119 results on '"Cerchietti, L"'
Search Results
2. The transcriptional modulator BCL6 as a molecular target for breast cancer therapy
- Author
-
Walker, S R, Liu, S, Xiang, M, Nicolais, M, Hatzi, K, Giannopoulou, E, Elemento, O, Cerchietti, L, Melnick, A, and Frank, D A
- Published
- 2015
- Full Text
- View/download PDF
3. 17P BCL6 and Notch pathway: A signaling axis leading to a novel druggable biotarget in triple-negative breast cancer
- Author
-
De Santis, F., primary, Romero-Cordoba, S.L., additional, Castagnoli, L., additional, Volpari, T., additional, Faraci, S., additional, Fucà, G., additional, Cerchietti, L., additional, Tagliabue, E., additional, De Braud, F., additional, Pupa, S.M., additional, and Di Nicola, M., additional
- Published
- 2021
- Full Text
- View/download PDF
4. Performance of TXRF among other competitors' analytical techniques
- Author
-
Vázquez, C., primary, Gonzáles Sintas, M.F., additional, and Cerchietti, L., additional
- Published
- 2019
- Full Text
- View/download PDF
5. MULTI-CENTER PHASE II STUDY OF ORAL AZACITIDINE (CC-486) PLUS CHOP AS INITIAL TREATMENT FOR PERIPHERAL T-CELL LYMPHOMA
- Author
-
Ruan, J., primary, Leonard, J.P., additional, Coleman, M., additional, Rutherford, S., additional, Van Besien, K., additional, Rodriguez, A., additional, Benderoff, L., additional, Mehta-Shah, N., additional, Moskowitz, A., additional, Sokol, L., additional, Cerchietti, L., additional, Inghirami, G., additional, and Martin, P., additional
- Published
- 2019
- Full Text
- View/download PDF
6. Abstract P1-07-02: Withdrawn
- Author
-
Di Nicola, M, primary, Volpari, T, additional, Fucà, G, additional, Romero Cordoba, S, additional, De Santis, F, additional, Rondinone, O, additional, Faraci, S, additional, Ferris, F, additional, Puricelli, C, additional, Castagnoli, L, additional, Marullo, R, additional, Cerchietti, L, additional, and Pupa, S, additional
- Published
- 2019
- Full Text
- View/download PDF
7. Performance of TXRF among other competitors' analytical techniques.
- Author
-
Vázquez, C., Gonzáles Sintas, M.F., and Cerchietti, L.
- Subjects
INDUCTIVELY coupled plasma atomic emission spectrometry ,FLUORESCENCE spectroscopy ,X-ray fluorescence ,X-ray reflection ,ARSENIC ,INDUCTIVELY coupled plasma mass spectrometry ,NITRIC acid ,SPECTROMETRY - Abstract
The main purpose of this presentation was to evaluate the performance of total reflection X‐ray fluorescence (TXRF) among conventional analytical techniques such as atomic absorption spectrometry (AAS), inductively coupled plasma‐optical emission spectrometry (ICP‐OES), and inductively coupled plasma‐mass spectrometry ICP‐MS). In order to perform the study, one sample and a blank acidified with nitric acid (2.0%–2.5% v/v) were distributed among 10 laboratories. The investigated analytes were arsenic, chromium, nickel, and lead within 0.050 to 1.50‐mg/L concentration range. A guide protocol was elaborated for the participants in order to normalize the manipulation of the samples. In the particular case of arsenic, atomic fluorescence spectrometry (AFS) was incorporated. The "En" statistical test was performed in order to evaluate the results reported by each participant against the assigned value of the sample. Results show an excellent performance of TXRF among the mentioned analytical techniques. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
8. PHASE II/III STUDY OF R‐MINICHOP ± ORAL AZACITIDINE IN PARTICIPANTS AGE 75 YEARS OR OLDER WITH DIFFUSE LARGE B CELL AND RELATED LYMPHOMAS.
- Author
-
Brem, E. A., Li, H., Beaven, A. W., Caimi, P., Cerchietti, L., Olin, R. L., Henry, N. L., Dilon, H., Little, R. F., Leblanc, M. L., Kahl, B., Leonard, J. P., Friedberg, J. W., and Smith, S. M.
- Subjects
DIFFUSE large B-cell lymphomas ,AZACITIDINE - Abstract
The phase II objective is to determine if oral azacitidine + R-miniCHOP should be tested further against R-miniCHOP based on estimates of 1-year PFS. S1918 incorporates the FIL Tool for baseline frailty assessment and a serial comprehensive geriatric assessment to evaluate effects of therapy on quality of life and functional status. Patients receive prephase therapy with prednisone 60-100 mg × 4-7 days to improve performance status and decrease early treatment-related mortality (Owens CN et al., 2015; Pfreundschuh M et al., 2008). [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
9. The transcriptional modulator BCL6 as a molecular target for breast cancer therapy
- Author
-
Walker, S R, primary, Liu, S, additional, Xiang, M, additional, Nicolais, M, additional, Hatzi, K, additional, Giannopoulou, E, additional, Elemento, O, additional, Cerchietti, L, additional, Melnick, A, additional, and Frank, D A, additional
- Published
- 2014
- Full Text
- View/download PDF
10. MiR-592 Regulates the Induction and Cell Death-Promoting Activity of p75NTR in Neuronal Ischemic Injury
- Author
-
Irmady, K., primary, Jackman, K. A., additional, Padow, V. A., additional, Shahani, N., additional, Martin, L. A., additional, Cerchietti, L., additional, Unsicker, K., additional, Iadecola, C., additional, and Hempstead, B. L., additional
- Published
- 2014
- Full Text
- View/download PDF
11. Oral morphine versus oral midazolam as upfront therapy to relief dyspnea in ambulatory cancer patients while its underlying cause is sought or treated
- Author
-
Cerchietti, L. C., primary, Navigante, A. H., additional, and Castro, M. A., additional
- Published
- 2008
- Full Text
- View/download PDF
12. Le Midazolam comme thérapeutique adjuvante de la Morphine dans le contrôle de la perception de la dyspnée sévère chez les patients présentant un cancer avancé
- Author
-
Navigante, A.-H., primary, Cerchietti, L.-C., additional, Castro, M.-A., additional, Luttera, M.-A., additional, and Cabalar, M.-E., additional
- Published
- 2006
- Full Text
- View/download PDF
13. Late toxicity in head and neck cancer (H&N) patients (pts) treated with hyperfractionated (HF) or accelerated (AC) radiotherapy (RT) and chemotherapy (CT)
- Author
-
Roth, B. M., primary, Giglio, R., additional, Cerchietti, L., additional, Blanco Saborio, A., additional, Cuartero, V., additional, Menendez, P., additional, Pogany, C., additional, Mickiewicz, E., additional, and Bonomi, M. R., additional
- Published
- 2004
- Full Text
- View/download PDF
14. Clinical trial. Hypodermoclysis for control of dehydration in terminal-stage cancer.
- Author
-
Cerchietti L, Navigante A, Sauri A, and Palazzo F
- Published
- 2000
15. A novel patient-derived tumorgraft model with TRAF1-ALK anaplastic large-cell lymphoma translocation
- Author
-
Thomas Tousseyn, Elena Lasorsa, M. Ponzoni, Cristina Abele, Andrea Acquaviva, S. A. Pileri, Pier Paolo Piccaluga, Domenico Novero, Maria Todaro, Antonella Barreca, Francesco Abate, Ivo Kwee, Giorgio Inghirami, F Di Giacomo, Javeed Iqbal, Indira Landra, Raul Rabadan, Silvio Aime, Wing C. Chan, Rodolfo Machiorlatti, Mangeng Cheng, Michela Boi, Enrico Tiacci, B Pera-Gresely, Francesco Bertoni, Leonard D. Shultz, J-A van der Krogt, Katia Messana, Bruce Ruggeri, Brunangelo Falini, Sabrina Aliberti, Fabrizio Tabbò, Marcello Gaudiano, Luca Bessone, Roberto Piva, R Crescenzo, Andrea Rinaldi, Iwona Wlodarska, Dario Livio Longo, Elisa Ficarra, Leandro Cerchietti, Abate, F., Todaro, M., Van Der Krogt, J.-A., Boi, M., Landra, I., Machiorlatti, R., Tabbò, F., Messana, K., Abele, C., Barreca, A., Novero, D., Gaudiano, M., Aliberti, S., Di Giacomo, F., Tousseyn, T., Lasorsa, E., Crescenzo, R., Bessone, L., Ficarra, E., Acquaviva, A., Rinaldi, A., Ponzoni, M., Longo, D.L., Aime, S., Cheng, M., Ruggeri, B., Piccaluga, P.P., Pileri, S., Tiacci, E., Falini, B., Pera-Gresely, B., Cerchietti, L., Iqbal, J., Chan, W.C., Shultz, L.D., Kwee, I., Piva, R., Wlodarska, I., Rabadan, R., Bertoni, F., Inghirami, G., The European T-cell Lymphoma Study Group [.., Agostinelli, C., ], European T-cell Lymphoma Study Group, Cavallo, F., Chiesa, N., Fienga, A., di Giacomo, F., Marchiorlatti, R., Martinoglio, B., Medico, E., Ferrero, GB., Mereu, E., Pellegrino, E., Scafò, I., Spaccarotella, E., Ubezzi, I., Urigu, S., Chiapella, A., Vitolo, U., Agnelli, L., Neri, A., Chilosi£££Anna Caliò Marco£££ AC., Zamó, A., Facchetti, F., Lonardi, S., De Chiara, A., Fulciniti, F., Ferreri, A., Piccaluga, PP., Van Loo, P., De Wolf-Peeters, C., Geissinger, E., Muller-Hermelink, HK., Rosenwald, A., Piris, MA., Rodriguez, ME., Chiattone, C., Paes, RA., Abate, F, Todaro, M, van der Krogt, Ja, Boi, M, Landra, I, Machiorlatti, R, Tabbò, F, Messana, K, Abele, C, Barreca, A, Novero, D, Gaudiano, M, Aliberti, S, Di Giacomo, F, Tousseyn, T, Lasorsa, E, Crescenzo, R, Bessone, L, Ficarra, E, Acquaviva, A, Rinaldi, A, Ponzoni, M, Longo, Dl, Aime, S, Cheng, M, Ruggeri, B, Piccaluga, Pp, Pileri, S, Tiacci, E, Falini, B, Pera-Gresely, B, Cerchietti, L, Iqbal, J, Chan, Wc, Shultz, Ld, Kwee, I, Piva, R, Wlodarska, I, Rabadan, R, Bertoni, F, Inghirami, G, and andThe European T-cell Lymphoma Study, Group
- Subjects
Pathology ,Cancer Research ,Lymphoma ,TRAF1 ,Messenger ,Drug Resistance ,Translocation, Genetic ,Fusion gene ,Mice ,Mice, Inbred NOD ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,Anaplastic lymphoma kinase ,Anaplastic ,Anaplastic Lymphoma Kinase ,Anaplastic large-cell lymphoma ,Animals ,Blotting, Western ,Flow Cytometry ,Gene Expression Profiling ,High-Throughput Nucleotide Sequencing ,Humans ,Immunoprecipitation ,In Situ Hybridization, Fluorescence ,Lymphoma, Large-Cell, Anaplastic ,NF-kappa B ,Proteasome Inhibitors ,Proto-Oncogene Proteins c-myc ,RNA, Messenger ,Real-Time Polymerase Chain Reaction ,Receptor Protein-Tyrosine Kinases ,Repressor Proteins ,Reverse Transcriptase Polymerase Chain Reaction ,Signal Transduction ,TNF Receptor-Associated Factor 1 ,Tumor Suppressor Protein p53 ,Xenograft Model Antitumor Assays ,Drug Resistance, Neoplasm ,In Situ Hybridization ,Hematology ,Cultured ,Blotting ,Medicine (all) ,Large-Cell ,Tumor Cells ,Proteasome Inhibitor ,Receptor Protein-Tyrosine Kinase ,Oncology ,Western ,Human ,medicine.medical_specialty ,fusion detection tool ,Xenograft Model Antitumor Assay ,medicine.drug_class ,Translocation ,Anesthesiology and Pain Medicine ,Biology ,anaplastic large-cell lymphomas (ALCL) ,RNA-Seq data ,Fluorescence ,Article ,Genetic ,Internal medicine ,PRDM1 ,medicine ,traslocation ,Animal ,Repressor Protein ,medicine.disease ,ALK inhibitor ,anaplastic lymphoma kinase (ALK) ,Cancer research ,Inbred NOD ,RNA ,Neoplasm ,Positive Regulatory Domain I-Binding Factor 1 ,Lymphoma, Large-Cell, Anaplastic/drug therapy ,Lymphoma, Large-Cell, Anaplastic/genetics ,NF-kappa B/genetics ,NF-kappa B/metabolism ,Proteasome Inhibitors/pharmacology ,Proto-Oncogene Proteins c-myc/genetics ,Proto-Oncogene Proteins c-myc/metabolism ,RNA, Messenger/genetics ,Receptor Protein-Tyrosine Kinases/genetics ,Receptor Protein-Tyrosine Kinases/metabolism ,Repressor Proteins/genetics ,Repressor Proteins/metabolism ,TNF Receptor-Associated Factor 1/genetics ,TNF Receptor-Associated Factor 1/metabolism ,Translocation, Genetic/genetics ,Tumor Suppressor Protein p53/genetics ,Tumor Suppressor Protein p53/metabolism - Abstract
