projects, Positron Emission Tomography Computed Tomography methods, Prospective Studies, Radiopharmaceuticals, Fluorodeoxyglucose F18, Multiple Myeloma diagnostic imaging, Multiple Myeloma pathology
- Abstract
Purpose: [
18 F]FDG PET/CT in multiple myeloma (MM) is currently the best technology to demonstrate patchy and extramedullary disease. However, [18 F]FDG PET has some limitations, and imaging with alternative tracers should be explored. In this study, we aimed to evaluate the performance of [18 F]fluciclovine PET compared to [18 F]FDG PET in newly diagnosed MM patients., Procedures: Thirteen newly diagnosed transplant eligible MM patients were imaged both with [18 F]FDG PET/CT and [18 F]fluciclovine PET/CT within 1 week in a prospective study. The subjects were visually assessed positive or negative for disease. The number of lesions and the SUVmax of selected lesions were measured for both tracers. Furthermore, tracer uptake ratios were obtained by dividing lesion SUVmax by blood or bone marrow SUVmax . Between-group differences and correlations were assessed with paired t-tests and Pearson tests. Bone marrow SUVs were compared to bone marrow plasma cell percentage in biopsy samples., Results: Nine subjects were assessed positively by [18 F]FDG PET (69%) and 12 positives by [18 F]fluciclovine PET (92%). All positive subjects had [18 F]fluciclovine scans that were qualitatively scored as easier to interpret visually than the [18 F]FDG scans. The number of lesions was also higher; seven of nine subjects with distinct hot spots on [18 F]fluciclovine PET had fewer or no visible lesions on [18 F]FDG PET. The mean lesion SUVmax values were 8.2 and 3.8 for [18 F]fluciclovine and [18 F]FDG, respectively. The mean tumour to blood values were 6.4 and 2.0 for [18 F]fluciclovine and [18 F]FDG, and the mean ratios between tumour and bone marrow were 2.1 and 1.5 for [18 F]fluciclovine and [18 F]FDG. The lesion SUVmax and ratios were significantly higher for [18 F]fluciclovine (all p < 0.01). Local [18 F]fluciclovine SUVmax or SUVmean values in os ilium and the percentage of plasma cells in bone marrow biopsies were linearly correlated (p = 0.048). There were no significant correlations between [18 F]FDG SUVs and plasma cells (p = 0.82)., Conclusions: Based on this pilot study, [18 F]fluciclovine is a promising tracer for MM. The visual and semi-quantitative evaluations indicate that [18 F]fluciclovine PET/CT can out-perform [18 F]FDG PET/CT at diagnosis., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
99. A prospective study of Skeletal survey versus Low-dose whole-body CT for Osteolytic lesions in Multiple Myeloma.
- Author
-
Gundesen MT, Asmussen JT, Haukås E, Schubert M, Abildgaard N, Schjesvold F, and Lund T
- Subjects
- Humans, Prospective Studies, Radiation Dosage, Retrospective Studies, Tomography, X-Ray Computed, Whole Body Imaging, Multiple Myeloma diagnosis, Multiple Myeloma diagnostic imaging, Osteolysis diagnostic imaging, Osteolysis etiology
- Abstract
Whole-body low-dose CT (WBLDCT) is recommended over classical skeletal surveys (CSS) for investigating bone disease in multiple myeloma (MM) based on retrospective studies. No prospective studies with serial follow-up scans exist., Objective: To compare WBLDCT to CSS for identifying progressive bone disease in MM in a prospective setting., Methods: Ninety-six patients with MM at Odense University Hospital and Stavanger Hospital were followed for up to four years. Patients were scanned with WBLDCT and CSS every year for the first two years and every six months thereafter or at suspicion of progression., Results: Nineteen cases of progressive bone disease were found using WBLDCT vs eight cases using CSS (p < 0.001). All cases of progressive bone disease using CSS were also identified by WBLDCT. Progression not found by CSS was primarily in the spine, sternum, and pelvis. Of the 19 cases, five patients had progressive bone disease only without other criteria for clinical progression. WBLDCT consistently identified more bone lesions per patient, 8.2 CI(6.8;9.6) vs CSS, 3.6 CI(2.7;4.5)., Conclusion: WBLDCT outperformed CSS for finding progressive bone disease and osteolytic lesions. More new lesions were found during follow-up by WBLDCT than CSS. Using CSS for lytic lesions will underestimate progression rates. Our data offer prospective evidence for the current recommendation using WBLDCT for skeletal evaluations in patients with multiple myeloma., (© 2022 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
100. Melflufen for the treatment of multiple myeloma.
- Author
-
Ocio EM, Nadeem O, Schjesvold F, Gay F, Touzeau C, Dimopoulos MA, Richardson PG, and Mateos MV
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols, Dexamethasone therapeutic use, Europe, Phenylalanine analogs & derivatives, Melphalan analogs & derivatives, Melphalan pharmacology, Melphalan therapeutic use, Multiple Myeloma chemically induced, Multiple Myeloma drug therapy, Multiple Myeloma pathology
- Abstract
Introduction: Melphalan flufenamide (melflufen) is a first-in-class peptide-drug conjugate that takes advantage of increased aminopeptidase activity inside tumor cells to rapidly release alkylating agents therein. Melflufen in combination with dexamethasone has been evaluated in multiple clinical trials in patients with relapsed/refractory multiple myeloma (MM)., Areas Covered: This profile covers the unique mechanism of action of melflufen, the preclinical results supporting its activity in cellular models of resistance to chemotherapy, its activity in animal models of MM, and the clinical pharmacokinetics of melflufen. Findings from clinical trials evaluating melflufen, including the pivotal phase II HORIZON study and the phase III OCEAN study, are discussed., Expert Opinion: Although MM treatment has improved, patients with disease refractory to multiple standard-of-care drug classes face a dismal prognosis. Melflufen demonstrated efficacy and tolerability in select populations, with an initial approval in the United States in patients with ≥ four previous lines of therapy and triple-class-refractory MM. Results from the phase III OCEAN study - currently under discussion with regulatory agencies in the United States and Europe - are more complex and have been put into context herein. Lastly, melflufen provides a proof-of-concept for the utility of the peptide-drug conjugate platform in relapsed/refractory MM.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.