Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jesús F. San-Miguel is active.

Publication


Featured researches published by Jesús F. San-Miguel.


The New England Journal of Medicine | 2015

Carfilzomib, Lenalidomide, and Dexamethasone for Relapsed Multiple Myeloma

A. Keith Stewart; S. Vincent Rajkumar; Meletios A. Dimopoulos; Tamas Masszi; Ivan Spicka; Albert Oriol; Roman Hájek; Laura Rosiñol; David Siegel; Georgi Mihaylov; Vesselina Goranova-Marinova; Peter Rajnics; Aleksandr Suvorov; Ruben Niesvizky; Andrzej J. Jakubowiak; Jesús F. San-Miguel; Heinz Ludwig; Michael Wang; Vladimír Maisnar; Jiri Minarik; William Bensinger; Maria Victoria Mateos; Dina Ben-Yehuda; Vishal Kukreti; Naseem Zojwalla; Margaret Tonda; Xinqun Yang; Biao Xing; Philippe Moreau; Antonio Palumbo

BACKGROUND Lenalidomide plus dexamethasone is a reference treatment for relapsed multiple myeloma. The combination of the proteasome inhibitor carfilzomib with lenalidomide and dexamethasone has shown efficacy in a phase 1 and 2 study in relapsed multiple myeloma. METHODS We randomly assigned 792 patients with relapsed multiple myeloma to carfilzomib with lenalidomide and dexamethasone (carfilzomib group) or lenalidomide and dexamethasone alone (control group). The primary end point was progression-free survival. RESULTS Progression-free survival was significantly improved with carfilzomib (median, 26.3 months, vs. 17.6 months in the control group; hazard ratio for progression or death, 0.69; 95% confidence interval [CI], 0.57 to 0.83; P=0.0001). The median overall survival was not reached in either group at the interim analysis. The Kaplan-Meier 24-month overall survival rates were 73.3% and 65.0% in the carfilzomib and control groups, respectively (hazard ratio for death, 0.79; 95% CI, 0.63 to 0.99; P=0.04). The rates of overall response (partial response or better) were 87.1% and 66.7% in the carfilzomib and control groups, respectively (P<0.001; 31.8% and 9.3% of patients in the respective groups had a complete response or better; 14.1% and 4.3% had a stringent complete response). Adverse events of grade 3 or higher were reported in 83.7% and 80.7% of patients in the carfilzomib and control groups, respectively; 15.3% and 17.7% of patients discontinued treatment owing to adverse events. Patients in the carfilzomib group reported superior health-related quality of life. CONCLUSIONS In patients with relapsed multiple myeloma, the addition of carfilzomib to lenalidomide and dexamethasone resulted in significantly improved progression-free survival at the interim analysis and had a favorable risk-benefit profile. (Funded by Onyx Pharmaceuticals; ClinicalTrials.gov number, NCT01080391.).


Leukemia | 2008

Efficacy and safety of bortezomib in patients with renal impairment: results from the APEX phase 3 study

Jesús F. San-Miguel; Paul G. Richardson; Pieter Sonneveld; Michael W. Schuster; David M. Irwin; Edward A. Stadtmauer; Thierry Facon; Jean Luc Harousseau; D. Ben-Yehuda; S. Lonial; H. Goldschmidt; D. Reece; J. Bladé; Mario Boccadoro; Jamie Cavenagh; Rachel Neuwirth; Anthony Boral; Dixie Lee Esseltine; Kenneth C. Anderson

Renal impairment is associated with poor prognosis in multiple myeloma (MM). This subgroup analysis of the phase 3 Assessment of Proteasome Inhibition for Extending Remissions (APEX) study of bortezomib vs high-dose dexamethasone assessed efficacy and safety in patients with relapsed MM with varying degrees of renal impairment (creatinine clearance (CrCl) <30, 30–50, 51–80 and >80 ml min−1). Time to progression (TTP), overall survival (OS) and safety were compared between subgroups with CrCl ⩽50 ml min−1 (severe-to-moderate) and >50 ml min−1 (no/mild impairment). Response rates with bortezomib were similar (36–47%) and time to response rapid (0.7–1.6 months) across subgroups. Although the trend was toward shorter TTP/OS in bortezomib patients with severe-to-moderate vs no/mild impairment, differences were not significant. OS was significantly shorter in dexamethasone patients with CrCl ⩽50 vs >50 ml min−1 (P=0.003), indicating that bortezomib is more effective than dexamethasone in overcoming the detrimental effect of renal impairment. Safety profile of bortezomib was comparable between subgroups. With dexamethasone, grade 3/4 adverse events (AEs), serious AEs and discontinuations for AEs were significantly elevated in patients with CrCl ⩽50 vs >50 ml min−1. These results indicate that bortezomib is active and well tolerated in patients with relapsed MM with varying degrees of renal insufficiency. Efficacy/safety were not substantially affected by severe-to-moderate vs no/mild impairment.