Although anaplastic large-cell lymphomas (ALCL) carrying anaplastic lymphoma kinase (ALK) have a relatively good prognosis, aggressive forms exist. We have identified a novel translocation, causing the fusion of the TRAF1 and ALK genes, in one patient who presented with a leukemic ALK+ ALCL (ALCL-11). To uncover the mechanisms leading to high-grade ALCL, we developed a human patient-derived tumorgraft (hPDT) line. Molecular characterization of primary and PDT cells demonstrated the activation of ALK and nuclear factor kappa B (NF kappa B) pathways. Genomic studies of ALCL-11 showed the TP53 loss and the in vivo subclonal expansion of lymphoma cells, lacking PRDM1/Blimp1 and carrying c-MYC gene amplification. The treatment with proteasome inhibitors of TRAF1-ALK cells led to the downregulation of p50/p52 and lymphoma growth inhibition. Moreover, a NF kappa B gene set classifier stratified ALCL in distinct subsets with different clinical outcome. Although a selective ALK inhibitor (CEP28122) resulted in a significant clinical response of hPDT mice, nevertheless the disease could not be eradicated. These data indicate that the activation of NF kappa B signaling contributes to the neoplastic phenotype of TRAF1-ALK ALCL. ALCL hPDTs are invaluable tools to validate the role of druggable molecules, predict therapeutic responses and implement patient specific therapies.
- Published
- 2015
16. Endothelial-leukemia interactions remodel drug responses uncovering T-ALL vulnerabilities
- Author
-
Luca Vincenzo Cappelli, Danilo Fiore, Jude M. Phillip, Liron Yoffe, Filomena Di Giacomo, William Chiu, Yang Hu, Clarisse Kayembe, Michael Ginsberg, Lorena Consolino, Jose Gabriel Barcia Duran, Nahuel Zamponi, Ari M. Melnick, Francesco Boccalatte, Wayne Tam, Olivier Elemento, Sabina Chiaretti, Anna Guarini, Robin Foà, Leandro Cerchietti, Shahin Rafii, Giorgio Inghirami, Cappelli, L. V., Fiore, D., Phillip, J. M., Yoffe, L., Di Giacomo, F., Chiu, W., Hu, Y., Kayembe, C., Ginsberg, M., Consolino, L., Barcia Duran, J. G., Zamponi, N., Melnick, A. M., Boccalatte, F., Tam, W., Elemento, O., Chiaretti, S., Guarini, A., Foa, R., Cerchietti, L., Rafii, S., and Inghirami, G.
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive and often incurable disease. To uncover therapeutic vulnerabilities, we first developed T-ALL patient–derived tumor xenografts (PDXs) and exposed PDX cells to a library of 433 clinical-stage compounds in vitro. We identified 39 broadly active drugs with antileukemia activity. Because endothelial cells (ECs) can alter drug responses in T-ALL, we developed an EC/T-ALL coculture system. We found that ECs provide protumorigenic signals and mitigate drug responses in T-ALL PDXs. Whereas ECs broadly rescued several compounds in most models, for some drugs the rescue was restricted to individual PDXs, suggesting unique crosstalk interactions and/or intrinsic tumor features. Mechanistically, cocultured T-ALL cells and ECs underwent bidirectional transcriptomic changes at the single-cell level, highlighting distinct “education signatures.” These changes were linked to bidirectional regulation of multiple pathways in T-ALL cells as well as in ECs. Remarkably, in vitro EC-educated T-ALL cells transcriptionally mirrored ex vivo splenic T-ALL at single-cell resolution. Last, 5 effective drugs from the 2 drug screenings were tested in vivo and shown to effectively delay tumor growth and dissemination thus prolonging overall survival. In sum, we developed a T-ALL/EC platform that elucidated leukemia-microenvironment interactions and identified effective compounds and therapeutic vulnerabilities.
- Published
- 2022
17. A Novel JAK1 Mutant Breast Implant-Associated Anaplastic Large Cell Lymphoma Patient-Derived Xenograft Fostering Pre-Clinical Discoveries
- Author
-
Robin Foà, Ahmet Dogan, Luca Vincenzo Cappelli, Steven M. Horwitz, Joseph Casano, Jude M Phillips, Paul Zumbo, Danilo Fiore, Liron Yoffe, David M. Weinstock, Leandro Cerchietti, Clarisse Kayembe, Federica Di Maggio, Elisa de Stanchina, Paola Ghione, Raul Rabadan, Zhaoqi Liu, Giorgio Inghirami, Wayne Tam, Inna Khodos, Shuhua Cheng, Doron Betel, Fiore, D., Cappelli, L. V., Zumbo, P., Phillips, J. M., Liu, Z., Cheng, S., Yoffe, L., Ghione, P., Di Maggio, F., Dogan, A., Khodos, I., de Stanchina, E., Casano, J., Kayembe, C., Tam, W., Betel, D., Foa', R., Cerchietti, L., Rabadan, R., Horwitz, S., Weinstock, D. M., and Inghirami, G.
- Subjects
0301 basic medicine ,Cancer Research ,Ruxolitinib ,precision medicine ,PDGFRA ,Biology ,lcsh:RC254-282 ,Article ,Deep sequencing ,drug discovery ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,JAK/STAT pathway ,pre-clinical model ,Anaplastic large-cell lymphoma ,Exome sequencing ,PDGFB ,Drug discovery ,patient-derived tumor xenograft ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Lymphoma ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine.drug - Abstract
Breast implant-associated lymphoma (BIA-ALCL) has recently been recognized as an independent peripheral T-cell lymphoma (PTCL) entity. In this study, we generated the first BIA-ALCL patient-derived tumor xenograft (PDTX) model (IL89) and a matching continuous cell line (IL89_CL#3488) to discover potential vulnerabilities and druggable targets. We characterized IL89 and IL89_CL#3488, both phenotypically and genotypically, and demonstrated that they closely resemble the matching human primary lymphoma. The tumor content underwent significant enrichment along passages, as confirmed by the increased variant allele frequency (VAF) of mutations. Known aberrations (JAK1 and KMT2C) were identified, together with novel hits, including PDGFB, PDGFRA, and SETBP1. A deep sequencing approach allowed the detection of mutations below the Whole Exome Sequencing (WES) sensitivity threshold, including JAK1G1097D, in the primary sample. RNA sequencing confirmed the expression of a signature of differentially expressed genes in BIA-ALCL. Next, we tested IL89&rsquo, s sensitivity to the JAK inhibitor ruxolitinib and observed a potent anti-tumor effect, both in vitro and in vivo. We also implemented a high-throughput drug screening approach to identify compounds associated with increased responses in the presence of ruxolitinib. In conclusion, these new IL89 BIA-ALCL models closely recapitulate the primary correspondent lymphoma and represent an informative platform for dissecting the molecular features of BIA-ALCL and performing pre-clinical drug discovery studies, fostering the development of new precision medicine approaches.
- Published
- 2020
- Full Text
- View/download PDF
18. Profiling Dynamic Patterns of Single-Cell Motility.
- Author
-
Maity D, Sivakumar N, Kamat P, Zamponi N, Min C, Du W, Jayatilaka H, Johnston A, Starich B, Agrawal A, Riley D, Venturutti L, Melnick A, Cerchietti L, Walston J, and Phillip JM
- Subjects
- Humans, Cell Movement physiology, Single-Cell Analysis methods
- Abstract
Cell motility plays an essential role in many biological processes as cells move and interact within their local microenvironments. Current methods for quantifying cell motility typically involve tracking individual cells over time, but the results are often presented as averaged values across cell populations. While informative, these ensemble approaches have limitations in assessing cellular heterogeneity and identifying generalizable patterns of single-cell behaviors, at baseline and in response to perturbations. In this study, CaMI is introduced, a computational framework designed to leverage the single-cell nature of motility data. CaMI identifies and classifies distinct spatio-temporal behaviors of individual cells, enabling robust classification of single-cell motility patterns in a large dataset (n = 74 253 cells). This framework allows quantification of spatial and temporal heterogeneities, determination of single-cell motility behaviors across various biological conditions and provides a visualization scheme for direct interpretation of dynamic cell behaviors. Importantly, CaMI reveals insights that conventional cell motility analyses may overlook, showcasing its utility in uncovering robust biological insights. Together, a multivariate framework is presented to classify emergent patterns of single-cell motility, emphasizing the critical role of cellular heterogeneity in shaping cell behaviors across populations., (© 2024 The Author(s). Advanced Science published by Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
19. Genetic mechanisms underlying tumor microenvironment composition and function in diffuse large B-cell lymphoma.
- Author
-
Cerchietti L
- Subjects
- Humans, Prognosis, Tumor Microenvironment genetics, Lymphoma, Large B-Cell, Diffuse pathology
- Abstract
Abstract: Cells in the tumor microenvironment (TME) of diffuse large B-cell lymphoma (DLBCL) show enormous diversity and plasticity, with functions that can range from tumor inhibitory to tumor supportive. The patient's age, immune status, and DLBCL treatments are factors that contribute to the shaping of this TME, but evidence suggests that genetic factors, arising principally in lymphoma cells themselves, are among the most important. Here, we review the current understanding of the role of these genetic drivers of DLBCL in establishing and modulating the lymphoma microenvironment. A better comprehension of the relationship between lymphoma genetic factors and TME biology should lead to better therapeutic interventions, especially immunotherapies., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
20. Germinal Center Dark Zone harbors ATR-dependent determinants of T-cell exclusion that are also identified in aggressive lymphoma.
