126 results on '"M. Alcoceba"'
Search Results
2. P1260: UNRAVELING THE GENETICS OF TRANSFORMED SPLENIC MARGINAL ZONE LYMPHOMA
- Author
-
M. Grau, C. López, A. Navarro, G. Clot, F. Nadeu, G. Bastidas, M. Alcoceba, M. J. Baptista, M. Blanes, F. Climent, D. Colomer, D. Costa, E. Domingo-Domènech, P. Forcada, A. Enjuanes, L. Escoda, G. Frigola, E. Giné, M. Lopez-Guerra, A. Rivas-Delgado, L. Vicente-Folch, A. Wotherspoon, E. Campo, A. López-Guillermo, E. Matutes, and S. Beà
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
3. P1282: DISEASE-SPECIFIC U1 SPLICEOSOMAL RNA MUTATIONS IN MATURE B-CELL NEOPLASMS
- Author
-
F. Nadeu, S. Shuai, G. Clot, L. K. Hilton, A. Diaz-Navarro, S. Martín, R. Royo, T. Baumann, M. Kulis, I. López-Oreja, M. Cossio, J. Lu, V. Ljungström, E. Young, K. Plevova, B. A. Knisbacher, Z. Lin, C. K. Hahn, P. Bousquets, M. Alcoceba, M. González, E. Colado, M. Aymerich, M. J. Terol, A. Rivas-Delgado, A. Enjuanes, S. Ruiz-Gaspà, T. Chatzikonstantinou, D. Hägerstrand, C. Jylhä, A. Skaftason, L. Mansouri, K. Stranska, M. Doubek, E. J. van Gastel-Mol, Z. Davis, R. Walewska, L. Scarfò, L. Trentin, A. Visentin, S. A. Parikh, K. G. Rabe, R. Moia, M. Armand, D. Rossi, F. Davi, G. Gaidano, N. E. Kay, T. Shanafelt, P. Ghia, D. Oscier, A. W. Langerak, S. Beà, A. López-Guillermo, D. Neuberg, C. J. Wu, G. Getz, S. Pospisilova, K. Stamatopoulos, R. Rosenquist, W. Huber, T. Zenz, D. Colomer, J. I. Martín-Subero, J. Delgado, R. D. Morin, L. D. Stein, X. S. Puente, and E. Campo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
4. P1277: MUTATIONAL LANDSCAPE AND COPY NUMBER ALTERATIONS IN TESTICULAR LARGE B-CELL LYMPHOMA
- Author
-
C. López, A. Rivas-Delgado, F. Nadeu, M. Grau, A. Rivero, J. Boschs-Schips, M. Alcoceba, G. Tapia, L. Luizaga, C. Bárcena, N. Kelleher, M. Pablo, O. Balague, G. Frigola, N. Villamor, L. Magnano, T. Baumann, A. Muntañola, J. M. Sancho-Cia, A. M. García-Sancho, E. Gonzalez Barca, F. Climent, E. Campo, E. Giné, A. López-Guillermo, and S. Beà
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
5. Immunotherapy: HIGH INCIDENCE OF CYTOPENIAS AFTER COMMERCIAL CAR-T CELL THERAPY IN A REAL-WORLD SETTING: UTILITY OF THE CAR-HEMATOTOX INDEX
- Author
-
A. Navarro-Bailon, A. Martin, V. Martin-Mendez, L. Prieto, E. Perez-Lopez, A. Cabero, M. Garcia-Blazquez, M. Fernandez-Crespo, S. Muntión, S. Preciado, M. Gomez-Redondo, M. Alaña, N. Albala, M. Lopez-Parra, P. Tamayo, E. Villaron, A. Yeguas, M. Alcoceba, A. Orfao, N. Gutierrez, L. Lopez-Corral, and F. Sanchez-Guijo
- Subjects
Cancer Research ,Transplantation ,Oncology ,Immunology ,Immunology and Allergy ,Cell Biology ,Genetics (clinical) - Published
- 2023
- Full Text
- View/download PDF
6. MIPI53. A NEW PROGNOSTIC INDEX FOR MANTLE CELL LYMPHOMA
- Author
-
Boente, M. Bastos, primary, Sánchez, M. Alcoceba, additional, Bailón, A. Navarro, additional, Herrera, A. Medina, additional, Álvarez, M. García, additional, Sarasquete, M. E. Alonso, additional, Santos, C. Chillón, additional, Echevarría, A. Balanzategui, additional, Gómez, A. Antón, additional, Niño, J., additional, Loizaga, I. de la Torre, additional, Bedera, N. H. Vidaña, additional, Sánchez, R. Maldonado, additional, Ruano, M. Hernández, additional, Sánchez, S. M. Lucas, additional, Villares, M. I. Sánchez, additional, González, A. I. Sánchez, additional, González de la Calle, V., additional, Díaz, M. González, additional, Sánchez, C. Jiménez, additional, Sanz, R. García, additional, and García‐Sancho, A. Martín, additional
- Published
- 2023
- Full Text
- View/download PDF
7. EHA2021 Virtual Congress Abstract BookEP631 OTHER MALIGNANCIES IN THE HISTORY OF CHRONIC LYMPHOCYTIC LEUKEMIA -, A RETROSPECTIVE, MULTICENTRE COHORT STUDY BY ERIC, THE EUROPEAN RESEARCH INITIATIVE ON CLL IN HARMONY. Abstract S143 CLINICAL IMPACT OF RECURRENT GENE MUTATIONS IN CHRONIC LYMPHOCYTIC LEUKEMIA: A RETROSPECTIVE, MULTI-CENTER COHORT STUDY BY ERIC, THE EUROPEAN RESEARCH INITIATIVE ON C
- Author
-
T. Chatzikonstantinou 1,2,*, L. Scarfò 1,3, J. Kotašková 4,5, G. Iacoboni 6, E. Minga 2, D. Chammou 2, G. Karakatsoulis 2, M. Alcoceba 7, S. El-Ashwah 8, F. Bacchiarri 9, M. Ali Khan 10,R. Cassin 11, S. Chatzileontiadou 12, A. Christian 13, R. Collado 14, M. D. De Deus Santos 15, Z. Davis 13, C. Demosthenous 16, M. Dimou 17, D. Donaldson 18, G. Dos Santos 19, B. Dreta 20,M. Efstathopoulou 21
- Published
- 2021
- Full Text
- View/download PDF
8. A case of aeromonas trota in an immunocompromised patient with Diarrhea
- Author
-
Universitat Rovira i Virgili, Fernández-Bravo A; Fort-Gallifa I; Ballester F; Pujol I; Gomez-bertomeu F; Domínguez M; Micó M; Alcoceba E; Simó-Sisó JM; Figueras MJ, Universitat Rovira i Virgili, and Fernández-Bravo A; Fort-Gallifa I; Ballester F; Pujol I; Gomez-bertomeu F; Domínguez M; Micó M; Alcoceba E; Simó-Sisó JM; Figueras MJ
- Abstract
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. According to recent literature, 95.4% of the Aeromonas strains associated with human clinical cases correspond to four species: Aeromonas caviae, Aeromonas dhakensis, Aeromonas veronii and Aeromonas hydrophila. However, other less prevalent species such as Aeromonas trota, are also described from clinical samples. Based on its low incidence, the latter species can be regarded as rare and it is the only Aeromonas species susceptible to ampicillin. From the taxonomic point of view, A. trota is considered a synonym of the species Aeromonas enteropelogenes. The objective of this study is to present a new clinical case associated with A. trota in order to increase the knowledge about this species. The strain was recovered from the feces of a 69-year-old patient with a diarrheal syndrome and peritoneal psammocarcinoma. The preliminary identification as Aeromonas sp. was obtained with the API 20E, but it was characterized as Aeromonas jandei and also as Aeromonas enteropelogenes with different scores with the matrix-assisted laser desorption ionization time of flight (MALDI-TOF). Based on the sequence of the rpoD gene, it was confirmed to be A. trota. The antimicrobial resistance pattern showed that the strain was susceptible to ampicillin, penicillins in combination with beta-lactamase inhibitors, quinolones, carbapenems, aminoglycosides and cephalosporins, except cephalothin. In conclusion, the recognition of an Aeromonas strain susceptible to ampicillin should alert the clinical microbiologist of the possible involvement of this rare species. Furthermore, the MALDI-TOF database should be updated indicating that the species A. enteropelogenes, is a synonym of A. trota.
- Published
- 2020
9. CAR T-CELLS ARE ARRIVING. IS ALLOGENEIC TRANSPLANT AN OBSOLETE APPROACH FOR DE NOVO/TRANSFORMED DLBCL IN THE CAR T-CELLS ERA? LONG-TERM FOLLOW-UP OF A SINGLE CENTRE UNIT
- Author
-
L. Prieto-García, M. Alcoceba, E. Pérez-López, L. López-Corral, M. Delgado, F. Sánchez-Guijo, A. Martín, A. Navarro-Bailón, M. Baile, M. López-Parra, P. Tamayo, N.C. Gutiérrez, L. Vázquez, M. Cabrero, and D. Caballero
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Single centre ,business.industry ,Long term follow up ,Internal medicine ,medicine ,Hematology ,General Medicine ,Car t cells ,business - Published
- 2019
- Full Text
- View/download PDF
10. Autologous and Allogeneic Stem-Cell Transplantation for Transformed Waldenström Macroglobulinemia.
- Author
-
Durot E, Kanagaratnam L, Zanwar S, Kaufman A, D'Sa S, Toussaint E, Roos-Weil D, Alcoceba M, Vos JMI, Michallet AS, Talaulikar D, Kastritis E, Khwaja J, Treon SP, Garcia-Sanz R, Morel P, Munoz J, Castillo JJ, Kapoor P, and Delmer A
- Published
- 2024
- Full Text
- View/download PDF
11. Quantifying HLA Mismatches at Epitope Level in Haplo-HSCT: Impact in the Outcome in Strategies Using PTCy.
- Author
-
Gil-Etayo FJ, Niño-Ramírez JE, Fonseca-Santos M, Arroyo-Sánchez D, Navarro-Bailón A, Vicente Parra A, Jiménez Hernaz I, Terradillos Sánchez P, Boix F, Alcoceba M, Marín L, Pérez-López E, Cabrero M, Martín-López AÁ, López M, Baile M, Avendaño A, Cabero A, García-Bacelar A, Vázquez L, Sánchez-Guijo F, García-Sanz R, López-Corral L, and Tejeda Velarde A
- Subjects
- Humans, Female, Male, Adult, Middle Aged, Transplantation, Haploidentical methods, Algorithms, Adolescent, Young Adult, Hematologic Neoplasms therapy, Hematologic Neoplasms immunology, Hematologic Neoplasms mortality, Aged, Immunosuppressive Agents therapeutic use, Hematopoietic Stem Cell Transplantation, Graft vs Host Disease immunology, HLA Antigens immunology, Epitopes immunology, Cyclophosphamide therapeutic use, Histocompatibility Testing methods
- Abstract
Haploidentical haematopoietic stem cell transplantation (haplo-HSCT) is one of the most effective therapies for treating malignant haematological disorders. However, HLA disparities are significant barriers to the success of this process since they increase the risk of graft versus host disease (GvHD). HLA disparities quantification could help to anticipate the probability and degree of GvHD, but the best tool for such quantification remains a challenge. The aim of this study was to quantify the degree of HLA epitope incompatibilities using PIRCHE (Predicted Indirectly Recognisable HLA Epitopes) algorithm for immunogenicity prediction and their potential relationship with GvHD degree and clinical outcome. We studied 145 patients who underwent a haplo-HSCT with post-transplant cyclophosphamide (PTCy) from a related donor between 2013 and 2020 at our centre evaluating molecular HLA mismatches by PIRCHE-algorithm. Patients with PIRCHE Score (PS) I + II > 38 in GvH direction, developed acute GvHD grade II-IV earlier than their counterparts (HR: 1.71, 95% CI: 1.02-2.87, p = 0.032). In addition, PS-B > 1 in GvH direction was an independent risk factor for chronic GvHD, (HR: 2.51, 95% CI 1.17-5.36, p = 0.017). A higher incidence of relapse was found for patients with PS-II > 28 HvG (HR: 9.16, 95% CI: 2.47-33.92, p < 0.001) which also favoured a worse GvHD/relapse-free survival in patients with PS-II > 27 in HvG direction (HR: 1.88, 95% CI: 1.11-3.18, p = 0.017). These findings indicate that the alloreactivity inferred by epitope HLA disparities is associated with post-transplant outcomes. Thus, analysing PS could benefit the selection of the most suitable donors allowing patient stratification based on HLA mismatches in the context of haplo-HSCT with PTCy., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