Lancet Oncology | 2010

Treatment-related peripheral neuropathy in multiple myeloma: the challenge continues

Michel Delforge; Joan Bladé; Meletios A. Dimopoulos; Thierry Facon; Martin Kropff; Heinz Ludwig; Antonio Palumbo; Philip Van Damme; Jesús F. San-Miguel; Pieter Sonneveld

Introduction of the proteasome inhibitor bortezomib and the immunomodulatory drugs thalidomide and lenalidomide has substantially improved outcomes for patients with multiple myeloma. As a result, these drugs have become cornerstones of current antimyeloma treatment regimens. However, after several years of clinical experience it has become apparent that peripheral neuropathy is the most common and potentially disabling non-haematological side-effect associated with thalidomide and bortezomib. Maximising treatment benefit while preserving quality of life therefore requires a careful balance between achieving optimum activity and minimising toxicity, including neuropathy, to further enhance efficacy. In this review, we discuss all aspects of drug-induced peripheral neuropathy in myeloma, with a particular focus on thalidomide and bortezomib.


Haematologica | 2010

In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma

Enrique M. Ocio; David Vilanova; Peter Atadja; Patricia Maiso; Edvan Crusoe; Diego Fernández-Lázaro; Mercedes Garayoa; Laura San-Segundo; Teresa Hernández-Iglesias; Enrique de Alava; Wenlin Shao; Yung-Mae Yao; Atanasio Pandiella; Jesús F. San-Miguel

Background Combinations of drug treatments based on bortezomib or lenalidomide plus steroids have resulted in very high response rates in multiple myeloma. However, most patients still relapse, indicating the need for novel combination partners to increase duration of response or to treat relapsed disease. We explored the antimyeloma activity of triple combinations of these well-established schemes with panobinostat, a novel deacetylase inhibitor with a multi-targeted profile. Design and Methods The activity of these combinations was explored in vitro in cell lines by using MTT and annex-in V, ex vivo by flow cytometry, and in vivo using two different murine models of human myeloma: one bearing a subcutaneous plasmacytoma and another with a disseminated myeloma. Moreover, gene expression profiling and immunohistochemical studies were performed. Results The addition of panobinostat (LBH589) to dexamethasone and either bortezomib or lenalidomide resulted in clear potentiation in multiple myeloma cell lines, freshly isolated plasma cells, and murine models of multiple myeloma. The quantification of the potency of these combinations by using the Chou-Talalay method showed synergistic combination indices for all of them. This effect derived from the deregulation of a cluster of genes that was completely different from the sum of genes affected by the single agents (895 and 1323 genes exclusively deregulated by panobinostat and dexamethasone plus bortezomib or lenalidomide, respectively). Functional experiments, such as annexin V staining, cell cycle analysis, and immunohistochemical studies also supported this potentiation. Anti-myeloma efficacy was confirmed in an extramedullary plasmacytoma model and a disseminated luciferized model, in which panobinostat also provided a marked benefit in bone disease. Conclusions The potent activity, together with the exclusive mechanistic profile, provides the rationale for the clinical evaluation of these drug combinations in multiple myeloma.