- Author
-
Cancila V, Morello G, Bertolazzi G, Chan AS, Bastianello G, Paysan D, Jaynes PW, Schiavoni G, Mattei F, Piconese S, Revuelta MV, Noto F, De Ninno A, Cammarata I, Pagni F, Venkatachalapathy S, Sangaletti S, Di Napoli A, Vacca D, Lonardi S, Lorenzi L, Ferreri AJM, Belmonte B, Varano G, Colombo MP, Bicciato S, Inghirami G, Cerchietti L, Ponzoni M, Zappasodi R, Facchetti F, Foiani M, Casola S, Jeyasekharan AD, and Tripodo C
- Abstract
The germinal center (GC) dark zone (DZ) and light zone (LZ) regions spatially separate expansion and diversification from selection of antigen-specific B-cells to ensure antibody affinity maturation and B cell memory. The DZ and LZ differ significantly in their immune composition despite the lack of a physical barrier, yet the determinants of this polarization are poorly understood. This study provides novel insights into signals controlling asymmetric T-cell distribution between DZ and LZ regions. We identify spatially-resolved DNA damage response and chromatin compaction molecular features that underlie DZ T-cell exclusion. The DZ spatial transcriptional signature linked to T-cell immune evasion clustered aggressive Diffuse Large B-cell Lymphomas (DLBCL) for differential T cell infiltration. We reveal the dependence of the DZ transcriptional core signature on the ATR kinase and dissect its role in restraining inflammatory responses contributing to establishing an immune-repulsive imprint in DLBCL. These insights may guide ATR-focused treatment strategies bolstering immunotherapy in tumors marked by DZ transcriptional and chromatin-associated features., Competing Interests: CONFLICT OF INTERESTS ADJ has received consultancy fees from DKSH/Beigene, Roche, Gilead, Turbine Ltd, AstraZeneca, Antengene, Janssen, MSD and IQVIA; and research funding from Janssen and AstraZeneca.
- Published
- 2024
- Full Text
- View/download PDF
21. XPO1 Enables Adaptive Regulation of mRNA Export Required for Genotoxic Stress Tolerance in Cancer Cells.
- Author
-
Marullo R, Rutherford SC, Revuelta MV, Zamponi N, Culjkovic-Kraljacic B, Kotlov N, Di Siervi N, Lara-Garcia J, Allan JN, Ruan J, Furman RR, Chen Z, Shore TB, Phillips AA, Mayer S, Hsu J, van Besien K, Leonard JP, Borden KLB, Inghirami G, Martin P, and Cerchietti L
- Subjects
- Humans, Active Transport, Cell Nucleus, Karyopherins metabolism, Cell Line, Tumor, Hydrazines pharmacology, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear metabolism, DNA Damage, RNA, Messenger genetics, RNA, Messenger metabolism, Lymphoma, Non-Hodgkin drug therapy, Lymphoma, Large B-Cell, Diffuse drug therapy
- Abstract
Exportin-1 (XPO1), the main soluble nuclear export receptor in eukaryotic cells, is frequently overexpressed in diffuse large B-cell lymphoma (DLBCL). A selective XPO1 inhibitor, selinexor, received approval as single agent for relapsed or refractory (R/R) DLBCL. Elucidating the mechanisms by which XPO1 overexpression supports cancer cells could facilitate further clinical development of XPO1 inhibitors. We uncovered here that XPO1 overexpression increases tolerance to genotoxic stress, leading to a poor response to chemoimmunotherapy. Upon DNA damage induced by MYC expression or exogenous compounds, XPO1 bound and exported EIF4E and THOC4 carrying DNA damage repair mRNAs, thereby increasing synthesis of DNA damage repair proteins under conditions of increased turnover. Consequently, XPO1 inhibition decreased the capacity of lymphoma cells to repair DNA damage and ultimately resulted in increased cytotoxicity. In a phase I clinical trial conducted in R/R DLBCL, the combination of selinexor with second-line chemoimmunotherapy was tolerated with early indication of efficacy. Overall, this study reveals that XPO1 overexpression plays a critical role in the increased tolerance of cancer cells to DNA damage while providing new insights to optimize the clinical development of XPO1 inhibitors., Significance: XPO1 regulates the dynamic ribonucleoprotein nuclear export in response to genotoxic stress to support tolerance and can be targeted to enhance the sensitivity of cancer cells to endogenous and exogenous DNA damage. See related commentary by Knittel and Reinhardt, p. 3., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
22. Multicenter phase 2 study of oral azacitidine (CC-486) plus CHOP as initial treatment for PTCL.
- Author
-
Ruan J, Moskowitz A, Mehta-Shah N, Sokol L, Chen Z, Kotlov N, Nos G, Sorokina M, Maksimov V, Sboner A, Sigouros M, van Besien K, Horwitz S, Rutherford SC, Mulvey E, Revuelta MV, Xiang J, Alonso A, Melnick A, Elemento O, Inghirami G, Leonard JP, Cerchietti L, and Martin P
- Subjects
- Humans, Azacitidine adverse effects, Doxorubicin, Prednisone adverse effects, Vincristine, Cyclophosphamide adverse effects, Immunologic Factors therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Tumor Microenvironment, Lymphoma, T-Cell, Peripheral pathology
- Abstract
Peripheral T-cell lymphomas (PTCL) with T-follicular helper phenotype (PTCL-TFH) has recurrent mutations affecting epigenetic regulators, which may contribute to aberrant DNA methylation and chemoresistance. This phase 2 study evaluated oral azacitidine (CC-486) plus cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) as initial treatment for PTCL. CC-486 at 300 mg daily was administered for 7 days before C1 of CHOP, and for 14 days before CHOP C2-6. The primary end point was end-of-treatment complete response (CR). Secondary end points included safety and survival. Correlative studies assessed mutations, gene expression, and methylation in tumor samples. Grade 3 to 4 hematologic toxicities were mostly neutropenia (71%), with febrile neutropenia uncommon (14%). Nonhematologic toxicities included fatigue (14%) and gastrointestinal symptoms (5%). In 20 evaluable patients, CR was 75%, including 88.2% for PTCL-TFH (n = 17). The 2-year progression-free survival (PFS) was 65.8% for all and 69.2% for PTCL-TFH, whereas 2-year overall survival (OS) was 68.4% for all and 76.1% for PTCL-TFH. The frequencies of the TET2, RHOA, DNMT3A, and IDH2 mutations were 76.5%, 41.1%, 23.5%, and 23.5%, respectively, with TET2 mutations significantly associated with CR (P = .007), favorable PFS (P = .004) and OS (P = .015), and DNMT3A mutations associated with adverse PFS (P = .016). CC-486 priming contributed to the reprograming of the tumor microenvironment by upregulation of genes related to apoptosis (P < .01) and inflammation (P < .01). DNA methylation did not show significant shift. This safe and active regimen is being further evaluated in the ALLIANCE randomized study A051902 in CD30-negative PTCL. This trial was registered at www.clinicaltrials.gov as #NCT03542266., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
23. The eukaryotic translation initiation factor eIF4E reprograms alternative splicing.
- Author
-
Ghram M, Morris G, Culjkovic-Kraljacic B, Mars JC, Gendron P, Skrabanek L, Revuelta MV, Cerchietti L, Guzman ML, and Borden KLB
- Subjects
- Humans, RNA Splicing Factors metabolism, Eukaryotic Initiation Factor-4E metabolism, RNA Splicing, Eukaryotic Initiation Factors genetics, Mutation, Alternative Splicing, Leukemia, Myeloid, Acute genetics
- Abstract
Aberrant splicing is typically attributed to splice-factor (SF) mutation and contributes to malignancies including acute myeloid leukemia (AML). Here, we discovered a mutation-independent means to extensively reprogram alternative splicing (AS). We showed that the dysregulated expression of eukaryotic translation initiation factor eIF4E elevated selective splice-factor production, thereby impacting multiple spliceosome complexes, including factors mutated in AML such as SF3B1 and U2AF1. These changes generated a splicing landscape that predominantly supported altered splice-site selection for ~800 transcripts in cell lines and ~4,600 transcripts in specimens from high-eIF4E AML patients otherwise harboring no known SF mutations. Nuclear RNA immunoprecipitations, export assays, polysome analyses, and mutational studies together revealed that eIF4E primarily increased SF production via its nuclear RNA export activity. By contrast, eIF4E dysregulation did not induce known SF mutations or alter spliceosome number. eIF4E interacted with the spliceosome and some pre-mRNAs, suggesting its direct involvement in specific splicing events. eIF4E induced simultaneous effects on numerous SF proteins, resulting in a much larger range of splicing alterations than in the case of mutation or dysregulation of individual SFs and providing a novel paradigm for splicing control and dysregulation., (© 2023 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
24. Preclinical models for prediction of immunotherapy outcomes and immune evasion mechanisms in genetically heterogeneous multiple myeloma.