12. Testicular large B-cell lymphoma is genetically similar to PCNSL and distinct from nodal DLBCL.
- Author
-
Rivas-Delgado A, López C, Clot G, Nadeu F, Grau M, Frigola G, Bosch-Schips J, Radke J, Ishaque N, Alcoceba M, Tapia G, Luizaga L, Barcena C, Kelleher N, Villamor N, Baumann T, Muntañola A, Sancho-Cia JM, García-Sancho AM, Gonzalez-Barca E, Matutes E, Brito JA, Karube K, Salaverria I, Enjuanes A, Wiemann S, Heppner FL, Siebert R, Climent F, Campo E, Giné E, López-Guillermo A, and Beà S
- Abstract
Testicular large B-cell lymphoma (TLBCL) is an infrequent and aggressive lymphoma arising in an immune-privileged site and has recently been recognized as a distinct entity from diffuse large B-cell lymphoma (DLBCL). We describe the genetic features of TLBCL and compare them with published series of nodal DLBCL and primary large B-cell lymphomas of the CNS (PCNSL). We collected 61 patients with TLBCL. We performed targeted next-generation sequencing, copy number arrays, and fluorescent in situ hybridization to assess chromosomal rearrangements in 40 cases with available material. Seventy percent of the cases showed localized stages. BCL6 rearrangements were detected in 36% of cases, and no concomitant BCL2 and MYC rearrangements were found. TLBCL had fewer copy number alterations ( p < 0.04) but more somatic variants ( p < 0.02) than nodal DLBCL and had more frequent 18q21.32-q23 ( BCL2 ) gains and 6q and 9p21.3 ( CDKN2A/B ) deletions. PIM1 , MYD88
L265P , CD79B , TBL1XR1 , MEF2B , CIITA , EP300, and ETV6 mutations were more frequent in TLBCL, and BCL10 mutations in nodal DLBCL. There were no major genetic differences between TLBCL and PCNSL. Localized or disseminated TLBCL displayed similar genomic profiles. Using LymphGen, the majority of cases were classified as MCD. However, we observed a subgroup of patients classified as BN2, both in localized and disseminated TLBCL, suggesting a degree of genetic heterogeneity in the TLBCL genetic profile. TLBCL has a distinctive genetic profile similar to PCNSL, supporting its recognition as a separate entity from DLBCL and might provide information to devise targeted therapeutic approaches., Competing Interests: Ferran Nadeu has received honoraria from Janssen, AbbVie, AstraZeneca, and SOPHiA GENETICS for speaking at educational activities; has received research support from Gilead; and has licensed the use of the protected IgCaller algorithm to Diagnóstica Longwood. Tycho Baumann has received consulting fees or honoraria from Janssen, Roche, Novartis, Merck, Gilead/Kite, Incyte, Lilly, Abbvie, AstraZeneca, and BeiGene. Alejandro Martin García‐Sancho has received consulting fees or honoraria from Janssen, Roche, BMS/Celgene, Kyowa Kirin, Clinigen, EUSAPharma, Novartis, Gilead/Kite, Incyte, Lilly, Takeda, ADC Therapeutics America, Miltenyi, Ideogen, Abbvie, and BeiGene. Elías Campo has been a consultant for GenMab, and Takeda; has received research support from AstraZeneca; received honoraria from Janssen, EUSPharma, Takeda, and Roche for speaking at educational activities; and is an inventor on a Lymphoma and Leukemia Molecular Profiling Project patent “Method for subtyping lymphoma subtypes by means of expression profiling” (PCT/US2014/64161) and a bioinformatic tool (IgCaller) licensed to Diagnostic Longwood. Eva Giné has received honoraria or consulting fees from Gilead, Kite Pharma, Janssen, Genmab, Miltenyi, and Lilly; has received research support from Janssen and travel expenses from Gilead and Kite Pharma. Armando López Guillermo served on the advisory board of Roche, Celgene, Novartis, and Gilead/Kite, received grants from Celgene and Gilead/Kite, and travel expenses from Kite/Gilead. The remaining authors declare no competing interests., (© 2024 The Author(s). HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)- Published
- 2024
- Full Text
- View/download PDF
13. Therapeutic strategies and treatment sequencing in patients with chronic lymphocytic leukemia: An international study of ERIC, the European Research Initiative on CLL.
- Author
-
Chatzikonstantinou T, Scarfò L, Minga E, Karakatsoulis G, Chamou D, Kotaskova J, Iacoboni G, Demosthenous C, Albi E, Alcoceba M, Al-Shemari S, Aurran-Schleinitz T, Bacchiarri F, Chatzileontiadou S, Collado R, Davis Z, de Deus Santos MD, Dimou M, Dmitrieva E, Donaldson D, Dos Santos G, Dreta B, Efstathopoulou M, El-Ashwah S, Enrico A, Frygier A, Galimberti S, Galitzia A, Gimeno E, Guarente V, Guieze R, Harrop S, Hatzimichael E, Herishanu Y, Hernández-Rivas JÁ, Jaksic O, Kalicińska E, Laribi K, Karakus V, Kater AP, Kho B, Kislova M, Konstantinou Ε, Koren-Michowitz M, Kotsianidis I, Kubova Z, Labrador J, Lad D, Laurenti L, Longval T, Lopez-Garcia A, Marquet J, Maslejova S, Mayor-Bastida C, Mihaljevic B, Milosevic I, Miras F, Moia R, Morawska M, Nath UK, Navarro-Bailón A, Olivieri J, Panovska-Stavridis I, Papaioannou M, Pierie C, Puiggros A, Reda G, Rigolin GM, Ruchlemer R, Schipani M, Schiwitza A, Shen Y, Shokralla T, Simkovic M, Smirnova S, Soliman DSA, Stilgenbauer S, Tadmor T, Tomic K, Tse E, Vassilakopoulos T, Visentin A, Vitale C, Vrachiolias G, Vukovic V, Walewska R, Xu Z, Yagci M, Yañez L, Yassin M, Zuchnicka J, Oscier D, Gozzetti A, Panagiotidis P, Bosch F, Sportoletti P, Espinet B, Pangalis GA, Popov VM, Mulligan S, Angelopoulou M, Demirkan F, Papajík T, Biderman B, Murru R, Coscia M, Tam C, Cuneo A, Gaidano G, Claus R, Stavroyianni N, Trentin L, Antic D, Smolej L, Kalashnikova OB, Catherwood M, Spacek M, Pospisilova S, Doubek M, Nikitin E, Chatzidimitriou A, Ghia P, and Stamatopoulos K
- Abstract
Competing Interests: Thomas Chatzikonstantinou received honoraria from AbbVie. Lydia Scarfò received honoraria from AbbVie, AstraZeneca, BeiGene, Lilly, Janssen, Octapharma. Gloria Iacoboni received honoraria and travel support from Novartis, Kite/Gilead, Bristol‐Myers Squibb, Abbvie, Autolus, Miltenyi, and AstraZeneca. Rosa Collado received support for attending meetings from Janssen‐Cilag and S.A. Sara Galimberti received honoraria support for attending meetings from AbbVie, AstraZeneca, Jazz, Novartis, and Incyte, honoraria from Roche, Celgene, Pfizer, and Janssen, and support for attending meetings from Jazz, AstraZeneca, and Roche. Romain Guieze received honoraria, consulting fees, and support for attending meetings from AbbVie, Beigene, Roche, Janssen, and AstraZeneca. Eleftheria Hatzimichael received honoraria from AbbVie, Janssen‐Cilag, AstraZeneca, and Roche. Yair Herishanu received honoraria from Janssen, AbbVie, Roche, AstraZeneca, Medion, and Lilly. José‐Ángel Hernández‐Rivas received honoraria as a consultant from Janssen, AbbVie, AstraZeneca, Lilly, and BeiGene and support for attending meetings from Janssen, AbbVie, AstraZeneca, and BeiGene. Ozren Jaksic received honoraria from Johnson and Johnson, AstraZeneca, and Lilly, honoraria from Johnson and Johnson, AbbVie, AstraZeneca, and Lilly, and support for attending meetings from Johnson and Johnson, and AbbVie. Kamel Laribi received consulting fees from AbbVie, AstraZeneca, Janssen, Beigene, Takeda, and Novartis. Maya Koren‐Michowitz received honoraria from Novartis, Pfizer, and Gad Medical LTD. and support for attending meetings from Novartis. Arnon P. Kater received advisory board fees and research money from Janssen, AbbVie, BMS, AstraZeneca, and Roche/Genentech, and support for attending meetings from Janssen and AbbVie. Ioannis Kotsianidis received honoraria and consulting fees from AbbVie and Janssen. Ivana Milosevic received honoraria from AbbVie, Roche, Sandoz, AstraZeneca, and Janssen, and support for attending meetings from AbbVie, Roche, and Takeda. Almudena Navarro‐Bailón received honoraria, advisory board fees, and support for attending meetings from AbbVie, AstraZeneca, Takeda, Janssen, and Beigene. Jacopo Olivieri received honoraria from AbbVie, AstraZeneca, and Janssen. Gianluigi Reda received consulting fees from AbbVie, AstraZeneca, Janssen, and Beigene, and is currently employed by AstraZeneca. Gian M. Rigolin received honoraria for participation in speaker's bureau from AbbVie, Astra Zeneca, Beigene, and Janssen, and support for attending meetings from Janssen. Mattia Schipani received honoraria and support for attending meetings from AstraZeneca, AbbVie, and Janssen‐Cilag and owns shares of stock in AbbVie, AstraZeneca, Merck, Eli Lilly, Sanofi, Johnson and Johnson, Pfizer, Gilead, and GSK. Tereza Shokralla and Stephan Stilgenbauer reports research funding from, consultancy or advisory role for, honoraria from, speakers' bureau participation for, and travel support from AbbVie, Amgen, AstraZeneca, BeiGene, Bristol Myers Squibb, Celgene, Gilead, GlaxoSmithKline, Hoffmann‐La Roche, Incyte, Infinity, Janssen, Novartis, and Sunesis. Eric Tse received support for attending meetings from Takeda. Theodoros Vassilakopoulos received honoraria from Takeda, Roche, Genesis Pharma, Merck, Novartis, Gilead, Sandoz, AstraZeneca, Integris, and Servier, and support for attending meetings from Takeda, Roche, Genesis Pharma, Merck, Pfizer, and Winmedica. Candida Vitale received honoraria from AbbVie, consulting fees from AstraZeneca, and support for attending meetings from AstraZeneca, Takeda, and Janssen. Renata Walewska received honoraria from AbbVie, AstraZeneca, and Beigene, support for attending meetings from Janssen, AbbVie, and AstraZeneca, and advisory board fees from AbbVie, AstraZeneca, Janssen, Beigene, and SecuraBio. Lucrecia Yañez received honoraria from AbbVie, AstraZeneca, Novartis, Gilead, Janssen, Jazz, MSD, and Pfizer, support for attending meetings from AbbVie, AstraZeneca, Gilead, Janssen, and Pfizer, and advisory board fees from AbbVie, AstraZeneca, Jazz, Janssen, Beigene, and Celgene. Francesc Bosch received consulting fees, honoraria, and payment for expert testimony from AbbVie, Genentech, Novartis, Takeda, Janssen, Roche, Mundipharma, Celgene/BMS, AstraZeneca, Lilly, Beigene, Gilead and TG Therapeutics, Advantage Allogene, Lava Therapeutics, and Enterome. Stephen Mulligan received advisory board fees from AbbVie, AstraZeneca, Janssen, Roche, and BeiGene. Maria Angelopoulou received consulting fees from AbbVie, Takeda, Janssen, Roche, Genesis, Gilead, and Amgen and honoraria from AbbVie, Takeda, Roche, Genesis, Gilead, and Novartis. Fatih Demirkan received support for attending meetings from Janssen and AbbVie. Tomas Papajík received honoraria and advisory board fees from AbbVie, Janssen‐Cilag, and AstraZeneca, and support for attending meetings from AstraZeneca. Marta Coscia received honoraria, advisory board fees, and support for attending meetings from AbbVie, AstraZeneca, and Janssen. Constantine Tam received honoraria from AbbVie, Beigene, Janssen, and LOXO. Antonio Cuneo received honoraria, advisory board fees, and support for attending meetings from AbbVie, AstraZeneca, Beigene, Janssen, and Lilly. Gianluca Gaidano received honoraria from Abbvie, AstraZeneca, BeiGene, Hikma, Incyte, Janssen, and Lilly. Niki Stavroyianni received honoraria from Janssen, AbbVie, AstraZeneca, and Lilly, and support for attending meetings from Janssen and AstraZeneca. Lukas Smolej received consulting fees, honoraria, and support for attending meetings from AbbVie, AstraZeneca, and Janssen and advisory board fees from AbbVie and AstraZeneca. Martin Spacek received honoraria and consulting and advisory board fees, and support for attending meetings from AbbVie, AstraZeneca, and Janssen. Michael Doubek received research support and honoraria from AbbVie, AstraZeneca, and Janssen. Eugene Nikitin received honoraria from AbbVie. Kostas Stamatopoulos received research support from AbbVie, AstraZeneca, Janssen, Novartis, and Roche; honoraria from AbbVie, AstraZeneca, Bristol Myers Squibb, Lilly, and Janssen. Paolo Ghia received research support from AbbVie, AstraZeneca, BMS, Janssen and honoraria from AbbVie, AstraZeneca, BeiGene, BMS, Galapagos, Genmab, Janssen, Loxo Oncology @Lilly, MSD, Roche, and is an Editor of HemaSphere. Georgios Karakatsoulis, Eva Minga, Dimitra Chamou, Jana Kotaskova, Christos Demosthenous, Elisa Albi, Miguel Alcoceba, Salem Al‐Shemari, Thérèse Aurran‐Schleinitz, Francesca Bacchiarri, Sofia Chatzileontiadou, Zadie Davis, Marcos Daniel de Deus Santos, Maria Dimou, Elena Dmitrieva, David Donaldson, Gimena Dos Santos, Barbara Dreta, Maria Efstathopoulou, Shaimaa El‐Ashwah, Alicia Enrico, Andrzej Frygier, Andrea Galitzia, Eva Gimeno, Valerio Guarente, Sean Harrop, Elżbieta Kalicińska, Volkan Karakus, Bonnie Kho, Maria Kislova, Εliana Konstantinou, Zuzana Kubova, Jorge Labrador, Deepesh Lad, Luca Laurenti, Thomas Longval, Alberto Lopez‐Garcia, Juan Marquet, Stanislava Maslejova, Carlota Mayor‐Bastida, Biljana Mihaljevic, Fatima Miras, Riccardo Moia, Marta Morawska, Uttam K. Nath, Irina Panovska‐Stavridis, Maria Papaioannou, Cheyenne Pierie, Anna Puiggros, Rosa Ruchlemer, Annett Schiwitza, Yandong Shen, Tereza Shokralla, Martin Simkovic, Svetlana Smirnova, Dina S. A. Soliman, Tamar Tadmor, Kristina Tomic, Andrea Visentin, George Vrachiolias, Vojin Vukovic, Zhenshu Xu, Munci Yagci, Mohamed Yassin, Jana Zuchnicka, David Oscier, Alessandro Gozzetti, Panagiotis Panagiotidis, Blanca Espinet, Paolo Sportoletti, Gerassimos A. Pangalis, Viola M. Popov, Bella Biderman, Roberta Murru, Rainer Claus, Livio Trentin, Darko Antic, Olga B. Kalashnikova, Mark Catherwood, Sarka Pospisilova, and Anastasia Chatzidimitriou have no conflict of interest to disclose.