Journal of Clinical Oncology | 2011

Phase Ib Study of Panobinostat and Bortezomib in Relapsed or Relapsed and Refractory Multiple Myeloma

Jesús F. San-Miguel; Paul G. Richardson; Andreas Günther; Orhan Sezer; David Siegel; Joan Bladé; Richard LeBlanc; Heather J. Sutherland; Monika Sopala; Kaushal Mishra; Song Mu; Priscille Bourquelot; Maria Victoria Mateos; Kenneth C. Anderson

PURPOSE Despite advancements, prognosis for patients with relapsed/refractory multiple myeloma (MM) is poor, and novel therapies are needed. Panobinostat is a potent deacetylase inhibitor that elicits synergistic effects on MM cells in combination with bortezomib. This phase Ib study sought to determine the maximum-tolerated dose (MTD) of panobinostat plus bortezomib in patients with relapsed or relapsed and refractory MM. PATIENTS AND METHODS In the dose-escalation phase (n = 47), panobinostat was administered orally thrice weekly every week in combination with bortezomib (21-day cycles). After MTD determination, patients were evaluated in an expansion phase (n = 15) that incorporated a 1-week treatment holiday of panobinostat, with dexamethasone added in cycle 2. Additional assessments included safety, pharmacokinetics, and efficacy per International Myeloma Working Group criteria. RESULTS The MTD was established at panobinostat 20 mg plus bortezomib 1.3 mg/m(2). Grade 3 or 4 adverse events (AEs) included thrombocytopenia (85.1%), neutropenia (63.8%), and asthenia (29.8%) in the escalation phase, and thrombocytopenia (66.7%), neutropenia (46.7%), and fatigue (20.0%) in the expansion phase. At MTD in the escalation phase, eight patients (47.1%) discontinued therapy as a result of AEs, whereas five patients (33.3%) discontinued treatment in the expansion phase. Expansion phase patients demonstrated greater median treatment duration. Overall response rate (ORR) was 73.3% in the expansion phase and 52.9% at the escalation phase MTD. Among bortezomib-refractory patients, the ORR was 26.3%, and 42.1% of patients had ≥ minimal response. CONCLUSION The MTD of panobinostat plus bortezomib was determined and demonstrated activity in patients with relapsed or relapsed/refractory MM, including bortezomib-refractory patients. A phase II/III clinical trial program (Panobinostat or Placebo With Bortezomib and Dexamethasone in Patients With Relapsed Multiple Myeloma [PANORAMA]) has been initiated.


British Journal of Haematology | 2000

Remission status defined by immunofixation vs.electrophoresis after autologous transplantation has a major impact on the outcome of multiple myeloma patients

Juan José Lahuerta; Joaquin Martinez-Lopez; Javier de la Serna; Joan Bladé; Carlos Grande; Adrian Alegre; Lourdes Vázquez; José García-Laraña; Ana Sureda; Javier de la Rubia; Eulogio Conde; Rafael Martínez; Katy Perez‐Equiza; José M. Moraleda; Angel Leon; Juan Besalduch; Rafael Cabrera; José D. Gonzalez-San Miguel; Alfonso Morales; Juan Carlos García-Ruiz; Joaquín Díaz-Mediavilla; Jesús F. San-Miguel

We have retrospectively analysed 344 multiple myeloma (MM) patients (202 de novo patients) treated in a non‐uniform way in whom high‐dose therapy and autologous stem cell transplantation (ASCT) response was simultaneously measured by both electrophoresis (EP) and immunofixation (IF). Patients in complete remission (CR) by EP were further subclassified as CR1 when IF was negative and CR2 when it remained positive. Partial responders (PR) were also subclassified as PR1 (very good PR, > 90% reduction in M‐component) or PR2 (50–90% reduction). CR1 patients showed a significantly better event‐free survival (EFS) [35% at 5 years, 95% confidence interval (CI) 17–53, median 46 months] and overall survival (OS) (72% at 5 years, CI 57–86, median not reached) compared with any other response group (univariate comparison P < 0·00000 to P = 0·004). In contrast, comparison of CR2 with PR1 and with PR2 did not define different prognostic subgroups (median EFS 30, 30 and 26 months respectively, P = 0·6; median survival 56, 44 and 42 months respectively, P = 0·5). The non‐responding patients had the worst outcome (5‐year OS 8%, median 7 months). Multivariate analysis confirmed both the absence of differences among CR2, PR1 and PR2 and the highly discriminatory prognostic capacity of a three‐category classification: (i) CR1 (ii) CR2 + PR1 + PR2, and (iii) non‐response (EFS P < 0·00000; OS P < 0·00000; both Cox models P < 0·00000). In the logistic regression analysis, the factors significantly associated with failure to achieve CR1 were the use of two or more up‐front chemotherapy lines, status of non‐response pre‐ASCT and inclusion of total body irradiation (TBI) in the preparative regimen. Tandem transplants or the use of multiple agents (busulphan and melphalan) in the preparative regimen resulted in a higher CR1 level; none of the biological factors explored influenced the possibility of achieving CR1. These results confirm that, in MM patients undergoing ASCT, achieving a negative IF identifies the patient subset with the best prognosis; accordingly, therapeutic strategies should be specifically designed to achieve negative IF.