- Author
-
Larrayoz M, Garcia-Barchino MJ, Celay J, Etxebeste A, Jimenez M, Perez C, Ordoñez R, Cobaleda C, Botta C, Fresquet V, Roa S, Goicoechea I, Maia C, Lasaga M, Chesi M, Bergsagel PL, Larrayoz MJ, Calasanz MJ, Campos-Sanchez E, Martinez-Cano J, Panizo C, Rodriguez-Otero P, Vicent S, Roncador G, Gonzalez P, Takahashi S, Katz SG, Walensky LD, Ruppert SM, Lasater EA, Amann M, Lozano T, Llopiz D, Sarobe P, Lasarte JJ, Planell N, Gomez-Cabrero D, Kudryashova O, Kurilovich A, Revuelta MV, Cerchietti L, Agirre X, San Miguel J, Paiva B, Prosper F, and Martinez-Climent JA
- Subjects
- Mice, Animals, CD8-Positive T-Lymphocytes, Immune Evasion, T-Lymphocytes, Regulatory, Immunotherapy adverse effects, Tumor Microenvironment genetics, Multiple Myeloma therapy, Multiple Myeloma drug therapy
- Abstract
The historical lack of preclinical models reflecting the genetic heterogeneity of multiple myeloma (MM) hampers the advance of therapeutic discoveries. To circumvent this limitation, we screened mice engineered to carry eight MM lesions (NF-κB, KRAS, MYC, TP53, BCL2, cyclin D1, MMSET/NSD2 and c-MAF) combinatorially activated in B lymphocytes following T cell-driven immunization. Fifteen genetically diverse models developed bone marrow (BM) tumors fulfilling MM pathogenesis. Integrative analyses of ∼500 mice and ∼1,000 patients revealed a common MAPK-MYC genetic pathway that accelerated time to progression from precursor states across genetically heterogeneous MM. MYC-dependent time to progression conditioned immune evasion mechanisms that remodeled the BM microenvironment differently. Rapid MYC-driven progressors exhibited a high number of activated/exhausted CD8
+ T cells with reduced immunosuppressive regulatory T (Treg ) cells, while late MYC acquisition in slow progressors was associated with lower CD8+ T cell infiltration and more abundant Treg cells. Single-cell transcriptomics and functional assays defined a high ratio of CD8+ T cells versus Treg cells as a predictor of response to immune checkpoint blockade (ICB). In clinical series, high CD8+ T/Treg cell ratios underlie early progression in untreated smoldering MM, and correlated with early relapse in newly diagnosed patients with MM under Len/Dex therapy. In ICB-refractory MM models, increasing CD8+ T cell cytotoxicity or depleting Treg cells reversed immunotherapy resistance and yielded prolonged MM control. Our experimental models enable the correlation of MM genetic and immunological traits with preclinical therapy responses, which may inform the next-generation immunotherapy trials., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
25. Endothelial cell-leukemia interactions remodel drug responses, uncovering T-ALL vulnerabilities.
- Author
-
Cappelli LV, Fiore D, Phillip JM, Yoffe L, Di Giacomo F, Chiu W, Hu Y, Kayembe C, Ginsberg M, Consolino L, Barcia Duran JG, Zamponi N, Melnick AM, Boccalatte F, Tam W, Elemento O, Chiaretti S, Guarini A, Foà R, Cerchietti L, Rafii S, and Inghirami G
- Subjects
- Humans, Cell Communication, Coculture Techniques, Tumor Microenvironment, Endothelial Cells metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism
- Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive and often incurable disease. To uncover therapeutic vulnerabilities, we first developed T-ALL patient-derived tumor xenografts (PDXs) and exposed PDX cells to a library of 433 clinical-stage compounds in vitro. We identified 39 broadly active drugs with antileukemia activity. Because endothelial cells (ECs) can alter drug responses in T-ALL, we developed an EC/T-ALL coculture system. We found that ECs provide protumorigenic signals and mitigate drug responses in T-ALL PDXs. Whereas ECs broadly rescued several compounds in most models, for some drugs the rescue was restricted to individual PDXs, suggesting unique crosstalk interactions and/or intrinsic tumor features. Mechanistically, cocultured T-ALL cells and ECs underwent bidirectional transcriptomic changes at the single-cell level, highlighting distinct "education signatures." These changes were linked to bidirectional regulation of multiple pathways in T-ALL cells as well as in ECs. Remarkably, in vitro EC-educated T-ALL cells transcriptionally mirrored ex vivo splenic T-ALL at single-cell resolution. Last, 5 effective drugs from the 2 drug screenings were tested in vivo and shown to effectively delay tumor growth and dissemination thus prolonging overall survival. In sum, we developed a T-ALL/EC platform that elucidated leukemia-microenvironment interactions and identified effective compounds and therapeutic vulnerabilities., (© 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
26. Inhibition of Integrin αVβ3 Signaling Improves the Antineoplastic Effect of Bexarotene in Cutaneous T-Cell Lymphoma.
- Author
-
Cayrol F, Revuelta MV, Debernardi M, Paulazo A, Phillip JM, Zamponi N, Sterle H, Díaz Flaqué MC, Magro C, Marullo R, Mulvey E, Ruan J, Cremaschi GA, and Cerchietti L
- Subjects
- Bexarotene pharmacology, Bexarotene therapeutic use, Humans, Integrin alphaVbeta3, Tetrahydronaphthalenes pharmacology, Tetrahydronaphthalenes therapeutic use, Thyroxine therapeutic use, Anticarcinogenic Agents pharmacology, Anticarcinogenic Agents therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Lymphoma, T-Cell, Cutaneous drug therapy, Lymphoma, T-Cell, Cutaneous pathology, Skin Neoplasms pathology
- Abstract
Bexarotene is a specific retinoid X receptor agonist that has been used for the treatment of cutaneous T-cell lymphoma (CTCL). Because bexarotene causes hypothyroidism, it requires the administration of levothyroxine. However, levothyroxine, in addition to its ubiquitous nuclear receptors, can activate the αVβ3 integrin that is overexpressed in CTCL, potentially interfering the antineoplastic effect of bexarotene. We thus investigated the biological effect of levothyroxine in relation to bexarotene treatment. Although in isolated CTCL cells levothyroxine decreased, in an αVβ3-dependent manner, the antineoplastic effect of bexarotene, levothyroxine supplementation in preclinical models was necessary to avoid suppression of lymphoma immunity. Accordingly, selective genetic and pharmacologic inhibition of integrin αVβ3 improved the antineoplastic effect of bexarotene plus levothyroxine replacement while maintaining lymphoma immunity. Our results provide a mechanistic rationale for clinical testing of integrin αVβ3 inhibitors as part of CTCL regimens based on bexarotene administration., Teaser: Inhibiting αVβ3 integrin improves the antineoplastic effect of bexarotene while maintaining lymphoma immunity., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
27. Precise reconstruction of the TME using bulk RNA-seq and a machine learning algorithm trained on artificial transcriptomes.
- Author
-
Zaitsev A, Chelushkin M, Dyikanov D, Cheremushkin I, Shpak B, Nomie K, Zyrin V, Nuzhdina E, Lozinsky Y, Zotova A, Degryse S, Kotlov N, Baisangurov A, Shatsky V, Afenteva D, Kuznetsov A, Paul SR, Davies DL, Reeves PM, Lanuti M, Goldberg MF, Tazearslan C, Chasse M, Wang I, Abdou M, Aslanian SM, Andrewes S, Hsieh JJ, Ramachandran A, Lyu Y, Galkin I, Svekolkin V, Cerchietti L, Poznansky MC, Ataullakhanov R, Fowler N, and Bagaev A
- Subjects
- Algorithms, CD8-Positive T-Lymphocytes, Humans, Machine Learning, RNA-Seq, Sequence Analysis, RNA, Tumor Microenvironment genetics, Neoplasms genetics, Transcriptome
- Abstract
Cellular deconvolution algorithms virtually reconstruct tissue composition by analyzing the gene expression of complex tissues. We present the decision tree machine learning algorithm, Kassandra, trained on a broad collection of >9,400 tissue and blood sorted cell RNA profiles incorporated into millions of artificial transcriptomes to accurately reconstruct the tumor microenvironment (TME). Bioinformatics correction for technical and biological variability, aberrant cancer cell expression inclusion, and accurate quantification and normalization of transcript expression increased Kassandra stability and robustness. Performance was validated on 4,000 H&E slides and 1,000 tissues by comparison with cytometric, immunohistochemical, or single-cell RNA-seq measurements. Kassandra accurately deconvolved TME elements, showing the role of these populations in tumor pathogenesis and other biological processes. Digital TME reconstruction revealed that the presence of PD-1-positive CD8
+ T cells strongly correlated with immunotherapy response and increased the predictive potential of established biomarkers, indicating that Kassandra could potentially be utilized in future clinical applications., Competing Interests: Declaration of interests N.F. is the Chief Medical Officer of BostonGene, Corp. and a professor at the University of Texas MD Anderson Cancer Center. A. Zaitsev, M. Chelushkin, V.Z., B.S., D.D., E. Nuzhdina, A. Bagaev, and R.A. are inventors on patent applications related to Kassandra. All other authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
28. BCL10 Mutations Define Distinct Dependencies Guiding Precision Therapy for DLBCL.
- Author
-
Xia M, David L, Teater M, Gutierrez J, Wang X, Meydan C, Lytle A, Slack GW, Scott DW, Morin RD, Onder O, Elenitoba-Johnson KSJ, Zamponi N, Cerchietti L, Lu T, Philippar U, Fontan L, Wu H, and Melnick AM
- Subjects
- CARD Signaling Adaptor Proteins genetics, Guanylate Cyclase genetics, Humans, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein genetics, Mutation, Signal Transduction, B-Cell CLL-Lymphoma 10 Protein genetics, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology
- Abstract
Activated B cell-like diffuse large B-cell lymphomas (ABC-DLBCL) have unfavorable outcomes and chronic activation of CARD11-BCL10-MALT1 (CBM) signal amplification complexes that form due to polymerization of BCL10 subunits, which is affected by recurrent somatic mutations in ABC-DLBCLs. Herein, we show that BCL10 mutants fall into at least two functionally distinct classes: missense mutations of the BCL10 CARD domain and truncation of its C-terminal tail. Truncating mutations abrogated a motif through which MALT1 inhibits BCL10 polymerization, trapping MALT1 in its activated filament-bound state. CARD missense mutations enhanced BCL10 filament formation, forming glutamine network structures that stabilize BCL10 filaments. Mutant forms of BCL10 were less dependent on upstream CARD11 activation and thus manifested resistance to BTK inhibitors, whereas BCL10 truncating but not CARD mutants were hypersensitive to MALT1 inhibitors. Therefore, BCL10 mutations are potential biomarkers for BTK inhibitor resistance in ABC-DLBCL, and further precision can be achieved by selecting therapy based on specific biochemical effects of distinct mutation classes., Significance: ABC-DLBCLs feature frequent mutations of signaling mediators that converge on the CBM complex. We use structure-function approaches to reveal that BCL10 mutations fall into two distinct biochemical classes. Both classes confer resistance to BTK inhibitors, whereas BCL10 truncations confer hyperresponsiveness to MALT1 inhibitors, providing a road map for precision therapies in ABC-DLBCLs. See related commentary by Phelan and Oellerich, p. 1844. This article is highlighted in the In This Issue feature, p. 1825., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
29. Variational autoencoders learn transferrable representations of metabolomics data.
- Author
-
Gomari DP, Schweickart A, Cerchietti L, Paietta E, Fernandez H, Al-Amin H, Suhre K, and Krumsiek J
- Subjects
- Humans, Metabolomics, Principal Component Analysis, Diabetes Mellitus, Type 2
- Abstract
Dimensionality reduction approaches are commonly used for the deconvolution of high-dimensional metabolomics datasets into underlying core metabolic processes. However, current state-of-the-art methods are widely incapable of detecting nonlinearities in metabolomics data. Variational Autoencoders (VAEs) are a deep learning method designed to learn nonlinear latent representations which generalize to unseen data. Here, we trained a VAE on a large-scale metabolomics population cohort of human blood samples consisting of over 4500 individuals. We analyzed the pathway composition of the latent space using a global feature importance score, which demonstrated that latent dimensions represent distinct cellular processes. To demonstrate model generalizability, we generated latent representations of unseen metabolomics datasets on type 2 diabetes, acute myeloid leukemia, and schizophrenia and found significant correlations with clinical patient groups. Notably, the VAE representations showed stronger effects than latent dimensions derived by linear and non-linear principal component analysis. Taken together, we demonstrate that the VAE is a powerful method that learns biologically meaningful, nonlinear, and transferrable latent representations of metabolomics data., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
30. Characterization of GECPAR, a noncoding RNA that regulates the transcriptional program of diffuse large B-cell lymphoma.
- Author
-
Napoli S, Cascione L, Rinaldi A, Spriano F, Guidetti F, Zhang F, Cacciapuoti MT, Mensah AA, Sartori G, Munz N, Forcato M, Bicciato S, Chiappella A, Ghione P, Elemento O, Cerchietti L, Inghirami G, and Bertoni F
- Subjects
- Humans, RNA genetics, RNA, Untranslated, Transcription, Genetic, Enhancer Elements, Genetic, Lymphoma, Large B-Cell, Diffuse genetics
- Abstract
Enhancers are regulatory regions of DNA, which play a key role in cell-type specific differentiation and development. Most active enhancers are transcribed into enhancer RNA (eRNA) that can regulate transcription of target genes by means of in cis as well as in trans action. eRNA stabilize contacts between distal genomic regions and mediate the interaction of DNA with master transcription factors. Here, we characterized an enhancer eRNA, GECPAR (germinal center proliferative adapter RNA), which is specifically transcribed in normal and neoplastic germinal center B cells from the super-enhancer of POU2AF1, a key regulatory gene of the germinal center reaction. Using diffuse large B-cell lymphoma cell line models, we demonstrated the tumor suppressor activity of GECPAR, which is mediated via its transcriptional regulation of proliferation and differentiation genes, particularly MYC and the Wnt pathway.
- Published
- 2022
- Full Text
- View/download PDF
31. SWOG 1918: A phase II/III randomized study of R-miniCHOP with or without oral azacitidine (CC-486) in participants age 75 years or older with newly diagnosed aggressive non-Hodgkin lymphomas - Aiming to improve therapy, outcomes, and validate a prospective frailty tool.
- Author
-
Brem EA, Li H, Beaven AW, Caimi PF, Cerchietti L, Alizadeh AA, Olin R, Henry NL, Dillon H, Little RF, Laubach C, LeBlanc M, Friedberg JW, and Smith SM
- Subjects
- Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Azacitidine adverse effects, Humans, Prospective Studies, Rituximab adverse effects, Frailty, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Non-Hodgkin drug therapy
- Abstract
Diffuse large B cell lymphoma (DLBCL) is an aggressive but potentially curable malignancy; however, cure is highly dependent on the ability to deliver intensive, anthracycline-based chemoimmunotherapy. Nearly one third of cases of DLBCL occur in patients over age 75 years, and advanced age is an important adverse feature in prognostic models. Despite this incidence in older patients, there is no clear accepted standard of care due to under-representation of this group in large randomized clinical trials. Furthermore, insufficient assessments of baseline frailty and prediction of toxicity hamper clinical decision-making. Here, we present an ongoing randomized study of R-miniCHOP chemoimmunotherapy with or without oral azacitidine (CC-486, Onureg) for patients age 75 and older with newly diagnosed DLBCL and associated aggressive lymphomas. The incorporation of an oral hypomethylating agent is based on increased tumor methylation as a biologic feature of older patients with DLBCL and a desire to minimize the injection burden for this population. This is the first randomized study in this population conducted in North America by the National Clinical Trials Network (NCTN) and will enroll up to 422 patients including 40 patients in a safety run-in phase. This study incorporates an objective assessment of baseline frailty (the FIL Tool) and a serial comprehensive geriatric assessment (CGA). Key correlative tests will include circulating tumor DNA (ctDNA) assays at pre-specified timepoints to explore if ctDNA quantity and methylation patterns correlate with response. S1918 has the potential to impact future trial design and to change the standard of care for patients 75 years and older with aggressive lymphoma given its randomized design, prospective incorporation of geriatric assessments, and exploration of ctDNA correlatives. Trial registration: The trial is registered with ClinicalTrial.gov Identifier NCT04799275., (Copyright © 2021 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
32. Phase 1 study of oral azacitidine (CC-486) plus R-CHOP in previously untreated intermediate- to high-risk DLBCL.
- Author
-
Martin P, Bartlett NL, Chavez JC, Reagan JL, Smith SM, LaCasce AS, Jones J, Drew J, Wu C, Mulvey E, Revuelta MV, Cerchietti L, and Leonard JP
- Subjects
- Administration, Oral, Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Azacitidine adverse effects, Cyclophosphamide administration & dosage, Cyclophosphamide adverse effects, Disease-Free Survival, Doxorubicin administration & dosage, Doxorubicin adverse effects, Female, Follow-Up Studies, Humans, Male, Maximum Tolerated Dose, Middle Aged, Prednisone administration & dosage, Prednisone adverse effects, Risk Factors, Rituximab administration & dosage, Rituximab adverse effects, Survival Rate, Vincristine administration & dosage, Vincristine adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Azacitidine administration & dosage, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse mortality
- Abstract
Resistance to standard immunochemotherapy remains an unmet challenge in diffuse large B-cell lymphoma (DLBCL), and aberrant DNA methylation may contribute to chemoresistance. Promising early-phase results were reported with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) plus subcutaneous azacitidine, a hypomethylating agent. In this phase 1 study, we evaluated CC-486 (oral azacitidine) plus 6 cycles of R-CHOP in patients with previously untreated intermediate- to high-risk DLBCL or grade 3B/transformed follicular lymphoma. CC-486 doses of 100, 150, 200, or 300 mg given 7 days before cycle 1 and on days 8-21 of cycles 1-5 were evaluated; additional patients were enrolled in the expansion phase to examine preliminary efficacy. The primary objectives were to determine the safety and the maximum tolerated dose (MTD) of CC-486 in combination with R-CHOP. The most common grade 3/4 toxicities were hematologic, including neutropenia (62.7%) and febrile neutropenia (25.4%); grade 3/4 nonhematologic toxicities were uncommon (<7%). The MTD was not established; 2 patients had dose-limiting toxicities (1 with grade 4 febrile neutropenia; 1 with grade 4 prolonged neutropenia). The recommended phase 2 dose was established as 300 mg. The overall response rate was 94.9%, with 52 patients (88.1%) achieving complete responses. With a median follow-up of 28.9 months, estimated 1- and 2-year progression-free survival rates were 84.1% and 78.6%, respectively. Overall, epigenetic priming with CC-486 before R-CHOP can be delivered with acceptable safety to patients with previously untreated intermediate- to high-risk DLBCL or grade 3B/transformed follicular lymphoma. ClinicalTrials.gov: NCT02343536., (© 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
33. Histamine H4 Receptor Agonism Induces Antitumor Effects in Human T-Cell Lymphoma.
- Author
-
Clauzure M, Táquez Delgado MA, Phillip JM, Revuelta MV, Cerchietti L, and Medina VA
- Subjects
- Apoptosis drug effects, Cell Line, Cell Line, Tumor, Cell Proliferation drug effects, HEK293 Cells, Histamine metabolism, Histamine Antagonists pharmacology, Humans, Lymphoma, T-Cell metabolism, Oxidative Stress drug effects, Antineoplastic Agents pharmacology, Histamine Agonists pharmacology, Lymphoma, T-Cell drug therapy, Receptors, Histamine H4 metabolism
- Abstract
The discovery of the human histamine H4 receptor (H4R) has contributed to our understanding of the role of histamine in numerous physiological and pathological conditions, including tumor development and progression. The lymph nodes of patients with malignant lymphomas have shown to contain high levels of histamine, however, less is known regarding the expression and function of the H4R in T-cell lymphoma (TCL). In this work we demonstrate the expression of H4R isoforms (mRNA and protein) in three human aggressive TCL (OCI-Ly12, Karpas 299, and HuT78). Histamine and specific H4R agonists (VUF8430 and JNJ28610244) significantly reduced cell viability in a dose-dependent manner ( p < 0.05). The combined treatment with the H4R antagonist (JNJ7777120, 10 µM) reversed the effects of the H4R ligands. Importantly, we screened a drug repurposing library of 433 FDA-approved compounds (1 μM) in combination with histamine (10 μM) in Hut78 cells. Histamine produced a favorable antitumor effect with 18 of these compounds, including the histone deacetylase inhibitor panobinostat. Apoptosis, proliferation, and oxidative stress studies confirmed the antitumoral effects of the combination. We conclude that the H4R is expressed in TCL, and it is involved in histamine-mediated responses.
- Published
- 2022
- Full Text
- View/download PDF
34. Translational Activation of ATF4 through Mitochondrial Anaplerotic Metabolic Pathways Is Required for DLBCL Growth and Survival.
- Author
-
Li M, Teater MR, Hong JY, Park NR, Duy C, Shen H, Wang L, Chen Z, Cerchietti L, Davidson SM, Lin H, and Melnick AM
- Subjects
- Activating Transcription Factor 4 genetics, Amino Acids metabolism, Citric Acid Cycle, Glutamine metabolism, Humans, Mitochondria metabolism, Lymphoma, Large B-Cell, Diffuse genetics, Sirtuin 3 genetics
- Abstract
Diffuse large B-cell lymphomas (DLBCL) are broadly dependent on anaplerotic metabolism regulated by mitochondrial SIRT3. Herein we find that translational upregulation of ATF4 is coupled with anaplerotic metabolism in DLBCLs due to nutrient deprivation caused by SIRT3 driving rapid flux of glutamine into the tricarboxylic acid (TCA) cycle. SIRT3 depletion led to ATF4 downregulation and cell death, which was rescued by ectopic ATF4 expression. Mechanistically, ATF4 translation is inhibited in SIRT3-deficient cells due to the increased pools of amino acids derived from compensatory autophagy and decreased glutamine consumption by the TCA cycle. Absence of ATF4 further aggravates this state through downregulation of its target genes, including genes for amino acid biosynthesis and import. Collectively, we identify a SIRT3-ATF4 axis required to maintain survival of DLBCL cells by enabling them to optimize amino acid uptake and utilization. Targeting ATF4 translation can potentiate the cytotoxic effect of SIRT3 inhibitor to DLBCL cells. SIGNIFICANCE: We discovered the link between SIRT3 and ATF4 in DLBCL cells, which connected lymphoma amino acid metabolism with ATF4 translation via metabolic stress signals. SIRT3-ATF4 axis is required in DLBCL cells regardless of subtype, which indicates a common metabolic vulnerability in DLBCLs and can serve as a therapeutic target. This article is highlighted in the In This Issue feature, p. 1 ., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
35. The metabolic adaptation evoked by arginine enhances the effect of radiation in brain metastases.
- Author
-
Marullo R, Castro M, Yomtoubian S, Calvo-Vidal MN, Revuelta MV, Krumsiek J, Cho A, Morgado PC, Yang S, Medina V, Roth BM, Bonomi M, Keshari KR, Mittal V, Navigante A, and Cerchietti L
- Abstract
Selected patients with brain metastases (BM) are candidates for radiotherapy. A lactatogenic metabolism, common in BM, has been associated with radioresistance. We demonstrated that BM express nitric oxide (NO) synthase 2 and that administration of its substrate l-arginine decreases tumor lactate in BM patients. In a placebo-controlled trial, we showed that administration of l-arginine before each fraction enhanced the effect of radiation, improving the control of BM. Studies in preclinical models demonstrated that l-arginine radiosensitization is a NO-mediated mechanism secondary to the metabolic adaptation induced in cancer cells. We showed that the decrease in tumor lactate was a consequence of reduced glycolysis that also impacted ATP and NAD
+ levels. These effects were associated with NO-dependent inhibition of GAPDH and hyperactivation of PARP upon nitrosative DNA damage. These metabolic changes ultimately impaired the repair of DNA damage induced by radiation in cancer cells while greatly sparing tumor-infiltrating lymphocytes.- Published
- 2021
- Full Text
- View/download PDF
36. Oncogenic HSP90 Facilitates Metabolic Alterations in Aggressive B-cell Lymphomas.
- Author
-
Calvo-Vidal MN, Zamponi N, Krumsiek J, Stockslager MA, Revuelta MV, Phillip JM, Marullo R, Tikhonova E, Kotlov N, Patel J, Yang SN, Yang L, Taldone T, Thieblemont C, Leonard JP, Martin P, Inghirami G, Chiosis G, Manalis SR, and Cerchietti L
- Subjects
- Animals, Carcinogenesis metabolism, Case-Control Studies, HSP90 Heat-Shock Proteins genetics, Humans, Lymphoma, Large B-Cell, Diffuse genetics, Mice, Protein Interaction Domains and Motifs, Proto-Oncogene Proteins c-myc genetics, Signal Transduction, Tumor Cells, Cultured, Carcinogenesis pathology, HSP90 Heat-Shock Proteins metabolism, Lymphoma, Large B-Cell, Diffuse metabolism, Lymphoma, Large B-Cell, Diffuse pathology, Metabolome, Proteolysis, Proto-Oncogene Proteins c-myc metabolism
- Abstract
HSP90 is critical for maintenance of the cellular proteostasis. In cancer cells, HSP90 also becomes a nucleating site for the stabilization of multiprotein complexes including signaling pathways and transcription complexes. Here we described the role of this HSP90 form, referred to as oncogenic HSP90, in the regulation of cytosolic metabolic pathways in proliferating B-cell lymphoma cells. Oncogenic HSP90 assisted in the organization of metabolic enzymes into non-membrane-bound functional compartments. Under experimental conditions that conserved cellular proteostasis, oncogenic HSP90 coordinated and sustained multiple metabolic pathways required for energy production and maintenance of cellular biomass as well as for secretion of extracellular metabolites. Conversely, inhibition of oncogenic HSP90, in absence of apparent client protein degradation, decreased the efficiency of MYC-driven metabolic reprogramming. This study reveals that oncogenic HSP90 supports metabolism in B-cell lymphoma cells and patients with diffuse large B-cell lymphoma, providing a novel mechanism of activity for HSP90 inhibitors. SIGNIFICANCE: The oncogenic form of HSP90 organizes and maintains functional multienzymatic metabolic hubs in cancer cells, suggesting the potential of repurposing oncogenic HSP90 selective inhibitors to disrupt metabolism in lymphoma cells., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
37. Clinical and Biological Subtypes of B-cell Lymphoma Revealed by Microenvironmental Signatures.
- Author
-
Kotlov N, Bagaev A, Revuelta MV, Phillip JM, Cacciapuoti MT, Antysheva Z, Svekolkin V, Tikhonova E, Miheecheva N, Kuzkina N, Nos G, Tabbo F, Frenkel F, Ghione P, Tsiper M, Almog N, Fowler N, Melnick AM, Leonard JP, Inghirami G, and Cerchietti L
- Subjects
- Gene Expression Profiling, Humans, Lymphoma, Large B-Cell, Diffuse pathology, Tumor Microenvironment, Lymphoma, Large B-Cell, Diffuse genetics
- Abstract
Diffuse large B-cell lymphoma (DLBCL) is a biologically and clinically heterogeneous disease. Transcriptomic and genetic characterization of DLBCL has increased the understanding of its intrinsic pathogenesis and provided potential therapeutic targets. However, the role of the microenvironment in DLBCL biology remains less understood. Here, we performed a transcriptomic analysis of the microenvironment of 4,655 DLBCLs from multiple independent cohorts and described four major lymphoma microenvironment categories that associate with distinct biological aberrations and clinical behavior. We also found evidence of genetic and epigenetic mechanisms deployed by cancer cells to evade microenvironmental constraints of lymphoma growth, supporting the rationale for implementing DNA hypomethylating agents in selected patients with DLBCL. In addition, our work uncovered new therapeutic vulnerabilities in the biochemical composition of the extracellular matrix that were exploited to decrease DLBCL proliferation in preclinical models. This novel classification provides a road map for the biological characterization and therapeutic exploitation of the DLBCL microenvironment. SIGNIFICANCE: In a translational relevant transcriptomic-based classification, we characterized the microenvironment as a critical component of the B-cell lymphoma biology and associated it with the DLBCL clinical behavior establishing a novel opportunity for targeting therapies. This article is highlighted in the In This Issue feature, p. 1307 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
38. The eukaryotic translation initiation factor eIF4E elevates steady-state m 7 G capping of coding and noncoding transcripts.
- Author
-
Culjkovic-Kraljacic B, Skrabanek L, Revuelta MV, Gasiorek J, Cowling VH, Cerchietti L, and Borden KLB
- Subjects
- Cell Line, Tumor, Eukaryotic Initiation Factor-4E chemistry, Eukaryotic Initiation Factor-4E genetics, Guanosine chemistry, Guanosine genetics, Guanosine metabolism, Humans, Polyribosomes metabolism, RNA Caps chemistry, RNA Caps genetics, RNA, Messenger chemistry, RNA, Messenger genetics, Transcriptome genetics, Eukaryotic Initiation Factor-4E metabolism, Guanosine analogs & derivatives, RNA Caps metabolism, RNA, Messenger metabolism
- Abstract
Methyl-7-guanosine (m
7 G) "capping" of coding and some noncoding RNAs is critical for their maturation and subsequent activity. Here, we discovered that eukaryotic translation initiation factor 4E (eIF4E), itself a cap-binding protein, drives the expression of the capping machinery and increased capping efficiency of ∼100 coding and noncoding RNAs. To quantify this, we developed enzymatic (cap quantification; CapQ) and quantitative cap immunoprecipitation (CapIP) methods. The CapQ method has the further advantage that it captures information about capping status independent of the type of 5' cap, i.e., it is not restricted to informing on m7 G caps. These methodological advances led to unanticipated revelations: 1) Many RNA populations are inefficiently capped at steady state (∼30 to 50%), and eIF4E overexpression increased this to ∼60 to 100%, depending on the RNA; 2) eIF4E physically associates with noncoding RNAs in the nucleus; and 3) approximately half of eIF4E-capping targets identified are noncoding RNAs. eIF4E's association with noncoding RNAs strongly positions it to act beyond translation. Coding and noncoding capping targets have activities that influence survival, cell morphology, and cell-to-cell interaction. Given that RNA export and translation machineries typically utilize capped RNA substrates, capping regulation provides means to titrate the protein-coding capacity of the transcriptome and, for noncoding RNAs, to regulate their activities. We also discovered a cap sensitivity element (CapSE) which conferred eIF4E-dependent capping sensitivity. Finally, we observed elevated capping for specific RNAs in high-eIF4E leukemia specimens, supporting a role for cap dysregulation in malignancy. In all, levels of capping RNAs can be regulated by eIF4E., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)- Published
- 2020
- Full Text
- View/download PDF
39. Selective dysregulation of ROCK2 activity promotes aberrant transcriptional networks in ABC diffuse large B-cell lymphoma.
- Author
-
Ricker E, Verma A, Marullo R, Gupta S, Ye C, Pannellini T, Manni M, Tam W, Inghirami G, Elemento O, Cerchietti L, and Pernis AB
- Subjects
- Cell Line, Tumor, Humans, Interferon Regulatory Factors genetics, Lymphoma, Large B-Cell, Diffuse metabolism, Lymphoma, Large B-Cell, Diffuse pathology, Phosphorylation, Proto-Oncogene Proteins c-myc metabolism, Gene Regulatory Networks, Lymphoma, Large B-Cell, Diffuse genetics, Transcription, Genetic, rho-Associated Kinases metabolism
- Abstract
Activated B-cell-like diffuse large B-cell lymphoma (ABC-DLBCL) is an aggressive subtype of lymphoma usually associated with inferior outcomes. ABC-DLBCL exhibits plasmablastic features and is characterized by aberrancies in the molecular networks controlled by IRF4. The signaling pathways that are dysregulated in ABC-DLBCL are, however, not fully understood. ROCK2 is a serine-threonine kinase whose role in lymphomagenesis is unknown. Here we show that ROCK2 activity is constitutively dysregulated in ABC-DLBCL but not in GCB-DLBCL and BL. We furthermore show that ROCK2 phosphorylates IRF4 and that the ROCK2-mediated phosphorylation of IRF4 modulates its ability to regulate a subset of target genes. In addition to its effects on IRF4, ROCK2 also controls the expression of MYC in ABC-DLBCL by regulating MYC protein levels. ROCK inhibition furthermore selectively decreases the proliferation and survival of ABC-DLBCL in vitro and inhibits ABC-DLBCL growth in xenograft models. Thus, dysregulated ROCK2 activity contributes to the aberrant molecular program of ABC-DLBCL via its dual ability to modulate both IRF4- and MYC-controlled gene networks and ROCK inhibition could represent an attractive therapeutic target for the treatment of ABC-DLBCL.
- Published
- 2020
- Full Text
- View/download PDF
40. Limits in the detection of m 6 A changes using MeRIP/m 6 A-seq.
- Author
-
McIntyre ABR, Gokhale NS, Cerchietti L, Jaffrey SR, Horner SM, and Mason CE
- Subjects
- Algorithms, Base Sequence, Humans, Immunoprecipitation, Methylation, RNA genetics, RNA, Messenger genetics, Sequence Analysis, RNA, Software, Adenosine genetics, RNA isolation & purification, RNA, Messenger isolation & purification
- Abstract
Many cellular mRNAs contain the modified base m
6 A, and recent studies have suggested that various stimuli can lead to changes in m6 A. The most common method to map m6 A and to predict changes in m6 A between conditions is methylated RNA immunoprecipitation sequencing (MeRIP-seq), through which methylated regions are detected as peaks in transcript coverage from immunoprecipitated RNA relative to input RNA. Here, we generated replicate controls and reanalyzed published MeRIP-seq data to estimate reproducibility across experiments. We found that m6 A peak overlap in mRNAs varies from ~30 to 60% between studies, even in the same cell type. We then assessed statistical methods to detect changes in m6 A peaks as distinct from changes in gene expression. However, from these published data sets, we detected few changes under most conditions and were unable to detect consistent changes across studies of similar stimuli. Overall, our work identifies limits to MeRIP-seq reproducibility in the detection both of peaks and of peak changes and proposes improved approaches for analysis of peak changes.- Published
- 2020
- Full Text
- View/download PDF
41. Inhibition of EZH2 Catalytic Activity Selectively Targets a Metastatic Subpopulation in Triple-Negative Breast Cancer.
- Author
-
Yomtoubian S, Lee SB, Verma A, Izzo F, Markowitz G, Choi H, Cerchietti L, Vahdat L, Brown KA, Andreopoulou E, Elemento O, Chang J, Inghirami G, Gao D, Ryu S, and Mittal V
- Subjects
- Animals, Base Sequence, Cell Compartmentation, Cell Differentiation, Cell Line, Tumor, Cell Proliferation, Disease Progression, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Epigenesis, Genetic, Female, GATA3 Transcription Factor metabolism, Humans, Mice, Inbred BALB C, Mice, SCID, Mutant Proteins metabolism, Neoplasm Invasiveness, Neoplasm Metastasis, Octamer Transcription Factor-3 metabolism, Phenotype, SOXB1 Transcription Factors metabolism, Triple Negative Breast Neoplasms genetics, Biocatalysis, Enhancer of Zeste Homolog 2 Protein metabolism, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology
- Abstract
Epigenetic changes are increasingly being appreciated as key events in breast cancer progression. However, breast cancer subtype-specific epigenetic regulation remains poorly investigated. Here we report that EZH2 is a leading candidate of epigenetic modulators associated with the TNBC subtype and that it predicts poor overall survival in TNBC patients. We demonstrate that specific pharmacological or genetic inhibition of EZH2 catalytic activity impairs distant metastasis. We further define a specific EZH2
high population with enhanced invasion, mammosphere formation, and metastatic potential that exhibits marked sensitivity to EZH2 inhibition. Mechanistically, EZH2 inhibition differentiates EZH2high basal cells to a luminal-like phenotype by derepressing GATA3 and renders them sensitive to endocrine therapy. Furthermore, dissection of human TNBC heterogeneity shows that EZH2high basal-like 1 and mesenchymal subtypes have exquisite sensitivity to EZH2 inhibition compared with the EZH2low luminal androgen receptor subtype. These preclinical findings provide a rationale for clinical development of EZH2 as a targeted therapy against TNBC metastasis., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
42. Application of total X-Ray fluorescence to gunshot residue determination.
- Author
-
Sarapura P, Gonzalez MF, Gonzalez F, Morzan E, Cerchietti L, and Custo G
- Abstract
Currently the great majority of the criminal acts have involved the use of firearms, for these reasons the evidences generate from these are one of the fundamental pillars of a forensic investigation. The firearm leaves evidence known as gunshot residue (GSR), which is principally composed of burnt and unburnt particles from the detonation, as well as fragments of the bullet, cartridge case, and the firearm. Gunshot residue (GSR) is produced when a firearm is discharged and large quantities of it can be transferred to an individual who has fired. SEM-EDX is the common technique used in the forensic laboratories, the analysis consists in detecting the particles and its elements. In this work we propose the use of X-ray Spectrometry by Total Reflection (TXRF) for the analysis of metals present in related samples in ballistic cases. The analysis was focused in the relationship of three elements present in GSR. A series of experiments with different persons firing gun of 9 mm was performed in a shooting range. Analytical XRFS signals corresponding to K line of Copper and L lines Barium and Lead were employed as the best discriminating variables. Machine Learning techniques, such as discriminant analysis, supported vector machines and partial least squares - discriminant analysis, enable the correct classification of all samples analyzed. A hundred samples were analyzed so far, this method has demonstrated a very high classification performance for detecting gunpowder residues in the skin., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
43. Non-oncogene Addiction to SIRT3 Plays a Critical Role in Lymphomagenesis.
- Author
-
Li M, Chiang YL, Lyssiotis CA, Teater MR, Hong JY, Shen H, Wang L, Hu J, Jing H, Chen Z, Jain N, Duy C, Mistry SJ, Cerchietti L, Cross JR, Cantley LC, Green MR, Lin H, and Melnick AM
- Subjects
- Acetyl Coenzyme A metabolism, Animals, Antineoplastic Agents pharmacology, Autophagic Cell Death drug effects, Citric Acid Cycle drug effects, Female, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Glutamine metabolism, HEK293 Cells, Histone Deacetylase Inhibitors pharmacology, Humans, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, MCF-7 Cells, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Molecular Targeted Therapy, Signal Transduction, Sirtuin 3 antagonists & inhibitors, Sirtuin 3 deficiency, Sirtuin 3 genetics, Xenograft Model Antitumor Assays, Cell Proliferation drug effects, Energy Metabolism drug effects, Lymphoma, Large B-Cell, Diffuse enzymology, Sirtuin 3 metabolism
- Abstract
Diffuse large B cell lymphomas (DLBCLs) are genetically heterogeneous and highly proliferative neoplasms derived from germinal center (GC) B cells. Here, we show that DLBCLs are dependent on mitochondrial lysine deacetylase SIRT3 for proliferation, survival, self-renewal, and tumor growth in vivo regardless of disease subtype and genetics. SIRT3 knockout attenuated B cell lymphomagenesis in VavP-Bcl2 mice without affecting normal GC formation. Mechanistically, SIRT3 depletion impaired glutamine flux to the TCA cycle via glutamate dehydrogenase and reduction in acetyl-CoA pools, which in turn induce autophagy and cell death. We developed a mitochondrial-targeted class I sirtuin inhibitor, YC8-02, which phenocopied the effects of SIRT3 depletion and killed DLBCL cells. SIRT3 is thus a metabolic non-oncogene addiction and therapeutic target for DLBCLs., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
44. BCL6 Evolved to Enable Stress Tolerance in Vertebrates and Is Broadly Required by Cancer Cells to Adapt to Stress.
- Author
-
Fernando TM, Marullo R, Pera Gresely B, Phillip JM, Yang SN, Lundell-Smith G, Torregroza I, Ahn H, Evans T, Győrffy B, Privé GG, Hirano M, Melnick AM, and Cerchietti L
- Subjects
- Animals, Apoptosis physiology, B-Lymphocytes cytology, B-Lymphocytes physiology, Cell Proliferation physiology, Cells, Cultured, Female, Germinal Center cytology, Germinal Center physiology, Heat Shock Transcription Factors biosynthesis, Heat Shock Transcription Factors genetics, Heat Shock Transcription Factors metabolism, Heat-Shock Response, Heterografts, Humans, Male, Mice, Mice, Knockout, Mice, SCID, Neoplasms enzymology, Neoplasms pathology, Proto-Oncogene Proteins c-bcl-6 genetics, Adaptation, Physiological physiology, Neoplasms drug therapy, Proto-Oncogene Proteins c-bcl-6 metabolism, Stress, Physiological physiology
- Abstract
Several lines of evidence link the canonical oncogene BCL6 to stress response. Here we demonstrate that BCL6 evolved in vertebrates as a component of the HSF1-driven stress response, which has been co-opted by the immune system to support germinal center formation and may have been decisive in the convergent evolution of humoral immunity in jawless and jawed vertebrates. We find that the highly conserved BTB corepressor binding site of BCL6 mediates stress adaptation across vertebrates. We demonstrate that pan-cancer cells hijack this stress tolerance mechanism to aberrantly express BCL6. Targeting the BCL6 BTB domain in cancer cells induces apoptosis and increases susceptibility to repeated doses of cytotoxic therapy. The chemosensitization effect upon BCL6 BTB inhibition is dependent on the derepression of TOX , implicating modulation of DNA repair as a downstream mechanism. Collectively, these data suggest a form of adaptive nononcogene addiction rooted in the natural selection of BCL6 during vertebrate evolution. SIGNIFICANCE: We demonstrate that HSF1 drives BCL6 expression to enable stress tolerance in vertebrates. We identify an HSF1-BCL6-TOX stress axis that is required by cancer cells to tolerate exposure to cytotoxic agents and points toward BCL6-targeted therapy as a way to more effectively kill a wide variety of solid tumors. This article is highlighted in the In This Issue feature, p. 565 ., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
45. Thyroid hormones induce doxorubicin chemosensitivity through enzymes involved in chemotherapy metabolism in lymphoma T cells.
- Author
-
Díaz Flaqué MC, Cayrol MF, Sterle HA, Del Rosario Aschero M, Díaz Albuja JA, Isse B, Farías RN, Cerchietti L, Rosemblit C, and Cremaschi GA
- Abstract
Thyroid hormones (THs) - 3,3',5-triiodo-L-thyronine (T3) and L-thyroxine (T4) - are important regulators of the metabolism and physiology of most normal tissues. Cytochrome P450 family 3A members are drug metabolizing enzymes involved in the activation and detoxification of several drugs. CYP3A4 is the major enzyme involved in the metabolism of chemotherapeutic drugs. In this work, we demonstrate that THs induce a significant increase in CYP3A4 mRNA levels, protein expression and metabolic activity through the membrane receptor integrin αvβ3 and the activation of signalling pathways through Stat1 and NF-κB. We reasoned that TH-induced CYP3A4 modulation may act as an important regulator in the metabolism of doxorubicin (Doxo). Experiments in vitro demonstrated that in CYP3A4-knocked down cells, no TH-mediated chemosensitivity to Doxo was observed. We also found that THs modulate these functions by activating the membrane receptor integrin αvβ3. In addition, we showed that the thyroid status can modulate CYP450 mRNA levels in tumor and liver tissues, and the tumor volume in response to chemotherapy in vivo . In fact, Doxo treatment in hypothyroid mice was associated with lower tumors, displaying lower levels of CYP enzymes, than euthyroid mice. However, higher mRNA levels of CYP enzymes were found in livers from Doxo treated hypothyroid mice respect to control. These results present a new mechanism by which TH could modulate chemotherapy response. These findings highlight the importance of evaluating thyroid status in patients during application of T-cell lymphoma therapeutic regimens., Competing Interests: CONFLICTS OF INTEREST The authors declare that there are no conflicts of interest that could be perceived as prejudicing the impartiality of the research reported.
- Published
- 2019
- Full Text
- View/download PDF
46. Five-year follow-up of lenalidomide plus rituximab as initial treatment of mantle cell lymphoma.
- Author
-
Ruan J, Martin P, Christos P, Cerchietti L, Tam W, Shah B, Schuster SJ, Rodriguez A, Hyman D, Calvo-Vidal MN, Smith SM, Svoboda J, Furman RR, Coleman M, and Leonard JP
- Subjects
- Adult, Aged, Aged, 80 and over, Angiogenesis Inhibitors adverse effects, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Female, Follow-Up Studies, Humans, Lenalidomide adverse effects, Male, Middle Aged, Neutropenia chemically induced, Rituximab adverse effects, Survival Analysis, Thrombocytopenia chemically induced, Treatment Outcome, Angiogenesis Inhibitors therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Lenalidomide therapeutic use, Lymphoma, Mantle-Cell drug therapy, Rituximab therapeutic use
- Abstract
We report 5-year follow-up of a multicenter phase 2 study of lenalidomide plus rituximab (LR) as initial treatment of mantle cell lymphoma (MCL). The regimen includes induction and maintenance with the LR doublet. Treatment was continuous until progression, with optional discontinuation after 3 years. The median age of the 38 participants was 65 years, with MCL international prognostic index scores balanced among low, intermediate, and high risk (34%, 34%, and 32%, respectively). Twenty-seven (75%) of the 36 evaluable patients completed ≥3 years of study treatment. At a median follow-up of 64 months (range, 21-78), the 3-year progression-free survival (PFS) and overall survival (OS) were 80% and 90%, respectively, with 5-year estimated PFS and OS of 64% and 77%, respectively. During maintenance, hematologic adverse events (AEs) included asymptomatic grade 3 or 4 cytopenias (42% neutropenia, 5% thrombocytopenia, 3% anemia) and mostly grade 1 or 2 infections managed in the outpatient setting (45% upper respiratory infection, 21% urinary tract infection, 13% sinusitis, 11% cellulitis, 8% pneumonia). Nonhematologic AEs, such as constitutional and inflammatory symptoms, occurred at reduced frequency and intensity compared with induction. A peripheral blood minimal residual disease (MRD) assay (clonoSEQ) showed MRD-negative complete remission in 8 of 10 subjects who had completed ≥3 years of treatment and with available samples for analysis. With longer follow-up, LR continues to demonstrate durable responses and manageable safety as initial induction and maintenance therapy for MCL (ClinicalTrials.gov NCT01472562)., (© 2018 by The American Society of Hematology.)
- Published
- 2018
- Full Text
- View/download PDF
47. Germline Lysine-Specific Demethylase 1 ( LSD1/KDM1A ) Mutations Confer Susceptibility to Multiple Myeloma.
- Author
-
Wei X, Calvo-Vidal MN, Chen S, Wu G, Revuelta MV, Sun J, Zhang J, Walsh MF, Nichols KE, Joseph V, Snyder C, Vachon CM, McKay JD, Wang SP, Jayabalan DS, Jacobs LM, Becirovic D, Waller RG, Artomov M, Viale A, Patel J, Phillip J, Chen-Kiang S, Curtin K, Salama M, Atanackovic D, Niesvizky R, Landgren O, Slager SL, Godley LA, Churpek J, Garber JE, Anderson KC, Daly MJ, Roeder RG, Dumontet C, Lynch HT, Mullighan CG, Camp NJ, Offit K, Klein RJ, Yu H, Cerchietti L, and Lipkin SM
- Subjects
- Animals, Cell Line, Tumor, Cyclin D2 biosynthesis, Genes, Tumor Suppressor, Germ Cells pathology, Histone Demethylases antagonists & inhibitors, Histones metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Mutation, Missense genetics, Paraproteins analysis, Plasma Cells pathology, RNA Interference, RNA, Small Interfering genetics, Genetic Predisposition to Disease genetics, Histone Demethylases genetics, Monoclonal Gammopathy of Undetermined Significance genetics, Multiple Myeloma genetics
- Abstract
Given the frequent and largely incurable occurrence of multiple myeloma, identification of germline genetic mutations that predispose cells to multiple myeloma may provide insight into disease etiology and the developmental mechanisms of its cell of origin, the plasma cell (PC). Here, we identified familial and early-onset multiple myeloma kindreds with truncating mutations in lysine-specific demethylase 1 (LSD1/KDM1A), an epigenetic transcriptional repressor that primarily demethylates histone H3 on lysine 4 and regulates hematopoietic stem cell self-renewal. In addition, we found higher rates of germline truncating and predicted deleterious missense KDM1A mutations in patients with multiple myeloma unselected for family history compared with controls. Both monoclonal gammopathy of undetermined significance (MGUS) and multiple myeloma cells have significantly lower KDM1A transcript levels compared with normal PCs. Transcriptome analysis of multiple myeloma cells from KDM1A mutation carriers shows enrichment of pathways and MYC target genes previously associated with myeloma pathogenesis. In mice, antigen challenge followed by pharmacologic inhibition of KDM1A promoted PC expansion, enhanced secondary immune response, elicited appearance of serum paraprotein, and mediated upregulation of MYC transcriptional targets. These changes are consistent with the development of MGUS. Collectively, our findings show that KDM1A is the first autosomal-dominant multiple myeloma germline predisposition gene providing new insights into its mechanistic roles as a tumor suppressor during post-germinal center B-cell differentiation. Significance: KDM1A is the first germline autosomal dominant predisposition gene identified in multiple myeloma and provides new insights into multiple myeloma etiology and the mechanistic role of KDM1A as a tumor suppressor during post-germinal center B-cell differentiation. Cancer Res; 78(10); 2747-59. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
48. Metabolomic Profiling Reveals Cellular Reprogramming of B-Cell Lymphoma by a Lysine Deacetylase Inhibitor through the Choline Pathway.
- Author
-
Pera B, Krumsiek J, Assouline SE, Marullo R, Patel J, Phillip JM, Román L, Mann KK, and Cerchietti L
- Subjects
- Animals, Cell Line, Tumor, Choline Kinase metabolism, Humans, Hydroxamic Acids pharmacology, Indoles pharmacology, Male, Metabolome drug effects, Mice, Morpholines pharmacology, Panobinostat, Pyrimidines pharmacology, Xenograft Model Antitumor Assays, Cellular Reprogramming drug effects, Choline metabolism, Histone Deacetylase Inhibitors pharmacology, Lymphoma, Large B-Cell, Diffuse metabolism, Lymphoma, Large B-Cell, Diffuse pathology, Lysine metabolism, Metabolomics methods, Signal Transduction drug effects
- Abstract
Despite the proven clinical antineoplastic activity of histone deacetylase inhibitors (HDACI), their effect has been reported to be lower than expected in B-cell lymphomas. Traditionally considered as "epigenetic drugs", HDACI modify the acetylation status of an extensive proteome, acting as general lysine deacetylase inhibitors (KDACI), and thus potentially impacting various branches of cellular metabolism. Here, we demonstrate through metabolomic profiling of patient plasma and cell lines that the KDACI panobinostat alters lipid metabolism and downstream survival signaling in diffuse large B-cell lymphomas (DLBCL). Specifically, panobinostat induces metabolic adaptations resulting in newly acquired dependency on the choline pathway and activation of PI3K signaling. This metabolic reprogramming decreased the antineoplastic effect of panobinostat. Conversely, inhibition of these metabolic adaptations resulted in superior anti-lymphoma effect as demonstrated by the combination of panobinostat with a choline pathway inhibitor. In conclusion, our study demonstrates the power of metabolomics in identifying unknown effects of KDACI, and emphasizes the need for a better understanding of these drugs in order to achieve successful clinical implementation., (Copyright © 2018 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
49. The eIF4E inhibitor ribavirin as a potential antilymphoma therapeutic: early clinical data<sup/>.
- Author
-
Rutherford SC, Stewart EN, Chen Z, Chadburn A, Wehrli NE, van Besien K, Martin P, Furman RR, Leonard JP, and Cerchietti L
- Subjects
- Antiviral Agents therapeutic use, Eukaryotic Initiation Factor-4E metabolism, Humans, Lymphoma metabolism, Lymphoma virology, Remission Induction, Respiratory Syncytial Virus Infections drug therapy, Respiratory Syncytial Virus Infections metabolism, Respiratory Syncytial Virus Infections virology, Respiratory Syncytial Viruses drug effects, Retrospective Studies, Eukaryotic Initiation Factor-4E antagonists & inhibitors, Lymphoma drug therapy, Ribavirin therapeutic use
- Published
- 2018
- Full Text
- View/download PDF
50. Award Winner in the Young Investigator Category, 2017 Society for Biomaterials Annual Meeting and Exposition, Minneapolis, MN, April 05-08, 2017: Lymph node stiffness-mimicking hydrogels regulate human B-cell lymphoma growth and cell surface receptor expression in a molecular subtype-specific manner.
- Author
-
Apoorva FNU, Tian YF, Pierpont TM, Bassen DM, Cerchietti L, Butcher JT, Weiss RS, and Singh A
- Subjects
- Animals, Cell Line, Tumor, Humans, Mice, Biomimetic Materials chemistry, Gene Expression Regulation, Neoplastic, Hydrogels chemistry, Integrins biosynthesis, Lymph Nodes chemistry, Lymph Nodes metabolism, Lymph Nodes pathology, Lymphoma, Large B-Cell, Diffuse metabolism, Lymphoma, Large B-Cell, Diffuse pathology, Neoplasm Proteins biosynthesis, Signal Transduction
- Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin's lymphoma, with multiple molecular subtypes. The activated B-cell-like DLBCL subtype accounts for roughly one-third of all the cases and has an inferior prognosis. There is a need to develop better class of therapeutics that could target molecular pathways in resistant DLBCLs; however, this requires DLBCLs to be studied in representative tumor microenvironments. The pathogenesis and progression of lymphoma has been mostly studied from the point of view of genetic alterations and intracellular pathway dysregulation. By comparison, the importance of lymphoma microenvironment in which these malignant cells arise and reside has not been studied in as much detail. We have recently elucidated the role of integrin signaling in lymphomas and demonstrated that inhibition of integrin-ligand interactions abrogated the proliferation of malignant cells in vitro and in patient-derived xenograft. Here we demonstrate the role of lymph node tissue stiffness on DLBCL in a B-cell molecular subtype specific manner. We engineered tunable bioartificial hydrogels that mimicked the stiffness of healthy and neoplastic lymph nodes of a transgenic mouse model and primary human lymphoma tumors. Our results demonstrate that molecularly diverse DLBCLs grow differentially in soft and high stiffness microenvironments, which further modulates the integrin and B-cell receptor expression level as well as response to therapeutics. We anticipate that our findings will be broadly useful to study lymphoma biology and discover new class of therapeutics that target B-cell tumors in physical environments. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1833-1844, 2017., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.