- Published
- 2024
- Full Text
- View/download PDF
14. Killer-cell immunoglobulin-like receptor polymorphism is associated with COVID-19 outcome: Results of a pilot observational study.
- Author
-
Niño-Ramírez JE, Alcoceba M, Gutiérrez-Zufiaurre MN, Marcos M, Gil-Etayo FJ, Bartol-Sánchez MR, Eiros R, Chillón MC, García-Álvarez M, Terradillos-Sánchez P, Presa D, Muñoz JL, López-Bernús A, López-Sánchez E, González-Calle D, Sánchez PL, Compán-Fernández O, González M, García-Sanz R, and Boix F
- Subjects
- Humans, Male, Female, Middle Aged, Pilot Projects, Retrospective Studies, Polymorphism, Genetic, Aged, Genotype, Genetic Predisposition to Disease, Adult, Severity of Illness Index, HLA Antigens genetics, COVID-19 genetics, COVID-19 immunology, COVID-19 virology, Receptors, KIR genetics, SARS-CoV-2 immunology
- Abstract
The pathogenesis of COVID-19 warrants unravelling. Genetic polymorphism analysis may help answer the variability in disease outcome. To determine the role of KIR and HLA polymorphisms in susceptibility, progression, and severity of SARS-CoV-2 infection, 458 patients and 667 controls enrolled in this retrospective observational study from April to December 2020. Mild/moderate and severe/death study groups were established. HLA-A, -B, -C, and KIR genotyping were performed using the Lifecodes® HLA-SSO and KIR-SSO kits on the Luminex® 200™ xMAP fluoroanalyser. A probability score using multivariate binary logistic regression analysis was calculated to estimate the likelihood of severe COVID-19. ROC analysis was used to calculate the best cut-off point for predicting a worse clinical outcome with high sensitivity and specificity. A p ≤ 0.05 was considered statistically significant. KIR AA genotype protected positively against severity/death from COVID-19. Furthermore, KIR3DL1, KIR2DL3 and KIR2DS4 genes protected patients from severe forms of COVID-19. KIR Bx genotype, as well as KIR2DL2, KIR2DS2, KIR2DS3 and KIR3DS1 were identified as biomarkers of severe COVID-19. Our logistic regression model, which included clinical and KIR/HLA variables, categorised our cohort of patients as high/low risk for severe COVID-19 disease with high sensitivity and specificity (Se = 94.29%, 95% CI [80.84-99.30]; Sp = 84.55%, 95% CI [79.26-88.94]; OR = 47.58, 95%CI [11.73-193.12], p < 0.0001). These results illustrate an association between KIR/HLA ligand polymorphism and different COVID-19 outcomes and remarks the possibility of use them as a surrogate biomarkers to detect severe patients in possible future infectious outbreaks., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
15. Liquid biopsy for molecular characterization of diffuse large B-cell lymphoma and early assessment of minimal residual disease.
- Author
-
Alcoceba M, Stewart JP, García-Álvarez M, Díaz LG, Jiménez C, Medina A, Chillón MC, Gazdova J, Blanco O, Díaz FJ, Peñarrubia MJ, Fernández S, Montes C, Cabero A, Caballero MD, García-Sanz R, González M, González D, Tamayo P, Gutiérrez NC, García-Sancho AM, and Sarasquete ME
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Adult, Liquid Biopsy methods, Aged, 80 and over, Positron Emission Tomography Computed Tomography, Rituximab therapeutic use, Rituximab administration & dosage, Cyclophosphamide therapeutic use, Cyclophosphamide administration & dosage, Biomarkers, Tumor blood, Vincristine therapeutic use, Vincristine administration & dosage, Prognosis, Doxorubicin therapeutic use, Doxorubicin administration & dosage, High-Throughput Nucleotide Sequencing, Prednisone therapeutic use, Prednisone administration & dosage, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse diagnosis, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse blood, Lymphoma, Large B-Cell, Diffuse mortality, Lymphoma, Large B-Cell, Diffuse pathology, Neoplasm, Residual diagnosis, Circulating Tumor DNA blood, Circulating Tumor DNA genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
Circulating tumour DNA (ctDNA) allows genotyping and minimal residual disease (MRD) detection in lymphomas. Using a next-generation sequencing (NGS) approach (EuroClonality-NDC), we evaluated the clinical and prognostic value of ctDNA in a series of R-CHOP-treated diffuse large B-cell lymphoma (DLBCL) patients at baseline (n = 68) and after two cycles (n = 59), monitored by metabolic imaging (positron emission tomography combined with computed tomography [PET/CT]). A molecular marker was identified in 61/68 (90%) ctDNA samples at diagnosis. Pretreatment high ctDNA levels significantly correlated with elevated lactate dehydrogenase, advanced stage, high-risk International Prognostic Index and a trend to shorter 2-year progression-free survival (PFS). Valuable NGS data after two cycles of treatment were obtained in 44 cases, and 38 achieved major molecular response (MMR; 2.5-log drop in ctDNA). PFS curves displayed statistically significant differences among those achieving MMR versus those not achieving MMR (2-year PFS of 76% vs. 0%, p < 0.001). Similarly, more than 66% reduction in ΔSUVmax by PET/CT identified two subgroups with different prognosis (2-year PFS of 83% vs. 38%; p < 0.001). Combining both approaches MMR and ΔSUVmax reduction, a better stratification was observed (2-year PFS of 84% vs. 17% vs. 0%, p < 0.001). EuroClonality-NDC panel allows the detection of a molecular marker in the ctDNA in 90% of DLBCL. ctDNA reduction at two cycles and its combination with interim PET results improve patient prognosis stratification., (© 2024 The Author(s). British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
16. Single-Cell DNA Sequencing and Immunophenotypic Profiling to Track Clonal Evolution in an Acute Myeloid Leukemia Patient.
- Author
-
García-Álvarez M, Yeguas A, Jiménez C, Medina-Herrera A, González-Calle V, Hernández-Ruano M, Maldonado R, Aires I, Casquero C, Sánchez-Villares I, Balanzategui A, Sarasquete ME, Alcoceba M, Vidriales MB, González-Díaz M, García-Sanz R, and Chillón MC
- Abstract
Single-cell DNA sequencing can address the sequence of somatic genetic events during myeloid transformation in relapsed acute myeloid leukemia (AML). We present an NPM1 -mutated AML patient with an initial low ratio of FLT3 -ITD (low-risk ELN-2017), treated with midostaurin combined with standard chemotherapy as front-line treatment, and with salvage therapy plus gilteritinib following allogenic stem cell transplantation after relapse. Simultaneous single-cell DNA sequencing and cell-surface immunophenotyping was used in diagnostic and relapse samples to understand the clinical scenario of this patient and to reconstruct the clonal composition of both tumors. Four independent clones were present before treatment: DNMT3A/DNMT3A/NPM1 (63.9%), DNMT3A/DNMT3A (13.9%), DNMT3A/DNMT3A/NPM1/FLT3 (13.8%), as well as a wild-type clone (8.3%), but only the minor clone with FLT3 -ITD survived and expanded after therapy, being the most represented one (58.6%) at relapse. FLT3 -ITD was subclonal and was found only in the myeloid blast population (CD38/CD117/CD123). Our study shows the usefulness of this approach to reveal the clonal architecture of the leukemia and the identification of small subclones at diagnosis and relapse that may explain how the neoplastic cells can escape from the activity of different treatments in a stepwise process that impedes the disease cure despite different stages of complete remission.
- Published
- 2023
- Full Text
- View/download PDF
17. IGLV3-21R110 mutation has prognostic value in patients with treatment-naive chronic lymphocytic leukemia.
- Author
-
Syrykh C, Pons-Brun B, Russiñol N, Playa-Albinyana H, Baumann T, Duran-Ferrer M, Kulis M, Carbó-Meix A, Mozas P, Alcoceba M, González M, Navarro-Bailón A, Colado E, Payer ÁR, Aymerich M, Terol MJ, Lu J, Knisbacher BA, Hahn CK, Ruiz-Gaspà S, Enjuanes A, Wu CJ, Getz G, Zenz T, López-Guillermo A, Martín-Subero JI, Colomer D, Delgado J, Campo E, and Nadeu F
- Subjects
- Humans, Prognosis, Mutation, Immunoglobulin Heavy Chains genetics, Leukemia, Lymphocytic, Chronic, B-Cell diagnosis, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Published
- 2023
- Full Text
- View/download PDF
18. Characterization of Three Somatic Mutations in the 3'UTR of RRAS2 and Their Inverse Correlation with Lymphocytosis in Chronic Lymphocytic Leukemia.
- Author
-
Lacuna M, Hortal AM, Cifuentes C, Gonzalo T, Alcoceba M, Bastos M, Bustelo XR, González M, and Alarcón B
- Subjects
- Humans, 3' Untranslated Regions genetics, B-Lymphocytes pathology, Mutation genetics, Membrane Proteins genetics, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Lymphocytosis genetics, Lymphocytosis diagnosis, Lymphocytosis pathology, Monomeric GTP-Binding Proteins genetics
- Abstract
Chronic lymphocytic leukemia (CLL) is a hematologic malignancy characterized by progressive accumulation of a rare population of CD5+ B-lymphocytes in peripheral blood, bone marrow, and lymphoid tissues. CLL exhibits remarkable clinical heterogeneity, with some patients presenting with indolent disease and others progressing rapidly to aggressive CLL. The significant heterogeneity of CLL underscores the importance of identifying novel prognostic markers. Recently, the RAS-related gene RRAS2 has emerged as both a driver oncogene and a potential marker for CLL progression, with higher RRAS2 expression associated with poorer disease prognosis. Although missense somatic mutations in the coding sequence of RRAS2 have not been described in CLL, this study reports the frequent detection of three somatic mutations in the 3' untranslated region (3'UTR) affecting positions +26, +53, and +180 downstream of the stop codon in the mRNA. An inverse relationship was observed between these three somatic mutations and RRAS2 mRNA expression, which correlated with lower blood lymphocytosis. These findings highlight the importance of RRAS2 overexpression in CLL development and prognosis and point to somatic mutations in its 3'UTR as novel mechanistic clues. Our results may contribute to the development of targeted therapeutic strategies and improved risk stratification for CLL patients.
- Published
- 2023
- Full Text
- View/download PDF
19. Other malignancies in the history of CLL: an international multicenter study conducted by ERIC, the European Research Initiative on CLL, in HARMONY.