Leukemia | 2009

Both expanded and uncultured mesenchymal stem cells from MDS patients are genomically abnormal, showing a specific genetic profile for the 5q-syndrome

O Lopez-Villar; J.L. García; F.M. Sanchez-Guijo; Cristina Robledo; Eva Villaron; P Hernández-Campo; M. Diez-Campelo; M V Barbado; José A. Pérez-Simón; Jesús María Hernández-Rivas; Jesús F. San-Miguel; M-C del Cañizo

The presence of cytogenetic aberrations on mesenchymal stem cells (MSC) from myelodysplastic syndrome (MDS) patients is controversial. The aim of the study is to characterize bone marrow (BM) derived MSC from patients with MDS using: kinetic studies, immunophenotyping, fluorescent in situ hybridization (FISH) and genetic changes by array-based comparative genomic hybridization (array-CGH). In all 36 cases of untreated MDS were studied. MDS–MSC achieved confluence at a significantly slower rate than donor-MSC, and the antigenic expression of CD105 and CD104 was lower. Array-CGH studies showed DNA genomic changes that were proved not to be somatic. These results were confirmed by FISH. To confirm that genomic changes were also present in freshly obtained MSCs they were enriched by sorting BM cells with the following phenotype: CD45−/CD73++/CD34−/CD271++. They also showed genomic changes that were confirmed by FISH. To analyze the relationship of these aberrations with clinical–biological data an unsupervized hierarchical cluster analysis was performed, two clusters were identified: the first one included the 5q− syndrome patients, whereas the other incorporated other MDS. Our results show, for the first time that MSC from MDS display genomic aberrations, assessed by array-CGH and FISH, some of them specially linked to a particular MDS subtype, the 5q− syndrome.


Leukemia | 2011

Optimizing the use of lenalidomide in relapsed or refractory multiple myeloma: Consensus statement

M. Dimopoulos; A. Palumbo; Michel Attal; Meral Beksac; Fe. Davies; Michel Delforge; Hermann Einsele; Roman Hájek; Jean Luc Harousseau; F. Leal da Costa; H. Ludwig; Ulf-Henrik Mellqvist; Gareth J. Morgan; Jesús F. San-Miguel; Sonja Zweegman; Pieter Sonneveld

An expert panel convened to reach a consensus regarding the optimal use of lenalidomide in combination with dexamethasone (Len/Dex) in patients with relapsed or refractory multiple myeloma (RRMM). On the basis of the available evidence, the panel agreed that Len/Dex is a valid and effective treatment option for most patients with RRMM. As with other therapies, using Len/Dex at first relapse is more effective regarding response rate and durability than using it after multiple salvage therapies. Len/Dex may be beneficial regardless of patient age, disease stage and renal function, although the starting dose of lenalidomide should be adjusted for renal impairment and cytopenias. Long-term treatment until there is evidence of disease progression may be recommended at the best-tolerated doses of both lenalidomide and dexamethasone. Recommendations regarding the prevention and management of adverse events, particularly venous thromboembolism and myelosuppression, were provided on the basis of the available evidence and practical experience of panel members. Ongoing trials will provide more insight into the effects of continuous lenalidomide-based therapy in myeloma.