- Author
-
Chatzikonstantinou T, Scarfò L, Karakatsoulis G, Minga E, Chamou D, Iacoboni G, Kotaskova J, Demosthenous C, Smolej L, Mulligan S, Alcoceba M, Al-Shemari S, Aurran-Schleinitz T, Bacchiarri F, Bellido M, Bijou F, Calleja A, Medina A, Khan MA, Cassin R, Chatzileontiadou S, Collado R, Christian A, Davis Z, Dimou M, Donaldson D, Santos GD, Dreta B, Efstathopoulou M, El-Ashwah S, Enrico A, Fresa A, Galimberti S, Galitzia A, García-Serra R, Gimeno E, González-Gascón-Y-Marín I, Gozzetti A, Guarente V, Guieze R, Gogia A, Gupta R, Harrop S, Hatzimichael E, Herishanu Y, Hernández-Rivas JÁ, Inchiappa L, Jaksic O, Janssen S, Kalicińska E, Laribi K, Karakus V, Kater AP, Kho B, Kislova M, Konstantinou E, Koren-Michowitz M, Kotsianidis I, Kreitman RJ, Labrador J, Lad D, Levin MD, Levy I, Longval T, Lopez-Garcia A, Marquet J, Martin-Rodríguez L, Maynadié M, Maslejova S, Mayor-Bastida C, Mihaljevic B, Milosevic I, Miras F, Moia R, Morawska M, Murru R, Nath UK, Navarro-Bailón A, Oliveira AC, Olivieri J, Oscier D, Panovska-Stavridis I, Papaioannou M, Papajík T, Kubova Z, Phumphukhieo P, Pierie C, Puiggros A, Rani L, Reda G, Rigolin GM, Ruchlemer R, Daniel de Deus Santos M, Schipani M, Schiwitza A, Shen Y, Simkovic M, Smirnova S, Abdelrahman Soliman DS, Spacek M, Tadmor T, Tomic K, Tse E, Vassilakopoulos T, Visentin A, Vitale C, von Tresckow J, Vrachiolias G, Vukovic V, Walewska R, Wasik-Szczepanek E, Xu Z, Yagci M, Yañez L, Yassin M, Zuchnicka J, Angelopoulou M, Antic D, Biderman B, Catherwood M, Claus R, Coscia M, Cuneo A, Demirkan F, Espinet B, Gaidano G, Kalashnikova OB, Laurenti L, Nikitin E, Pangalis GA, Panagiotidis P, Popov VM, Pospisilova S, Sportoletti P, Stavroyianni N, Tam C, Trentin L, Chatzidimitriou A, Bosch F, Doubek M, Ghia P, and Stamatopoulos K
- Abstract
Background: Patients with chronic lymphocytic leukemia (CLL) have a higher risk of developing other malignancies (OMs) compared to the general population. However, the impact of CLL-related risk factors and CLL-directed treatment is still unclear and represents the focus of this work., Methods: We conducted a retrospective international multicenter study to assess the incidence of OMs and detect potential risk factors in 19,705 patients with CLL, small lymphocytic lymphoma, or high-count CLL-like monoclonal B-cell lymphocytosis, diagnosed between 2000 and 2016. Data collection took place between October 2020 and March 2022., Findings: In 129,254 years of follow-up after CLL diagnosis, 3513 OMs were diagnosed (27.2 OMs/1000 person-years). The most common hematological OMs were Richter transformation, myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Non-melanoma skin (NMSC) and prostate cancers were the most common solid tumors (STs).The only predictor for MDS and AML development was treatment with fludarabine and cyclophosphamide with/without rituximab (FC ± R) (OR = 3.7; 95% CI = 2.79-4.91; p < 0.001). STs were more frequent in males and patients with unmutated immunoglobulin heavy variable genes (OR = 1.77; 95% CI = 1.49-2.11; p < 0.001/OR = 1.89; 95% CI = 1.6-2.24; p < 0.001).CLL-directed treatment was associated with non-melanoma skin and prostate cancers (OR = 1.8; 95% CI = 1.36-2.41; p < 0.001/OR = 2.11; 95% CI = 1.12-3.97; p = 0.021). In contrast, breast cancers were more frequent in untreated patients (OR = 0.17; 95% CI = 0.08-0.33; p < 0.001).Patients with CLL and an OM had inferior overall survival (OS) than those without. AML and MDS conferred the worst OS (p < 0.001)., Interpretation: OMs in CLL impact on OS. Treatment for CLL increased the risk for AML/MDS, prostate cancer, and NMSC. FCR was associated with increased risk for AML/MDS., Funding: AbbVie, and EU/EFPIAInnovative Medicines Initiative Joint Undertaking HARMONY grant n° 116026., Competing Interests: TC received honoraria from AbbVie. LS received consulting fees from AbbVie, BeiGene, AstraZeneca, Lilly, and Janssen, honoraria from Janssen and Octapharma, support for attending meetings from BeiGene and Janssen and advisory board fees from Merck. SMu received advisory board fees from AbbVie, AstraZeneca, Janssen, Roche and BeiGene. JAHR received honoraria as a consultant from Janssen, AbbVie, AstraZeneca, Lilly, and BeiGene and support for attending meetings from Janssen, AbbVie, AstraZeneca, and BeiGene. LI received honoraria from AbbVie, Roche, and Janssen-Cilag. APK received advisory board fees and research money from Janssen, AbbVie, BMS, AstraZeneca, Roche/Genentech, support for attending meetings from Janssen and AbbVie. M-DL received travel expenses from Janssen and AbbVie. GMR received honoraria for participation to speaker's bureau from AbbVie, Astra Zeneca, Beigene, and Janssen, and support for attending meetings from Janssen. JVT received consulting fees from AbbVie, AstraZeneca, BeiGene, and Janssen, honoraria for scientific talks from AbbVie, AstraZeneca, BeiGene, Janssen, Lilly, and Roche, travel support from AbbVie, AstraZeneca, BeiGene, Janssen, Roche, and Lilly, and advisory boards fees for AbbVie, Amgen, AstraZeneca, BeiGene. GI received honoraria from Novartis, BMS, Sandoz, AstraZeneca, Janssen, Kite/Gilead, and Miltenyi, support for attending meetings from Kite/Gilead, AstraZeneca, and AbbVie and advisory board fees from Kite/Gilead, Novartis, BMS, and Autolus. FBi received support for attending meetings from AbbVie. RCo received support for attending meetings from Janssen-Cilag and S.A. SG received honoraria support for attending meetings from AbbVie, AstraZeneca, Jazz, Novartis, and Incyte, honoraria from Roche, Celgene, Pfizer, and Janssen, and support for attending meetings from Jazz, AstraZeneca, and Roche. RGS received support for attending meetings from AbbVie and S.L.U. RG received honoraria, consulting fees, and support for attending meetings from AbbVie, Beigene, Roche, Janssen, AstraZeneca. YH received honoraria from Janssen, AbbVie, Roche, AstraZeneca, Medion, and Lilly. OJ received honoraria from Johnson and Johnson, AstraZeneca, and Lilly, honoraria from Johnson and Johnson, AbbVie, AstraZeneca, and Lilly, and support for attending meetings from Johnson and Johnson, and AbbVie. LK received consulting fees from AbbVie, AstraZeneca, Janssen, Beigene, Takeda, and Novartis. MKM received honoraria from Novartis, Pfizer, and Gad Medical LTD and support for attending meetings from Novartis. IKo received honoraria and consulting fees from AbbVie and Janssen. IM received honoraria from AbbVie, Roche, Sandoz, AstraZeneca, and Janssen, and support for attending meetings from AbbVie, Roche, and Takeda. ANB received honoraria, advisory board fees and support for attending meetings from AbbVie, AstraZeneca, Takeda, Janssen, and Beigene. JO received honoraria from AbbVie, AstraZeneca, and Janssen. GR received consulting fees from AbbVie, AstraZeneca, Janssen, and Beigene, and is currently employed by AstraZeneca. TP received honoraria and advisory board fees from AbbVie, Janssen-Cilag, and AstraZeneca and support for attending meeting from AstraZeneca. MS received honoraria and support for attending meeting from AstraZeneca, AbbVie, and Janssen-Cilag and owns shares of stock in AbbVie, AstraZeneca, Merck, Eli Lilly, Sanofi, Johnson and Johnson, Pfizer, Gilead, and GSK. MSp received honoraria and consulting and advisory board fees, and support for attending meeting from AbbVie, AstraZeneca, and Janssen. ET received support for attending meetings from Takeda. TV received honoraria from Takeda, Roche, Genesis Pharma, Merck, Novartis, Gilead, Sandoz, AstraZeneca, Integris, and Servier and support for attending meetings from Takeda, Roche, Genesis Pharma, Merck, Pfizer, and Winmedica. CV received honoraria from AbbVie, consulting fees from AstraZeneca and support for attending meeting from AstraZeneca, Takeda, and Janssen. RW received honoraria from AbbVie, AstraZeneca, and Beigene, support for attending meetings from Janssen, AbbVie, and AstraZeneca and advisory board fees from AbbVie, AstraZeneca, Janssen, Beigene, and SecuraBio. EWS received honoraria from AbbVie, Roche, and Janssen-Cilag and support for attending meetings from AbbVie. LY received honoraria from AbbVie, AstraZeneca, Novartis, Gilead, Janssen, Jazz, MSD, and Pfizer, support for attending meetings from AbbVie, AstraZeneca, Gilead, Janssen, and Pfizer, and advisory board fees from AbbVie, AstraZeneca, Jazz, Janssen, Beigene, and Celgene. MAn received consulting fees from AbbVie, Takeda, Janssen, Roche, Genesis, Gilead, and Amgen and honoraria from AbbVie, Takeda, Roche, Genesis, Gilead, and Novartis. MC received honoraria, advisory board fees, and support for attending meetings from AbbVie, AstraZeneca, and Janssen. ACu received honoraria, advisory board fees, and support for attending meetings from AbbVie, AstraZeneca, Beigene, Janssen, and Lilly. FD support for attending meetings from Janssen and AbbVie. GG received honoraria, and advisory board fees from AbbVie, AstraZeneca, Beigene, Janssen and advisory board fees from Lilly. EN received honoraria from AbbVie. LSm received consulting fees, honoraria and support for attending meetings from AbbVie, AstraZeneca, and Janssen and advisory board fees from AbbVie and AstraZeneca. NS received honoraria from Janssen, AbbVie, AstraZeneca, and Lilly, and support for attending meetings from Janssen and AstraZeneca. CT received honoraria from AbbVie, Beigene, Janssen, and LOXO. FB received consulting fees, honoraria and payment for expert testimony from AbbVie, Genentech, Novartis, Takeda, Janssen, Roche, Mundipharma, Celgene/BMS, AstraZeneca, Lilly, Beigene, Gilead and TG Therapeutics, Advantage Allogene, Lava Therapeutics, and Enterome. MDo received honoraria and advisory board fees from AbbVie, AstraZeneca and Janssen, advisory board fees from Swixx, and support for attending meetings from Janssen. PG received honoraria and consulting fees from AbbVie, AstraZeneca, BMS, Janssen, Lilly/Loxo Oncology, MSD, and Roche; grant support from AbbVie, AstraZeneca, BMS, Janssen. KS received honoraria from Janssen, AbbVie, Lilly and AstraZeneca, consulting fees and support for attending meetings from Janssen and AstraZeneca. GK, EM, DC, JK, CD, MA, SA, TAS, FBa, MB, ACa, AM, AKM, RC, SC, ACh, ZD, MDi, DD, GDS, BD, ME, SEA, AE, AF, AG, EG, IGGM, AGo, AjG, RGu, SH, EH, SJ, EKa, VK, BK, MK, EK, RJK, JL, DL, IL, TL, ALG, JM, LMR, MMa, SM, CMB, BM, FM, RM, MMo, RMu, UKN, ACO, DO, IPS, MP, ZK, PP, CP, AP, LR, RR, MDDS, AS, YS, MSi, SS, DSAS, TT, KT, AV, GV, VV, ZX, MYa, MY, JZ, DA, BB, MCa, RCl, BE, OBK, LL, GP, PPa, VMP, SP, PS, LT, AC, have no conflict of interest to disclose., (© 2023 Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
20. Clonal architecture and evolutionary history of Waldenström's macroglobulinemia at the single-cell level.
- Author
-
García-Sanz R, García-Álvarez M, Medina A, Askari E, González-Calle V, Casanova M, de la Torre-Loizaga I, Escalante-Barrigón F, Bastos-Boente M, Bárez A, Vidaña-Bedera N, Alonso JM, Sarasquete ME, González M, Chillón MC, Alcoceba M, and Jiménez C
- Subjects
- Humans, Single-Cell Analysis, Protein Array Analysis, Myeloid Differentiation Factor 88 genetics, DNA Mutational Analysis, Waldenstrom Macroglobulinemia diagnosis, Waldenstrom Macroglobulinemia genetics, Clonal Evolution, Mutation, DNA Copy Number Variations
- Abstract
To provide insight into the subclonal architecture and co-dependency patterns of the alterations in Waldenström's macroglobulinemia (WM), we performed single-cell mutational and protein profiling of eight patients. A custom panel was designed to screen for mutations and copy number alterations at the single-cell level in samples taken from patients at diagnosis (n=5) or at disease progression (n=3). Results showed that in asymptomatic WM at diagnosis, MYD88L265P was the predominant clonal alteration; other events, if present, were secondary and subclonal to MYD88L265P. In symptomatic WM, clonal diversity was more evident, uncovering combinations of alterations that synergized to promote clonal expansion and dominance. At disease progression, a dominant clone was observed, sometimes accompanied by other less complex minor clones, which could be consistent with a clonal selection process. Clonal diversity was also reduced, probably due to the effect of treatment. Finally, we combined protein expression with mutational analysis to map somatic genotype with the immunophenotype. Our findings provide a comprehensive view of the clonality of tumor populations in WM and how clonal complexity can evolve and impact disease progression., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
21. Unraveling the genetics of transformed splenic marginal zone lymphoma.
- Author
-
Grau M, López C, Navarro A, Frigola G, Nadeu F, Clot G, Bastidas-Mora G, Alcoceba M, Baptista MJ, Blanes M, Colomer D, Costa D, Domingo-Domènech E, Enjuanes A, Escoda L, Forcada P, Giné E, Lopez-Guerra M, Ramón O, Rivas-Delgado A, Vicente Folch L, Wotherspoon A, Climent F, Campo E, López-Guillermo A, Matutes E, and Beà S
- Subjects
- Humans, Mutation, Translocation, Genetic, Splenic Neoplasms genetics, Splenic Neoplasms diagnosis, Splenic Neoplasms pathology, Lymphoma, Large B-Cell, Diffuse genetics, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
The genetic mechanisms associated with splenic marginal zone lymphoma (SMZL) transformation are not well defined. We studied 41 patients with SMZL that eventually underwent large B-cell lymphoma transformation. Tumor material was obtained either only at diagnosis (9 patients), at diagnosis and transformation (18 patients), and only at transformation (14 patients). Samples were categorized in 2 groups: (1) at diagnosis (SMZL, n = 27 samples), and (2) at transformation (SMZL-T, n = 32 samples). Using copy number arrays and a next-generation sequencing custom panel, we identified that the main genomic alterations in SMZL-T involved TNFAIP3, KMT2D, TP53, ARID1A, KLF2, 1q gains, and losses of 9p21.3 (CDKN2A/B) and 7q31-q32. Compared with SMZL, SMZL-T had higher genomic complexity, and higher incidence of TNFAIP3 and TP53 alterations, 9p21.3 (CDKN2A/B) losses, and 6p gains. SMZL and SMZL-T clones arose by divergent evolution from a common altered precursor cell that acquired different genetic alterations in virtually all evaluable cases (92%, 12 of 13 cases). Using whole-genome sequencing of diagnostic and transformation samples in 1 patient, we observed that the SMZL-T sample carried more genomic aberrations than the diagnostic sample, identified a translocation t(14;19)(q32;q13) present in both samples, and detected a focal B2M deletion due to chromothripsis acquired at transformation. Survival analysis showed that KLF2 mutations, complex karyotype, and International Prognostic Index score at transformation were predictive of a shorter survival from transformation (P = .001; P = .042; and P = .007; respectively). In summary, SMZL-T are characterized by higher genomic complexity than SMZL, and characteristic genomic alterations that could represent key players in the transformation event., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. Primary Cutaneous Marginal Zone Lymphoproliferative Disorder of Donor Origin after Allogeneic Hematopoietic Stem Cell Transplantation.
- Author
-
Bejarano L, Sayagués JM, Alcoceba M, Balanzategui A, Lopez-Corral L, Cañueto J, and Santos-Briz A
- Subjects
- Humans, Female, Adult, In Situ Hybridization, Fluorescence, Plasma Cells pathology, Lymphoproliferative Disorders etiology, Lymphoproliferative Disorders pathology, Hematopoietic Stem Cell Transplantation adverse effects, Skin Diseases, Epstein-Barr Virus Infections pathology
- Abstract
Abstract: Primary cutaneous posttransplant lymphoproliferative disorders (PTLDs) after allogeneic hematopoietic stem cell transplant (allo-HSCT) are exceedingly rare, with only 6 published cases, all of them consisting in T-cell neoplasms. In this report, we present for the first time a donor-derived B-cell PTLD consisting in a primary, cutaneous, B-cell, marginal zone, lymphoproliferative disorder (PCMZLPD). The patient, a 37-year-old woman with a history of Hodgkin lymphoma received an allo-HSCT from her healthy, matched, related father, achieving complete host chimerism in the bone marrow and peripheral blood. However, 8 years after the allo-HSCT, she presented asymptomatic skin lesions consisting in oval, well-defined, slightly raised erythematous plaques, located on the arms, trunk, and legs. Skin biopsies of 2 lesions demonstrated a class-switched IgG+, EBV-, PCMZLPD, showing kappa light chain restriction and monoclonal rearrangement of the IgH gene. Microsatellite genotyping and 2-color fluorescence in situ hybridization (X and Y chromosomes) confirmed that the origin of the neoplastic cells was the donor graft. The lesions showed an indolent behavior, good response to topical corticosteroids, and no need for systemic treatment. Our case broadens the spectrum of PTLD, a diverse group of lymphoid and/or plasmacytic proliferations with variable clinical presentations and histopathological features., Competing Interests: The authors declare no conflicts of interest., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
23. Do GWAS-Identified Risk Variants for Chronic Lymphocytic Leukemia Influence Overall Patient Survival and Disease Progression?
- Author
-
Cabrera-Serrano AJ, Sánchez-Maldonado JM, Ter Horst R, Macauda A, García-Martín P, Benavente Y, Landi S, Clay-Gilmour A, Niazi Y, Espinet B, Rodríguez-Sevilla JJ, Pérez EM, Maffei R, Blanco G, Giaccherini M, Cerhan JR, Marasca R, López-Nevot MÁ, Chen-Liang T, Thomsen H, Gámez I, Campa D, Moreno V, de Sanjosé S, Marcos-Gragera R, García-Álvarez M, Dierssen-Sotos T, Jerez A, Butrym A, Norman AD, Luppi M, Slager SL, Hemminki K, Li Y, Berndt SI, Casabonne D, Alcoceba M, Puiggros A, Netea MG, Försti A, Canzian F, and Sainz J
- Subjects
- Adult, Humans, Genome-Wide Association Study, Risk Factors, Disease Progression, Genetic Predisposition to Disease, Polymorphism, Single Nucleotide, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia among adults worldwide. Although genome-wide association studies (GWAS) have uncovered the germline genetic component underlying CLL susceptibility, the potential use of GWAS-identified risk variants to predict disease progression and patient survival remains unexplored. Here, we evaluated whether 41 GWAS-identified risk variants for CLL could influence overall survival (OS) and disease progression, defined as time to first treatment (TTFT) in a cohort of 1039 CLL cases ascertained through the CRuCIAL consortium. Although this is the largest study assessing the effect of GWAS-identified susceptibility variants for CLL on OS, we only found a weak association of ten single nucleotide polymorphisms (SNPs) with OS ( p < 0.05) that did not remain significant after correction for multiple testing. In line with these results, polygenic risk scores (PRSs) built with these SNPs in the CRuCIAL cohort showed a modest association with OS and a low capacity to predict patient survival, with an area under the receiver operating characteristic curve (AUROC) of 0.57. Similarly, seven SNPs were associated with TTFT ( p < 0.05); however, these did not reach the multiple testing significance threshold, and the meta-analysis with previous published data did not confirm any of the associations. As expected, PRSs built with these SNPs showed reduced accuracy in prediction of disease progression (AUROC = 0.62). These results suggest that susceptibility variants for CLL do not impact overall survival and disease progression in CLL patients.
- Published
- 2023
- Full Text
- View/download PDF
24. Detection and characterization of the novel HLA-DPA1*02:66:02N allele, with a premature stop codon in exon 2.
- Author
-
Niño Ramírez JE, Balas A, Gil-Etayo FJ, Jiménez Hernaz I, Terradillos Sánchez P, Vicente Parra A, Balanzategui A, Alcoceba M, García Sanz R, and Tejeda Velarde A
- Subjects
- Humans, Alleles, Exons genetics, Codon, Histocompatibility Testing methods, Codon, Nonsense, HLA-DP alpha-Chains genetics
- Abstract
The failure to identify HLA null alleles in bone marrow transplantation could be life-threatening because this could result in an HLA mismatch with the ability to trigger the graft-vs-host disease (GVHD) and to reduce patient's survival. In this report we describe the identification and characterization of the novel HLA-DPA1*02:66:02N allele with a non-sense codon in exon 2. This new allele was discovered in two unrelated bone marrow donors during routine HLA-typing using next-generation sequencing (NGS). DPA1*02:66:02N is homologous to DPA1*02:01:01:03 with a single nucleotide difference in exon 2, codon 50, where the replacement of C located at genomic position 3825 by T, causes the formation of a premature stop codon (TGA), resulting in a null allele. This description illustrates the benefits of HLA typing by NGS since it permits to reduce ambiguities, identify new alleles, analyze multiple HLA loci and improve transplantation outcome., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 American Society for Histocompatibility and Immunogenetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. Systematic Evaluation of Antigenic Stimulation in Chronic Lymphocytic Leukemia: Humoral Immunity as Biomarkers for Disease Evolution.
- Author
-
Landeira-Viñuela A, Alcoceba-Sanchez M, Navarro-Bailón A, Arias-Hidalgo C, Juanes-Velasco P, Sánchez-Santos JM, Lecrevisse Q, Pedreira CE, García-Vaquero ML, Hernández ÁP, Montalvillo E, Góngora R, De Las Rivas J, González-Díaz M, Orfao A, and Fuentes M
- Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in the Western world. Studies of CLL antibody reactivity have shown differential targets to autoantigens and antimicrobial molecular motifs that support the current hypothesis of CLL pathogenesis., Methods: In this study, we conducted a quantitative serum analysis of 7 immunoglobulins in CLL and monoclonal B-cell lymphocytosis (MBL) patients (bead-suspension protein arrays) and a serological profile (IgG and IgM) study of autoantibodies and antimicrobial antigens (protein microarrays)., Results: Significant differences in the IgA levels were observed according to disease progression and evolution as well as significant alterations in IgG1 according to IGHV mutational status. More representative IgG autoantibodies in the cohort were against nonmutagenic proteins and IgM autoantibodies were against vesicle proteins. Antimicrobial IgG and IgM were detected against microbes associated with respiratory tract infections., Conclusions: Quantitative differences in immunoglobulin serum levels could be potential biomarkers for disease progression. In the top 5 tumoral antigens, we detected autoantibodies (IgM and IgG) against proteins related to cell homeostasis and metabolism in the studied cohort. The top 5 microbial antigens were associated with respiratory and gastrointestinal infections; moreover, the subsets with better prognostics were characterized by a reactivation of Cytomegalovirus. The viral humoral response could be a potential prognosis biomarker for disease progression.
- Published
- 2023
- Full Text
- View/download PDF
26. An Optimized Single Nucleotide Polymorphism-Based Detection Method Suggests That Allelic Variants in the 3' Untranslated Region of RRAS2 Correlate with Treatment Response in Chronic Lymphocytic Leukemia Patients.
- Author
-
Hortal A, Lacuna M, Cifuentes C, Alcoceba M, Bustelo XR, González M, and Alarcón B
- Abstract
Unlike classical RAS genes, oncogenic mutations on RRAS2 are seldomly found in human cancer. By contrast, RRAS2 is frequently found overexpressed in a number of human tumors, including B and T cell lymphomas, breast, gastric, head and neck cancers. In this regard, we have recently shown that overexpression of wild-type RRAS2 drives the formation of both chronic lymphocytic leukemia (CLL) and breast cancer in mice. In support for the relevance of overexpression of wild type RRAS2 in human cancer, we have found that RRAS2 expression is influenced by the presence of a specific single nucleotide polymorphism (SNP) located in the 3'-untranslated region (UTR) of the RRAS2 mRNA. Perhaps more importantly, the presence of the alternate C, rather than the G allele, at the RRAS2 SNP designated as rs8570 is also associated with worse patient prognosis in CLL. This indicates that the detection of this SNP allelic variants can be informative to predict RRAS2 expression levels and disease long-term evolution in patients. Here, we describe a polymerase chain reaction (PCR)-based method that facilitates the rapid and easy determination of G and C allelic variants of the SNP. Using this approach, we confirm that the C allelic variant is associated with higher expression levels of RRAS2 transcripts and poor patient prognosis. However, we have also found that expression of the C allelic variants correlates with better response to ibrutinib, a Bruton kinase inhibitor commonly used in CLL treatments. This suggests that this method for detecting the RRAS2 rs8570 SNP might be a useful as a tool to predict both patient prognosis and response to targeted therapy in CLL.
- Published
- 2023
- Full Text
- View/download PDF
27. Genetic and phenotypic characterisation of HIV-associated aggressive B-cell non-Hodgkin lymphomas, which do not occur specifically in this population: diagnostic and prognostic implications.
- Author
-
Baptista MJ, Tapia G, Muñoz-Marmol AM, Muncunill J, Garcia O, Montoto S, Gribben JG, Calaminici M, Martinez A, Veloza L, Martínez-Trillos A, Aldamiz T, Menarguez J, Terol MJ, Ferrandez A, Alcoceba M, Briones J, González-Barca E, Climent F, Muntañola A, Moraleda JM, Provencio M, Abrisqueta P, Abella E, Colomo L, García-Ballesteros C, Garcia-Caro M, Sancho JM, Ribera JM, Mate JL, and Navarro JT
- Subjects
- Humans, Prognosis, Gene Rearrangement, Chromosome Aberrations, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-bcl-6 genetics, Lymphoma, Large B-Cell, Diffuse diagnosis, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Burkitt Lymphoma genetics, HIV Infections diagnosis
- Abstract
The frequency of aggressive subtypes of B-cell non-Hodgkin lymphoma (B-NHL), such as high-grade B-cell lymphomas (HGBL) with MYC and BCL2 and/or BCL6 rearrangement (HGBL-DH/TH) or Burkitt-like lymphoma (BL) with 11q aberration, is not well known in the HIV setting. We aimed to characterise HIV-associated aggressive B-NHL according to the 2017 WHO criteria, and to identify genotypic and phenotypic features with prognostic impact. Seventy-five HIV-associated aggressive B-NHL were studied by immunohistochemistry (CD10, BCL2, BCL6, MUM1, MYC, and CD30), EBV-encoded RNAs (EBERs), and fluorescence in situ hybridisation (FISH) to evaluate the status of the MYC, BCL2, and BCL6 genes and chromosome 11q. The 2017 WHO classification criteria and the Hans algorithm, for the cell-of-origin classification of diffuse large B-cell lymphomas (DLBCL), were applied. In DLBCL cases, the frequencies of MYC and BCL6 rearrangements (14.9 and 27.7%, respectively) were similar to those described in HIV-negative patients, but BCL2 rearrangements were infrequent (4.3%). MYC expression was identified in 23.4% of DLBCL cases, and coexpression of MYC and BCL2 in 13.0%, which was associated with a worse prognosis. As for BL cases, the expression of MUM1 (30.4%) conferred a worse prognosis. Finally, the prevalence of HGBL-DH/TH and BL-like with 11q aberration are reported in the HIV setting. The phenotypic and genotypic characteristics of HIV-associated aggressive B-NHL are similar to those of the general population, except for the low frequency of BCL2 rearrangements in DLBCL. MYC and BCL2 coexpression in DLBCL, and MUM-1 expression in BL, have a negative prognostic impact on HIV-infected individuals., (© 2022 The Authors. Histopathology published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
28. A gene expression assay based on chronic lymphocytic leukemia activation in the microenvironment to predict progression.
- Author
-
Abrisqueta P, Medina D, Villacampa G, Lu J, Alcoceba M, Carabia J, Boix J, Tazón-Vega B, Iacoboni G, Bobillo S, Marín-Niebla A, González M, Zenz T, Crespo M, and Bosch F
- Subjects
- Humans, Prognosis, Mutation, Proportional Hazards Models, Transcriptome, Tumor Microenvironment genetics, Leukemia, Lymphocytic, Chronic, B-Cell diagnosis, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell therapy
- Abstract
Several gene expression profiles with a strong correlation with patient outcomes have been previously described in chronic lymphocytic leukemia (CLL), although their applicability as biomarkers in clinical practice has been particularly limited. Here we describe the training and validation of a gene expression signature for predicting early progression in patients with CLL based on the analysis of 200 genes related to microenvironment signaling on the NanoString platform. In the training cohort (n = 154), the CLL15 assay containing a 15-gene signature was associated with the time to first treatment (TtFT) (hazard ratio [HR], 2.83; 95% CI, 2.17-3.68; P < .001). The prognostic value of the CLL15 score (HR, 1.71; 95% CI, 1.15-2.52; P = .007) was further confirmed in an external independent validation cohort (n = 112). Notably, the CLL15 score improved the prognostic capacity over IGHV mutational status and the International Prognostic Score for asymptomatic early-stage (IPS-E) CLL. In multivariate analysis, the CLL15 score (HR, 1.83; 95% CI, 1.32-2.56; P < .001) and the IPS-E CLL (HR, 2.23; 95% CI, 1.59-3.12; P < .001) were independently associated with TtFT. The newly developed and validated CLL15 assay successfully translated previous gene signatures such as the microenvironment signaling into a new gene expression-based assay with prognostic implications in CLL., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Unravelling soluble immune checkpoints in chronic lymphocytic leukemia: Physiological immunomodulators or immune dysfunction.
- Author
-
Landeira-Viñuela A, Arias-Hidalgo C, Juanes-Velasco P, Alcoceba M, Navarro-Bailón A, Pedreira CE, Lecrevisse Q, Díaz-Muñoz L, Sánchez-Santos JM, Hernández ÁP, García-Vaquero ML, Góngora R, De Las Rivas J, González M, Orfao A, and Fuentes M
- Subjects
- Biomarkers, Chemokine CXCL10, Endothelial Cells pathology, Humans, Immunoglobulin Heavy Chains genetics, Immunologic Factors, Interleukin-4, Tumor Microenvironment, Leukemia, Lymphocytic, Chronic, B-Cell
- Abstract
Chronic lymphocytic leukemia (CLL) is a lymphoid neoplasm characterized by the accumulation of mature B cells. The diagnosis is established by the detection of monoclonal B lymphocytes in peripheral blood, even in early stages [monoclonal B-cell lymphocytosis (MBL
hi )], and its clinical course is highly heterogeneous. In fact, there are well-characterized multiple prognostic factors that are also related to the observed genetic heterogenicity, such as immunoglobulin heavy chain variable region (IGHV) mutational status, del17p, and TP53 mutations, among others. Moreover, a dysregulation of the immune system (innate and adaptive immunity) has been observed in CLL patients, with strong impact on immune surveillance and consequently on the onset, evolution, and therapy response. In addition, the tumor microenvironment is highly complex and heterogeneous (i.e., matrix, fibroblast, endothelial cells, and immune cells), playing a critical role in the evolution of CLL. In this study, a quantitative profile of 103 proteins (cytokines, chemokines, growth/regulatory factors, immune checkpoints, and soluble receptors) in 67 serum samples (57 CLL and 10 MBLhi ) has been systematically evaluated. Also, differential profiles of soluble immune factors that discriminate between MBLhi and CLL (sCD47, sCD27, sTIMD-4, sIL-2R, and sULBP-1), disease progression (sCD48, sCD27, sArginase-1, sLAG-3, IL-4, and sIL-2R), or among profiles correlated with other prognostic factors, such as IGHV mutational status (CXCL11/I-TAC, CXCL10/IP-10, sHEVM, and sLAG-3), were deciphered. These results pave the way to explore the role of soluble immune checkpoints as a promising source of biomarkers in CLL, to provide novel insights into the immune suppression process and/or dysfunction, mostly on T cells, in combination with cellular balance disruption and microenvironment polarization leading to tumor escape., 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 Landeira-Viñuela, Arias-Hidalgo, Juanes-Velasco, Alcoceba, Navarro-Bailón, Pedreira, Lecrevisse, Díaz-Muñoz, Sánchez-Santos, Hernández, García-Vaquero, Góngora, De Las Rivas, González, Orfao and Fuentes.)- Published
- 2022
- Full Text
- View/download PDF
30. Detection of early seeding of Richter transformation in chronic lymphocytic leukemia.
- Author
-
Nadeu F, Royo R, Massoni-Badosa R, Playa-Albinyana H, Garcia-Torre B, Duran-Ferrer M, Dawson KJ, Kulis M, Diaz-Navarro A, Villamor N, Melero JL, Chapaprieta V, Dueso-Barroso A, Delgado J, Moia R, Ruiz-Gil S, Marchese D, Giró A, Verdaguer-Dot N, Romo M, Clot G, Rozman M, Frigola G, Rivas-Delgado A, Baumann T, Alcoceba M, González M, Climent F, Abrisqueta P, Castellví J, Bosch F, Aymerich M, Enjuanes A, Ruiz-Gaspà S, López-Guillermo A, Jares P, Beà S, Capella-Gutierrez S, Gelpí JL, López-Bigas N, Torrents D, Campbell PJ, Gut I, Rossi D, Gaidano G, Puente XS, Garcia-Roves PM, Colomer D, Heyn H, Maura F, Martín-Subero JI, and Campo E
- Subjects
- Cell Transformation, Neoplastic genetics, Disease Progression, Humans, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology
- Abstract
Richter transformation (RT) is a paradigmatic evolution of chronic lymphocytic leukemia (CLL) into a very aggressive large B cell lymphoma conferring a dismal prognosis. The mechanisms driving RT remain largely unknown. We characterized the whole genome, epigenome and transcriptome, combined with single-cell DNA/RNA-sequencing analyses and functional experiments, of 19 cases of CLL developing RT. Studying 54 longitudinal samples covering up to 19 years of disease course, we uncovered minute subclones carrying genomic, immunogenetic and transcriptomic features of RT cells already at CLL diagnosis, which were dormant for up to 19 years before transformation. We also identified new driver alterations, discovered a new mutational signature (SBS-RT), recognized an oxidative phosphorylation (OXPHOS)
high -B cell receptor (BCR)low -signaling transcriptional axis in RT and showed that OXPHOS inhibition reduces the proliferation of RT cells. These findings demonstrate the early seeding of subclones driving advanced stages of cancer evolution and uncover potential therapeutic targets for RT., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
31. Interlaboratory Analytical Validation of a Next-Generation Sequencing Strategy for Clonotypic Assessment and Minimal Residual Disease Monitoring in Multiple Myeloma.
- Author
-
Medina A, Jiménez C, Puig N, Sarasquete ME, Flores-Montero J, García-Álvarez M, Prieto-Conde I, Chillón C, Alcoceba M, González-Calle V, Gutiérrez NC, Jacobsen A, Vigil E, Hutt K, Huang Y, Orfao A, González M, Miller J, and García-Sanz R
- Subjects
- Humans, High-Throughput Nucleotide Sequencing methods, Neoplasm, Residual diagnosis, Neoplasm, Residual genetics, Reproducibility of Results, Multiple Myeloma diagnosis, Multiple Myeloma genetics
- Abstract
Context.—: Minimal residual disease (MRD) is a major prognostic factor in multiple myeloma, although validated technologies are limited., Objective.—: To standardize the performance of the LymphoTrack next-generation sequencing (NGS) assays (Invivoscribe), targeting clonal immunoglobulin rearrangements, in order to reproduce the detection of tumor clonotypes and MRD quantitation in myeloma., Design.—: The quantification ability of the assay was evaluated through serial dilution experiments. Paired samples from 101 patients were tested by LymphoTrack, using Sanger sequencing and EuroFlow's next-generation flow (NGF) assay as validated references for diagnostic and follow-up evaluation, respectively. MRD studies using LymphoTrack were performed in parallel at 2 laboratories to evaluate reproducibility., Results.—: Sensitivity was set as 1.3 tumor cells per total number of input cells. Clonality was confirmed in 99% and 100% of cases with Sanger and NGS, respectively, showing great concordance (97.9%), although several samples had minor discordances in the nucleotide sequence of rearrangements. Parallel NGS was performed in 82 follow-up cases, achieving a median sensitivity of 0.001%, while for NGF, median sensitivity was 0.0002%. Reproducibility of LymphoTrack-based MRD studies (85.4%) and correlation with NGF (R2 > 0.800) were high. Bland-Altman tests showed highly significant levels of agreement between flow and sequencing., Conclusions.—: Taken together, we have shown that LymphoTrack is a suitable strategy for clonality detection and MRD evaluation, with results comparable to gold standard procedures.
- Published
- 2022
- Full Text
- View/download PDF
32. Identification of the novel HLA-A*23:01:01:27 allele in an acute myeloid patient and related donor.
- Author
-
Bastos M, Alcoceba M, Chillón MC, García-Sanz R, and Boix F
- Subjects
- Alleles, HLA-A Antigens genetics, Humans, Polymorphism, Single Nucleotide, Tissue Donors
- Abstract
A single nucleotide polymorphism in intron 1 determines the new HLA-A*23:01:01:27 allele., (© 2022 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
33. High frequency of central nervous system involvement in transformed Waldenström macroglobulinemia.
- Author
-
Durot E, Kanagaratnam L, Zanwar S, Toussaint E, Kastritis E, D'Sa S, Alcoceba M, Tomowiak C, Hivert B, Protin C, Abeykoon JP, Vos JMI, Michallet AS, Rodier C, Dupuis J, Leprêtre S, Merabet F, Roussel X, Zini JM, Regny C, Patel A, Morel P, Roos-Weil D, Treon SP, Dimopoulos MA, Garcia-Sanz R, Kapoor P, Castillo JJ, and Delmer AJ
- Subjects
- Central Nervous System, Humans, Waldenstrom Macroglobulinemia
- Published
- 2022
- Full Text
- View/download PDF
34. Validation and functional characterization of GWAS-identified variants for chronic lymphocytic leukemia: a CRuCIAL study.
- Author
-
García-Martín P, Díez AM, Maldonado JMS, Serrano AJC, Ter Horst R, Benavente Y, Landi S, Macauda A, Clay-Gilmour A, Hernández-Mohedo F, Niazi Y, González-Sierra P, Espinet B, Rodríguez-Sevilla JJ, Maffei R, Blanco G, Giaccherini M, Puiggros A, Cerhan J, Marasca R, Cañadas-Garre M, López-Nevot MÁ, Chen-Liang T, Thomsen H, Gámez I, Moreno V, Marcos-Gragera R, García-Álvarez M, Llorca J, Jerez A, Berndt S, Butrym A, Norman AD, Casabonne D, Luppi M, Slager SL, Hemminki K, Li Y, Alcoceba M, Campa D, Canzian F, de Sanjosé S, Försti A, Netea MG, Jurado M, and Sainz J
- Subjects
- Genetic Predisposition to Disease, Genome-Wide Association Study, Humans, Polymorphism, Single Nucleotide, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Published
- 2022
- Full Text
- View/download PDF
35. MYD88 Mutations: Transforming the Landscape of IgM Monoclonal Gammopathies.
- Author
-
Alcoceba M, García-Álvarez M, Medina A, Maldonado R, González-Calle V, Chillón MC, Sarasquete ME, González M, García-Sanz R, and Jiménez C
- Subjects
- Aged, Humans, Immunoglobulin M genetics, Immunoglobulin M metabolism, Lymphoma genetics, Lymphoma metabolism, Lymphoma, B-Cell genetics, Lymphoma, B-Cell metabolism, Mutation, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 metabolism, Waldenstrom Macroglobulinemia diagnosis, Waldenstrom Macroglobulinemia genetics, Waldenstrom Macroglobulinemia metabolism
- Abstract
The MYD88 gene has a physiological role in the innate immune system. Somatic mutations in MYD88 , including the most common L265P, have been associated with the development of certain types of lymphoma. MYD88
L265P is present in more than 90% of patients with Waldenström's macroglobulinemia (WM) and IgM monoclonal gammopathy of undetermined significance (IgM-MGUS). The absence of MYD88 mutations in WM patients has been associated with a higher risk of transformation into aggressive lymphoma, resistance to certain therapies (BTK inhibitors), and shorter overall survival. The MyD88 signaling pathway has also been used as a target for specific therapies. In this review, we summarize the clinical applications of MYD88 testing in the diagnosis, prognosis, follow-up, and treatment of patients. Although MYD88L265P is not specific to WM, few tumors present a single causative mutation in a recurrent position. The role of the oncogene in the pathogenesis of WM is still unclear, especially considering that the mutation can be found in normal B cells of patients, as recently reported. This may have important implications for early lymphoma detection in healthy elderly individuals and for the treatment response assessment based on a MYD88L265P analysis.- Published
- 2022
- Full Text
- View/download PDF
36. Overexpression of wild type RRAS2, without oncogenic mutations, drives chronic lymphocytic leukemia.
- Author
-
Hortal AM, Oeste CL, Cifuentes C, Alcoceba M, Fernández-Pisonero I, Clavaín L, Tercero R, Mendoza P, Domínguez V, García-Flores M, Pintado B, Abia D, García-Macías C, Navarro-Bailón A, Bustelo XR, González M, and Alarcón B
- Subjects
- Animals, Genes, ras, Humans, Membrane Proteins genetics, Mice, Mutation, Receptors, Antigen, B-Cell, Signal Transduction, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Monomeric GTP-Binding Proteins genetics
- Abstract
Background: Chronic lymphocytic leukemia (CLL) is the most frequent, and still incurable, form of leukemia in the Western World. It is widely accepted that cancer results from an evolutionary process shaped by the acquisition of driver mutations which confer selective growth advantage to cells that harbor them. Clear examples are missense mutations in classic RAS genes (KRAS, HRAS and NRAS) that underlie the development of approximately 13% of human cancers. Although autonomous B cell antigen receptor (BCR) signaling is involved and mutations in many tumor suppressor genes and oncogenes have been identified, an oncogenic driver gene has not still been identified for CLL., Methods: Conditional knock-in mice were generated to overexpress wild type RRAS2 and prove its driver role. RT-qPCR analysis of a human CLL sample cohort was carried out to measure RRAS2 transcriptional expression. Sanger DNA sequencing was used to identify a SNP in the 3'UTR region of RRAS2 in human CLL samples. RNAseq of murine CLL was carried out to identify activated pathways, molecular mechanisms and to pinpoint somatic mutations accompanying RRAS2 overexpression. Flow cytometry was used for phenotypic characterization and shRNA techniques to knockdown RRAS2 expression in human CLL., Results: RRAS2 mRNA is found overexpressed in its wild type form in 82% of the human CLL samples analyzed (n = 178, mean and median = 5-fold) as well as in the explored metadata. A single nucleotide polymorphism (rs8570) in the 3'UTR of the RRAS2 mRNA has been identified in CLL patients, linking higher expression of RRAS2 with more aggressive disease. Deliberate overexpression of wild type RRAS2 in mice, but not an oncogenic Q72L mutation in the coding sequence, provokes the development of CLL. Overexpression of wild type RRAS2 in mice is accompanied by a strong convergent selection of somatic mutations in genes that have been identified in human CLL. R-RAS2 protein is physically bound to the BCR and mediates BCR signals in CLL., Conclusions: The results indicate that overexpression of wild type RRAS2 is behind the development of CLL., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
37. Revised International Prognostic Index and genetic alterations are associated with early failure to R-CHOP in patients with diffuse large B-cell lymphoma.
- Author
-
Dlouhy I, Karube K, Enjuanes A, Salaverria I, Nadeu F, Ramis-Zaldivar JE, Valero JG, Rivas-Delgado A, Magnano L, Martin-García D, Pérez-Galán P, Clot G, Rovira J, Jares P, Balagué O, Giné E, Mozas P, Briones J, Sancho JM, Salar A, Mercadal S, Alcoceba M, Valera A, Campo E, and López-Guillermo A
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Biomarkers, Tumor, Biopsy, Cyclophosphamide adverse effects, Cyclophosphamide therapeutic use, DNA Copy Number Variations, DNA Mutational Analysis, Doxorubicin adverse effects, Doxorubicin therapeutic use, Female, Gene Expression Profiling, Genetic Predisposition to Disease, Humans, In Situ Hybridization, Fluorescence, Lymphoma, Large B-Cell, Diffuse diagnosis, Lymphoma, Large B-Cell, Diffuse mortality, Male, Middle Aged, Neoplasm Grading, Neoplasm Staging, Odds Ratio, Prednisone adverse effects, Prednisone therapeutic use, Prognosis, Rituximab adverse effects, Rituximab therapeutic use, Treatment Failure, Treatment Outcome, Vincristine adverse effects, Vincristine therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Genetic Variation, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics
- Abstract
Relapsed or refractory diffuse large B-cell lymphoma (DLBCL) cases have a poor outcome. Here we analysed clinico-biological features in 373 DLBCL patients homogeneously treated with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP), in order to identify variables associated with early failure to treatment (EF), defined as primary refractoriness or relapse within 12 months from diagnosis. In addition to clinical features, mutational status of 106 genes was studied by targeted next-generation sequencing in 111 cases, copy number alterations in 87, and gene expression profile (GEP) in 39. Ninety-seven cases (26%) were identified as EF and showed significantly shorter overall survival (OS). Patients with B symptoms, advanced stage, high levels of serum lactate dehydrogenase (LDH) or β2-microglobulin, low lymphocyte/monocyte ratio and higher Revised International Prognostic Index (R-IPI) scores, as well as those with BCL2 rearrangements more frequently showed EF, with R-IPI being the most important in logistic regression. Mutations in NOTCH2, gains in 5p15·33 (TERT), 12q13 (CDK2), 12q14·1 (CDK4) and 12q15 (MDM2) showed predictive importance for EF independently from R-IPI. GEP studies showed that EF cases were significantly enriched in sets related to cell cycle regulation and inflammatory response, while cases in response showed over-representation of gene sets related to extra-cellular matrix and tumour microenvironment., (© 2021 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
38. Clinico-biological features and outcome of patients with splenic marginal zone lymphoma with histological transformation.
- Author
-
Bastidas-Mora G, Beà S, Navarro A, Gine E, Costa D, Delgado J, Baumann T, Magnano L, Rivas-Delgado A, Villamor N, Colomer D, Lopez-Guerra M, Rozman M, Balagué O, Martínez D, Baptista MJ, Escoda L, Alcoceba M, Blanes M, Climent F, Campo E, Wotherspoon A, López-Guillermo A, and Matutes E
- Subjects
- Adult, Aged, Biomarkers, Tumor, Cell Transformation, Neoplastic, Cytogenetic Analysis, Disease Management, Disease Susceptibility, Female, Humans, Immunohistochemistry, Immunophenotyping, In Situ Hybridization, Fluorescence, Incidence, Lymphoma, B-Cell, Marginal Zone epidemiology, Lymphoma, B-Cell, Marginal Zone etiology, Lymphoma, B-Cell, Marginal Zone metabolism, Male, Middle Aged, Neoplasm Grading, Neoplasm Staging, Prognosis, Splenic Neoplasms epidemiology, Splenic Neoplasms etiology, Splenic Neoplasms metabolism, Lymphoma, B-Cell, Marginal Zone diagnosis, Spleen pathology, Splenic Neoplasms diagnosis
- Abstract
We describe 36 patients with splenic marginal zone lymphoma (SMZL) with transformation (SMZL-T), including 15 from a series of 84 patients with SMZL diagnosed at the Hospital Clinic of Barcelona (HCB) and 21 diagnosed with SMZL-T in other centres. In the HCB cohort, the cumulative incidence of transformation at 5 years was 15%. Predictors for transformation were cytopenias, hypoalbuminaemia, complex karyotype (CK) and both the Intergruppo Italiano Linfomi (ILL) and simplified Haemoglobin, Platelet count, lactate dehydrogenase (LDH) and extrahilar Lymphadenopathy (HPLL)/ABC scores (P < 0·05). The only independent predictor for transformation in multivariate analysis was CK [hazard ratio (HR) 4·025, P = 0·05]. Patients with SMZL-T had a significantly higher risk of death than the remainder (HR 3·89, P < 0·001). Of the 36 patients with SMZL-T, one developed Hodgkin lymphoma and 35 a diffuse large B-cell lymphoma, 71% with a non-germinal centre phenotype. The main features were B symptoms, lymphadenopathy, and high serum LDH. CK was observed in 12/22 (55%) SMZL-T and fluorescence in situ hybridisation detected abnormalities of MYC proto-oncogene, basic helix-loop-helix transcription factor (MYC), B-cell leukaemia/lymphoma 2 (BCL2) and/or BCL6 in six of 14 (43%). In all, 21 patients received immunochemotherapy, six chemotherapy, one radiotherapy and three splenectomy. The complete response (CR) rate was 61% and the median survival from transformation was 4·92 years. Predictors for a worse survival in multivariate analysis were high-risk International Prognostic Index (HR 5·294, P = 0·016) and lack of CR (HR 2·67, P < 0·001)., (© 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
39. Liquid biopsy: a non-invasive approach for Hodgkin lymphoma genotyping.
- Author
-
Alcoceba M, García-Álvarez M, Chillón MC, Jiménez C, Medina A, Antón A, Blanco O, Díaz LG, Tamayo P, González-Calle V, Vidal MJ, Cuello R, Díaz Gálvez FJ, Queizán JA, Martín A, González M, García-Sanz R, and Sarasquete ME
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Alleles, Female, Genotype, High-Throughput Nucleotide Sequencing, Hodgkin Disease blood, Humans, Male, Middle Aged, Mutation, Prognosis, Prospective Studies, Young Adult, DNA, Neoplasm blood, Genotyping Techniques, Hodgkin Disease genetics, Liquid Biopsy
- Abstract
The Hodgkin lymphoma (HL) genomic landscape is hardly known due to the scarcity of tumour cells in the tissue. Liquid biopsy employing circulating tumour DNA (ctDNA) can emerge as an alternative tool for non-invasive genotyping. By using a custom next generation sequencing (NGS) panel in combination with unique molecule identifiers, we aimed to identify somatic variants in the ctDNA of 60 HL at diagnosis. A total of 277 variants were detected in 36 of the 49 samples (73·5%) with a good quality ctDNA sample. The median number of variants detected per patient was five (range 1-23) with a median variant allele frequency of 4·2% (0·84-28%). Genotyping revealed somatic variants in the following genes: SOCS1 (28%), IGLL5 (26%), TNFAIP3 (23%), GNA13 (23%), STAT6 (21%) and B2M (19%). Moreover, several poor prognosis features (high LDH, low serum albumin, B-symptoms, IPI ≥ 3 or at an advanced stage) were related to significantly higher amounts of ctDNA. Variant detection in ctDNA by NGS is a feasible approach to depict the genetic features of HL patients at diagnosis. Our data favour the implementation of liquid biopsy genotyping for the routine evaluation of HL patients., (© 2021 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
40. Primary refractory follicular lymphoma: a poor outcome entity with high risk of transformation to aggressive B cell lymphoma.
- Author
-
Alonso-Álvarez S, Manni M, Montoto S, Sarkozy C, Morschhauser F, Wondergem MJ, Guarini A, Magnano L, Alcoceba M, Chamuleau M, Galimberti S, Gomes da Silva M, Holte H, Zucca E, Lockmer S, Aurer I, Marcheselli L, Stepanishyna Y, Caballero Barrigón MD, Salles G, and Federico M
- Subjects
- Aged, Aged, 80 and over, Cell Transformation, Neoplastic, Female, Humans, Lymphoma, Follicular mortality, Lymphoma, Follicular therapy, Male, Middle Aged, Retrospective Studies, Lymphoma, B-Cell pathology, Lymphoma, Follicular pathology
- Abstract
Background: Primary refractory (PREF) follicular lymphoma (FL) has a completely different clinical course from that of FL that responds to front-line treatments. In addition to having poor responses to salvage therapies, it seems that patients with PREF are at increased risk of histological transformation (HT). The Aristotle consortium presented the opportunity of investigating the risk of HT in a very large series of cases. Thus, we investigated the risk of HT in patients with PREF FL compared with that of responding patients or in stable disease and ultimately their outcome., Methods: Six thousand three hundred thirty-nine patients from the Aristotle database were included in the analysis. These patients had a histologically confirmed grade 1, 2 or 3a FL diagnosed between 1997 and 2013. The primary end-points were the cumulative incidence (CI) of HT at the first progression or relapse and the survival after transformation., Findings: The 5-year CI of HT among patients with PREF was 34% (95% confidence interval (CI): 27-43), whilst it was 7.1% (95% CI: 6.0-8.5) in the group of patients with partial response (PR) or stable disease (SD) (PR + SD) and 3.5% (95% CI: 3.0-4.2) in the group of patients achieving complete response (CR). The 5-year survival after relapse (SAR) was 33% (95% CI: 28-39) for the PREF group, 57% (95% CI 54-61) in patients with PR, 51% (95% CI 43-58) in the SD group after first-line therapy and 63% (95% CI: 66-72) in patients with CR after initial treatment (p-value <0.001). The 5-year SAR for those patients with PREF who developed HT was 21% (95% CI: 12-31), clearly diminished when compared with those patients with PREF who did not experience HT (38% [95% CI: 31-44]) (p-value = 0.001)., Interpretation: Patients with PREF FL have a dismal outcome and an associated very high rate of HT that further worsens their poor prognosis., Competing Interests: Conflict of interest statement The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
41. The novel HLA-DQB1*06:03:27 allele characterised by sequence-based typing in a European bone marrow donor.
- Author
-
Boix F, Chillón MC, García-Sánchez S, Alcoceba M, and Marín-Rubio LA
- Subjects
- Alleles, HLA-DQ beta-Chains genetics, Histocompatibility Testing, Humans, Bone Marrow
- Abstract
A single nucleotide substitution in exon 3 of HLA-DQB1*06:03:01 results in a new allele, HLA-DQB1*06:03:27., (© 2021 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
42. The Hydropathy Index of the HCDR3 Region of the B-Cell Receptor Identifies Two Subgroups of IGHV-Mutated Chronic Lymphocytic Leukemia Patients With Distinct Outcome.
- Author
-
Rodríguez-Caballero A, Fuentes Herrero B, Oliva Ariza G, Criado I, Alcoceba M, Prieto C, Pérez Caro M, García-Montero AC, González Díaz M, Forconi F, Sarmento-Ribeiro AB, Almeida J, and Orfao A
- Abstract
The HCDR3 sequences of the B-cell receptor (BCR) undergo constraints in length, amino acid use, and charge during maturation of B-cell precursors and after antigen encounter, leading to BCR and antibodies with high affinity to specific antigens. Chronic lymphocytic leukemia consists of an expansion of B-cells with a mixed immature and "antigen-experienced" phenotype, with either a mutated (M-CLL) or unmutated (U-CLL) tumor BCR, associated with distinct patient outcomes. Here, we investigated the hydropathy index of the BCR of 138 CLL patients and its association with the IGHV mutational status and patient outcome. Overall, two clearly distinct subgroups of M-CLL patients emerged, based on a neutral (mean hydropathy index of -0.1) vs. negatively charged BCR (mean hydropathy index of -1.1) with molecular features closer to those of B-cell precursors and peripheral/mature B-cells, respectively. Despite that M-CLL with neutral HCDR3 did not show traits associated with a mature B-cell repertoire, important differences in IGHV gene usage of tumor cells and patient outcome were observed in this subgroup of patients once compared to both U-CLL and M-CLL with negatively charged HCDR3 sequences. Compared to M-CLL with negatively charged HCDR3 sequences, M-CLL with neutral HCDR3 sequences showed predominance of men, more advanced stages of the disease, and a greater frequency of genetic alterations-e.g., del(17p)-together with a higher rate of disease progression and shorter time to therapy (TTT), independently of other prognostic factors. Our data suggest that the hydropathy index of the HCDR3 sequences of CLL cells allows the identification of a subgroup of M-CLL with intermediate prognostic features between U-CLL and the more favorable subgroup of M-CLL with a negatively charged BCR., 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 © 2021 Rodríguez-Caballero, Fuentes Herrero, Oliva Ariza, Criado, Alcoceba, Prieto, Pérez Caro, García-Montero, González Díaz, Forconi, Sarmento-Ribeiro, Almeida and Orfao.)
- Published
- 2021
- Full Text
- View/download PDF
43. Genomic mutation profile in progressive chronic lymphocytic leukemia patients prior to first-line chemoimmunotherapy with FCR and rituximab maintenance (REM).
- Author
-
González-Rincón J, Garcia-Vela JA, Gómez S, Fernández-Cuevas B, Nova-Gurumeta S, Pérez-Sanz N, Alcoceba M, González M, Anguita E, López-Jiménez J, González-Barca E, Yáñez L, Pérez-Persona E, de la Serna J, Fernández-Zarzoso M, Deben G, Peñalver FJ, Fernández MC, de Oteyza JP, Andreu MÁ, Ruíz-Guinaldo MÁ, Paz-Arias R, García-Malo MD, Recasens V, Collado R, Córdoba R, Navarro-Matilla B, Sánchez-Beato M, and García-Marco JA
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Combined Modality Therapy, DNA Mutational Analysis, Female, Genomics, Humans, Male, Middle Aged, RNA Splicing, Vidarabine administration & dosage, Cyclophosphamide administration & dosage, Gene Expression Regulation, Leukemic, Immunotherapy methods, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Mutation, Rituximab administration & dosage, Vidarabine analogs & derivatives
- Abstract
Chronic Lymphocytic Leukemia (CLL) is the most prevalent leukemia in Western countries and is notable for its variable clinical course. This variability is partly reflected by the mutational status of IGHV genes. Many CLL samples have been studied in recent years by next-generation sequencing. These studies have identified recurrent somatic mutations in NOTCH1, SF3B1, ATM, TP53, BIRC3 and others genes that play roles in cell cycle, DNA repair, RNA metabolism and splicing. In this study, we have taken a deep-targeted massive sequencing approach to analyze the impact of mutations in the most frequently mutated genes in patients with CLL enrolled in the REM (rituximab en mantenimiento) clinical trial. The mutational status of our patients with CLL, except for the TP53 gene, does not seem to affect the good results obtained with maintenance therapy with rituximab after front-line FCR treatment., Competing Interests: J.A. Garcia-Marco has received honoraria for advisory board and speaker’s bureau from Mundipharma, Glaxo, AbbVie, Roche, Gilead, and Janssen, and research support from Hoffman-La Roche and Janssen. Jaime Pérez de Oteyza declares a consulting and advisory role for Hoffman-La Roche. The other authors have no conflicts of interest to declare. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2021
- Full Text
- View/download PDF
44. Anti-TRBC1 Antibody-Based Flow Cytometric Detection of T-Cell Clonality: Standardization of Sample Preparation and Diagnostic Implementation.
- Author
-
Muñoz-García N, Lima M, Villamor N, Morán-Plata FJ, Barrena S, Mateos S, Caldas C, Balanzategui A, Alcoceba M, Domínguez A, Gómez F, Langerak AW, van Dongen JJM, Orfao A, and Almeida J
- Abstract
A single antibody (anti-TRBC1; JOVI-1 antibody clone) against one of the two mutually exclusive T-cell receptor β-chain constant domains was identified as a potentially useful flow-cytometry (FCM) marker to assess Tαβ-cell clonality. We optimized the TRBC1-FCM approach for detecting clonal Tαβ-cells and validated the method in 211 normal, reactive and pathological samples. TRBC1 labeling significantly improved in the presence of CD3. Purified TRBC1
+ and TRBC1- monoclonal and polyclonal Tαβ-cells rearranged TRBJ1 in 44/47 (94%) and TRBJ1+TRBJ2 in 48 of 48 (100%) populations, respectively, which confirmed the high specificity of this assay. Additionally, TRBC1+ /TRBC1- ratios within different Tαβ-cell subsets are provided as reference for polyclonal cells, among which a bimodal pattern of TRBC1-expression profile was found for all TCRVβ families, whereas highly-variable TRBC1+ /TRBC1- ratios were observed in more mature vs. naïve Tαβ-cell subsets (vs. total T-cells). In 112/117 (96%) samples containing clonal Tαβ-cells in which the approach was validated, monotypic expression of TRBC1 was confirmed. Dilutional experiments showed a level of detection for detecting clonal Tαβ-cells of ≤10-4 in seven out of eight pathological samples. These results support implementation of the optimized TRBC1-FCM approach as a fast, specific and accurate method for assessing T-cell clonality in diagnostic-FCM panels, and for minimal (residual) disease detection in mature Tαβ+ leukemia/lymphoma patients.- Published
- 2021
- Full Text
- View/download PDF
45. Validation of the EuroClonality-NGS DNA capture panel as an integrated genomic tool for lymphoproliferative disorders.
- Author
-
Stewart JP, Gazdova J, Darzentas N, Wren D, Proszek P, Fazio G, Songia S, Alcoceba M, Sarasquete ME, Villarese P, van der Klift MY, Heezen KC, McCafferty N, Pal K, Catherwood M, Kim CS, Srivastava S, Kroeze LI, Hodges E, Stamatopoulos K, Klapper W, Genuardi E, Ferrero S, van den Brand M, Cazzaniga G, Davi F, Sutton LA, Garcia-Sanz R, Groenen PJTA, Macintyre EA, Brüggemann M, Pott C, Langerak AW, and Gonzalez D
- Subjects
- DNA, Genomics, Humans, Immunoglobulins, Gene Rearrangement, Lymphoproliferative Disorders diagnosis, Lymphoproliferative Disorders genetics
- Abstract
Current diagnostic standards for lymphoproliferative disorders include multiple tests for detection of clonal immunoglobulin (IG) and/or T-cell receptor (TCR) rearrangements, translocations, copy-number alterations (CNAs), and somatic mutations. The EuroClonality-NGS DNA Capture (EuroClonality-NDC) assay was designed as an integrated tool to characterize these alterations by capturing IGH switch regions along with variable, diversity, and joining genes of all IG and TCR loci in addition to clinically relevant genes for CNA and mutation analysis. Diagnostic performance against standard-of-care clinical testing was assessed in a cohort of 280 B- and T-cell malignancies from 10 European laboratories, including 88 formalin-fixed paraffin-embedded samples and 21 reactive lesions. DNA samples were subjected to the EuroClonality-NDC protocol in 7 EuroClonality-NGS laboratories and analyzed using a bespoke bioinformatic pipeline. The EuroClonality-NDC assay detected B-cell clonality in 191 (97%) of 197 B-cell malignancies and T-cell clonality in 71 (97%) of 73 T-cell malignancies. Limit of detection (LOD) for IG/TCR rearrangements was established at 5% using cell line blends. Chromosomal translocations were detected in 145 (95%) of 152 cases known to be positive. CNAs were validated for immunogenetic and oncogenetic regions, highlighting their novel role in confirming clonality in somatically hypermutated cases. Single-nucleotide variant LOD was determined as 4% allele frequency, and an orthogonal validation using 32 samples resulted in 98% concordance. The EuroClonality-NDC assay is a robust tool providing a single end-to-end workflow for simultaneous detection of B- and T-cell clonality, translocations, CNAs, and sequence variants., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
46. Allele and haplotype frequencies of HLA-A, -B, -C, -DRB1, -DQB1 and -DQA1 in Castile and Leon region from North West of Spain.
- Author
-
Boix F, Marín-Rubio LA, Alcoceba M, García-Álvarez M, Chillón MC, García-Sáncez S, Burillo S, Terradillos-Sánchez P, Jiménez-Hernaz I, Lucas-Sánchez S, Balanzategui A, Abad-Molina C, Corral R, González M, and García-Sanz R
- Subjects
- Bone Marrow Cells metabolism, Genomics methods, Histocompatibility Testing, Humans, Polymorphism, Genetic, Registries, Spain, Alleles, Gene Frequency, Genetics, Population, HLA Antigens genetics, Haplotypes
- Abstract
Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2021
- Full Text
- View/download PDF
47. 6q deletion in Waldenström macroglobulinaemia negatively affects time to transformation and survival.
- Author
-
García-Sanz R, Dogliotti I, Zaccaria GM, Ocio EM, Rubio A, Murillo I, Escalante F, Aguilera C, García-Mateo A, García de Coca A, Hernández R, Dávila J, Puig N, García-Álvarez M, Chillón MDC, Alcoceba M, Medina A, González de la Calle V, Sarasquete ME, González M, Gutiérrez NC, and Jiménez C
- Subjects
- Aged, Asymptomatic Diseases, Bone Marrow Cells chemistry, Bone Marrow Cells ultrastructure, Chromosome Deletion, Chromosomes, Human, Pair 6 genetics, Female, Humans, Immunoglobulin M blood, Immunophenotyping, In Situ Hybridization, Fluorescence, Male, Middle Aged, Monoclonal Gammopathy of Undetermined Significance genetics, Paraproteins analysis, Prognosis, Progression-Free Survival, Risk Assessment, Survival Analysis, Time Factors, Treatment Outcome, Waldenstrom Macroglobulinemia pathology, Cell Transformation, Neoplastic genetics, Waldenstrom Macroglobulinemia genetics
- Abstract
Deletion of the long arm of chromosome 6 (del6q) is the most frequent cytogenetic abnormality in Waldenström macroglobulinaemia (WM), occurring in approximately 50% of patients. Its effect on patient outcome has not been completely established. We used fluorescence in situ hybridisation to analyse the prevalence of del6q in selected CD19+ bone marrow cells of 225 patients with newly diagnosed immunoglobulin M (IgM) monoclonal gammopathies. Del6q was identified in one of 27 (4%) cases of IgM-monoclonal gammopathy of undetermined significance, nine of 105 (9%) of asymptomatic WM (aWM), and 28/93 (30%) of symptomatic WM (sWM), and was associated with adverse prognostic features and higher International Prognostic Scoring System for WM (IPSSWM) score. Asymptomatic patients with del6q ultimately required therapy more often and had a shorter time to transformation (TT) to symptomatic disease (median TT, 30 months vs. 199 months, respectively, P < 0·001). When treatment was required, 6q-deleted patients had shorter progression-free survival (median 20 vs. 47 months, P < 0·001). The presence of del6q translated into shorter overall survival (OS), irrespective of the initial diagnosis, with a median OS of 90 compared with 131 months in non-del6q patients (P = 0·01). In summary, our study shows that del6q in IgM gammopathy is associated with symptomatic disease, need for treatment and poorer clinical outcomes., (© 2020 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
48. Building a network of TP53 and IGHV testing reference centers across Spain: the Red53 initiative.
- Author
-
Bosch F, Navarro B, Crespo M, Alcoceba M, Sánchez JB, Tazón B, Serrano A, Álvarez MG, Serrano LG, Alonso-Torres P, Villanueva M, Loriente C, Abrisqueta P, Peiró M, García-Marco JA, González M, and Terol MJ
- Subjects
- Biomarkers, Tumor genetics, Clinical Laboratory Services supply & distribution, Cohort Studies, Community Networks organization & administration, Humans, Implementation Science, Intersectoral Collaboration, Job Satisfaction, Leukemia, Lymphocytic, Chronic, B-Cell diagnosis, Leukemia, Lymphocytic, Chronic, B-Cell epidemiology, Molecular Diagnostic Techniques methods, Mutation, Prognosis, Spain epidemiology, Surveys and Questionnaires, Clinical Laboratory Services organization & administration, DNA Mutational Analysis methods, Immunoglobulin Heavy Chains genetics, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Referral and Consultation organization & administration, Tumor Suppressor Protein p53 genetics
- Abstract
Among the different biomarkers predicting response in chronic lymphocytic leukemia (CLL), the most influential parameters are the mutational status of the IGHV genes and the presence of TP53 gene disruptions. Nevertheless, these important assessments are not readily available in most centers dealing with CLL patients. To provide this molecular testing across the country, the Spanish Cooperative Group on CLL (GELLC) established a network of four analytical reference centers. A total of 2153 samples from 256 centers were analyzed over a period of 30 months. In 9% of the patients, we found pathological mutations in the TP53 gene, whereas 48.96% were classified as IGHV unmutated. Results of the satisfaction survey of the program showed a Net Promoter Score of 85.15. Building a national network for molecular testing in CLL allowed the CLL population a broad access to complex biomarkers analysis that should translate into a more accurate and informed therapeutic decision-making.
- Published
- 2021
- Full Text
- View/download PDF
49. Genetic complexity impacts the clinical outcome of follicular lymphoma patients.
- Author
-
García-Álvarez M, Alonso-Álvarez S, Prieto-Conde I, Jiménez C, Sarasquete ME, Chillón MC, Medina A, Balanzategui A, Maldonado R, Antón A, Rodríguez M, Blanco O, Díaz LG, Tamayo P, Blanco P, Esteban C, González-Calle V, Puig N, Gutiérrez N, Martín A, García-Sanz R, González M, Caballero MD, and Alcoceba M
- Subjects
- Humans, Translocation, Genetic, Lymphoma, Follicular diagnosis, Lymphoma, Follicular genetics
- Published
- 2021
- Full Text
- View/download PDF
50. STAT3 and STAT5B Mutations in T/NK-Cell Chronic Lymphoproliferative Disorders of Large Granular Lymphocytes (LGL): Association with Disease Features.
- Author
-
Muñoz-García N, Jara-Acevedo M, Caldas C, Bárcena P, López A, Puig N, Alcoceba M, Fernández P, Villamor N, Flores-Montero JA, Gómez K, Lemes MA, Hernández JC, Álvarez-Twose I, Guerra JL, González M, Orfao A, and Almeida J
- Abstract
STAT3 and STAT5B ( STAT3/STAT5B ) mutations are the most common mutations in T-cell large granular lymphocytic leukemia (T-LGLL) and chronic lymphoproliferative disorders of NK cells (CLPD-NK), but their clinical impact remains unknown. We investigated the frequency and type of STAT3/STAT5B mutations in FACS-sorted populations of expanded T/NK-LGL from 100 (82 clonal; 6 oligoclonal; 12 polyclonal) patients, and its relationship with disease features. Seventeen non-LGL T-CLPD patients and 628 age-matched healthy donors were analyzed as controls. STAT3 ( n = 30) and STAT5B ( n = 1) mutations were detected in 28/82 clonal T/NK-LGLL patients (34%), while absent (0/18, 0%) among oligoclonal/polyclonal LGL-lymphocytosis. Mutations were found across all diagnostic subgroups: TCD8
+ -LGLL, 36%; CLPD-NK, 38%; TCD4+ -LGLL, 7%; Tαβ+ DP-LGLL, 100%; Tαβ+ DN-LGLL, 50%; Tγδ+ -LGLL, 44%. STAT3 -mutated T-LGLL/CLPD-NK showed overall reduced ( p < 0.05) blood counts of most normal leukocyte subsets, with a higher rate (vs. nonmutated LGLL) of neutropenia ( p = 0.04), severe neutropenia ( p = 0.02), and cases requiring treatment ( p = 0.0001), together with a shorter time-to-therapy ( p = 0.0001), particularly in non-Y640F STAT3- mutated patients. These findings confirm and extend on previous observations about the high prevalence of STAT3 mutations across different subtypes of LGLL, and its association with a more marked decrease of all major blood-cell subsets and a shortened time-to-therapy.- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.