Bone Marrow Transplantation | 1998

Autologous peripheral blood stem cell transplantation for multiple myeloma: a report of 259 cases from the Spanish Registry

A Alegre; Joaquín Díaz-Mediavilla; Jesús F. San-Miguel; Rafael Martínez; J. García Laraña; Anna Sureda; Juan-José Lahuerta; D Morales; J. Bladé; D Caballero; J de la Rubia; Antonio Escudero; Jose L. Diez-Martin; Fernando Hernandez-Navarro; J. Rifón; J. Odriozola; Salut Brunet; J. De La Serna; Juan Besalduch; Mj Vidal; C Solano; Arturo Vera-Ponce de León; Jj Sánchez; C Martínez-Chamorro; J.M. Fernández-Rañada

Between January 1989 and November 1995, 259 patients with multiple myeloma (MM), 22 stage I, 57 stage II and 180 stage III at diagnosis were treated with myeloablative high-dose therapy followed by autologous peripheral blood stem cell (PBSC) transplantation. The median time from diagnosis to transplantation was 17 months (6–112). At the time of transplant, 56 patients were in CR, 153 in PR, 25 were nonresponders and 25 had progressive disease. Mobilization of stem cells was performed with G-CSF alone in 141 cases, chemotherapy plus G-CSF in 65, chemotherapy plus GM-CSF in 36 and chemotherapy alone in 17 patients. The conditioning regimen consisted of high-dose melphalan alone in 96 patients, melphalan plus TBI in 73, busulfan plus melphalan in 56, busulfan plus cyclophosphamide in 27 and cyclophosphamide plus TBI in seven. The median durations of neutropenia (>0.5 × 109/l) and thrombocytopenia (>20 × 109/l) were 12 (5–118) and 13 days (5–360), respectively. Transplant-related mortality occurred in 11 patients (4%). Once a stable graft was achieved, 114 patients (44%) received maintenance treatment with recombinant alpha interferon (IFN-α). Among the 248 patients evaluable for response 125 (51%) had a CR and 100 had a PR (40%). The median duration of progression-free survival (PFS) and overall survival (OS) after transplantation was 23 and 35 months, respectively. Univariate analysis showed that response status pretransplant, only one line of primary induction treatment and IFN-αmaintenance treatment post-transplant significantly influenced OS. Female sex, pretransplant responsive disease, and treatment with IFN-α post-transplant were the factors significantly influencing PFS. The conditioning regimen and method of stem cell mobilization had no significant impact on OS and PFS. On multivariate analysis the only independent factors associated with a longer survival were the number of chemotherapy courses prior to autologous PBSC transplantation and the pretransplant response status. The present analysis from the Spanish Registry confirms the feasibility of autologous PBSC transplantation in myeloma patients with a very low toxicity (4% toxic deaths). The high complete response rate after transplantation is encouraging. The best results are obtained when the procedure is performed early after the first line of induction therapy and in patients with chemosensitive disease. Whether early high-dose therapy followed by autotransplantation in responding patients is superior to conventional chemotherapy is currently being investigated in prospective randomized studies.


British Journal of Haematology | 2010

Monoclonal gammopathy of undetermined significance: A consensus statement: Guideline

James R. Berenson; Kenneth C. Anderson; Robert A. Audell; Ralph V. Boccia; Morton Coleman; Meletios A. Dimopoulos; Matthew T. Drake; Rafael Fonseca; Jean Luc Harousseau; Douglas E. Joshua; Sagar Lonial; Ruben Niesvizky; Antonio Palumbo; G. David Roodman; Jesús F. San-Miguel; Seema Singhal; Donna M. Weber; Maurizio Zangari; Eric Wirtschafter; Ori Yellin; Robert A. Kyle

On February 25, 2009, a panel of international experts on plasma cell dyscrasia and skeletal disease met to discuss monoclonal gammopathy of undetermined significance (MGUS). This non‐malignant B‐cell disorder is the most common plasma cell dyscrasia and is associated with an increased risk of developing serious B‐cell disorders. Individuals with MGUS also have an increased risk of osteoporosis and osteopenia associated with an increased likelihood of developing fractures especially in the vertebral column, peripheral neuropathy and thromboembolic events. The goal of the meeting was to develop a consensus statement regarding the appropriate tests to screen, evaluate and follow‐up patients with MGUS. The panel also addressed the identification and treatment of MGUS‐related skeletal problems, thromboembolic events and neurological complications. The following consensus statement outlines the conclusions and marks the first time that a consensus statement for the screening and treatment of MGUS has been clearly stated.

Collaboration


Dive into the Jesús F. San-Miguel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meletios A. Dimopoulos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joan Bladé

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Albert Oriol

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Norma C. Gutiérrez

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge