180 results on '"Ruella, M."'
Search Results
2. Combination of everolimus and itacitinib in patients with Hodgkin lymphoma relapsed/refractory to brentuximab vedotin (BV) and checkpoint inhibitors (CPI)
- Author
-
Svoboda, J., primary, Landsburg, D. L., additional, Nasta, S. D., additional, Barta, S. K., additional, Chong, E. A., additional, Carter, J., additional, Delp, G., additional, Duca, A., additional, Ballard, H. J., additional, Emanuel, S. A., additional, McWilliams, T., additional, Hwang, W., additional, Hughes, M. E., additional, Ruella, M., additional, and Schuster, S. J., additional
- Published
- 2023
- Full Text
- View/download PDF
3. FOURTH GENERATION HUCART19‐IL18 PRODUCES DURABLE RESPONSES IN LYMPHOMA PATIENTS PREVIOUSLY RELAPSED/REFRACTORY TO ANTI‐CD19 CAR T‐CELL THERAPY
- Author
-
Svoboda, J., primary, Landsburg, D. L., additional, Chong, E. A., additional, Barta, S. K., additional, Nasta, S. D., additional, Ruella, M., additional, Hexner, E. O., additional, Marshall, A., additional, Leskowitz, R., additional, Four, M., additional, Shea, J., additional, Cervini, A., additional, Davis, M. M., additional, Please, G., additional, Hasenmayer, D., additional, Amortegui, J. R., additional, Hwang, W., additional, Frey, N., additional, Siegel, D. L., additional, Fraietta, J., additional, Levine, B., additional, Porter, D. L., additional, Schuster, S. J., additional, and June, C. H., additional
- Published
- 2023
- Full Text
- View/download PDF
4. Immunotherapy: CLINICAL DEVELOPMENT OF A NOVEL CD5 KNOCKED OUT ANTI-CD5 CAR T CELL PRODUCT FOR T CELL LYMPHOMA
- Author
-
Patel, R., primary, Ghilardi, G., additional, Porazzi, P., additional, Yang, S., additional, Qian, D., additional, Zhang, Y., additional, Pajarillo, R., additional, Schuster, S., additional, Barta, S., additional, Nimmagadda, A., additional, Snook, A., additional, Siciliano, N., additional, and Ruella, M., additional
- Published
- 2023
- Full Text
- View/download PDF
5. Immunotherapy: THE BTLA-HVEM AXIS IS A CRUCIAL IMMUNE CHECKPOINT OF T CELL IMMUNOTHERAPIES FOR CANCER
- Author
-
Guruprasad, P., primary, Carturan, A., additional, Zhang, Y., additional, Ghilardi, G., additional, Kumashie, K., additional, Kim, K., additional, Lee, Y., additional, Kim, J., additional, Lee, J., additional, Shestov, M., additional, Pajarillo, R., additional, Harris, J., additional, Lee, Y.G., additional, Wang, M., additional, Joshi, A., additional, Ballard, H., additional, Cohen, I., additional, Ugwuanyi, O., additional, Hong, A., additional, Paruzzo, L., additional, Patel, R., additional, Shestova, O., additional, Porazzi, P., additional, Svoboda, J., additional, and Ruella, M., additional
- Published
- 2023
- Full Text
- View/download PDF
6. Strategy to prevent epitope masking in CAR.CD19+ B-cell leukemia blasts
- Author
-
Quintarelli, C, Guercio, M, Manni, S, Boffa, I, Sinibaldi, M, DI Cecca, S, Caruso, S, Abbaszadeh, Z, Camera, A, Cembrola, B, Ciccone, R, Orfao, A, Martin-Martin, L, Gutierrez-Herrero, S, Herrero-Garcia, M, Cazzaniga, G, Nunes, V, Songia, S, Marcatili, P, Marin, F, Ruella, M, Bertaina, V, Vinti, L, Del Bufalo, F, Algeri, M, Merli, P, De Angelis, B, Locatelli, F, Quintarelli C., Guercio M., Manni S., Boffa I., Sinibaldi M., DI Cecca S., Caruso S., Abbaszadeh Z., Camera A., Cembrola B., Ciccone R., Orfao A., Martin-Martin L., Gutierrez-Herrero S., Herrero-Garcia M., Cazzaniga G., Nunes V., Songia S., Marcatili P., Marin F. I., Ruella M., Bertaina V., Vinti L., Del Bufalo F., Algeri M., Merli P., De Angelis B., Locatelli F., Quintarelli, C, Guercio, M, Manni, S, Boffa, I, Sinibaldi, M, DI Cecca, S, Caruso, S, Abbaszadeh, Z, Camera, A, Cembrola, B, Ciccone, R, Orfao, A, Martin-Martin, L, Gutierrez-Herrero, S, Herrero-Garcia, M, Cazzaniga, G, Nunes, V, Songia, S, Marcatili, P, Marin, F, Ruella, M, Bertaina, V, Vinti, L, Del Bufalo, F, Algeri, M, Merli, P, De Angelis, B, Locatelli, F, Quintarelli C., Guercio M., Manni S., Boffa I., Sinibaldi M., DI Cecca S., Caruso S., Abbaszadeh Z., Camera A., Cembrola B., Ciccone R., Orfao A., Martin-Martin L., Gutierrez-Herrero S., Herrero-Garcia M., Cazzaniga G., Nunes V., Songia S., Marcatili P., Marin F. I., Ruella M., Bertaina V., Vinti L., Del Bufalo F., Algeri M., Merli P., De Angelis B., and Locatelli F.
- Abstract
Chimeric antigen receptor T-cells (CAR T-cells) for the treatment of relapsing/refractory B-cell precursor acute lymphoblastic leukemia have led to exciting clinical results. However, CAR T-cell approaches revealed a potential risk of CD19-/CAR+ leukemic relapse due to inadvertent transduction of leukemia cells. BackgroundMethods We evaluated the impact of a high percentage of leukemia blast contamination in patient-derived starting material (SM) on CAR T-cell drug product (DP) manufacturing. In vitro as well as in vivo models were employed to identify characteristics of the construct associated with better profile of safety in case of inadvertent B-cell leukemia transduction during CAR T-cell manufacturing. Results The presence of large amounts of CD19+ cells in SM did not affect the transduction level of DPs, as well as the CAR T-cell rate of expansion at the end of standard production of 14 days. DPs were deeply characterized by flow cytometry and molecular biology for Ig-rearrangements, showing that the level of B-cell contamination in DPs did not correlate with the percentage of CD19+ cells in SM, in the studied patient cohort. Moreover, we investigated whether CAR design may affect the control of CAR+ leukemia cells. We provided evidences that CAR.CD19 short linker (SL) prevents complete epitope masking in CD19+CAR+ leukemia cells and we demonstrated in vitro and in vivo that CD19 +CAR(SL)+leukemic cells are killed by CAR.CD19 T-cells. Conclusions Taken together, these data suggest that a VL-VH SL may result in a safe CAR-T product, even when manufacturing starts from biological materials characterized by heavy contamination of leukemia blasts.
- Published
- 2021
7. Kinase inhibitor ibrutinib to prevent cytokine-release syndrome after anti-CD19 chimeric antigen receptor T cells for B-cell neoplasms
- Author
-
Ruella, M, Kenderian, S S, Shestova, O, Klichinsky, M, Melenhorst, J J, Wasik, M A, Lacey, S F, June, C H, and Gill, S
- Published
- 2017
- Full Text
- View/download PDF
8. CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia
- Author
-
Kenderian, S S, Ruella, M, Shestova, O, Klichinsky, M, Aikawa, V, Morrissette, J J D, Scholler, J, Song, D, Porter, D L, Carroll, M, June, C H, and Gill, S
- Published
- 2015
- Full Text
- View/download PDF
9. Cervicodorsal extramedullary anaplastic ependymoma with intracranial metastasis
- Author
-
Marco Del Pont, F., primary, Villamil, F., additional, Lorefice, E., additional, Caffaratti, G., additional, Centeno, T.R., additional, Ruella, M., additional, and Mormandi, R., additional
- Published
- 2021
- Full Text
- View/download PDF
10. Terson syndrome caused by intraventricular hemorrhage associated with arteriovenous malformation: Case report
- Author
-
Villamil, F., primary, Ruella, M., additional, Giovannini, S.J.M., additional, Ries Centeno, T., additional, Marco Del Pont, F., additional, Millar, P., additional, and Alcorta, S.C., additional
- Published
- 2021
- Full Text
- View/download PDF
11. A serious challenge in fighting the COVID 19 pandemic: SARS CoV 2 vaccines in oncologic patients
- Author
-
Ruella, M., primary and Ghilardi, G., additional
- Published
- 2021
- Full Text
- View/download PDF
12. Bone marrow-derived cell mobilization by G-CSF to enhance osseointegration of bone substitute in high tibial osteotomy
- Author
-
Marmotti, A., Castoldi, F., Rossi, R., Marenco, S., Risso, A., Ruella, M., Tron, A., Borrè, A., Blonna, D., and Tarella, C.
- Published
- 2013
- Full Text
- View/download PDF
13. Immunogenicity of CAR T cells in cancer therapy
- Author
-
Shah, N.N., Wayne, A.S., Turtle, C.J., Abou-el-Enein, M., Ahmed, N., Ruella, M., Wagner, D.L., Savoldo, B., Hegde, M., Hamieh, M., Fritsche, E., and Pulsipher, M.A.
- Abstract
Patient-derived T cells genetically reprogrammed to express CD19-specific chimeric antigen receptors (CARs) have shown remarkable clinical responses and are commercially available for the treatment of patients with certain advanced-stage B cell malignancies. Nonetheless, several trials have revealed pre-existing and/or treatment-induced immune responses to the mouse-derived single-chain variable fragments included in these constructs. These responses might have contributed to both treatment failure and the limited success of redosing strategies observed in some patients. Data from early phase clinical trials suggest that CAR T cells are also associated with immunogenicity-related events in patients with solid tumours. Generally, the clinical implications of anti-CAR immune responses are poorly understood and highly variable between different CAR constructs and malignancies. These observations highlight an urgent need to uncover the mechanisms of immunogenicity in patients receiving CAR T cells and develop validated assays to enable clinical detection. In this Review, we describe the current clinical evidence of anti-CAR immune responses and discuss how new CAR T cell technologies might impact the risk of immunogenicity. We then suggest ways to reduce the risks of anti-CAR immune responses to CAR T cell products that are advancing towards the clinic. Finally, we summarize measures that investigators could consider in order to systematically monitor and better comprehend the possible effects of immunogenicity during trials involving CAR T cells as well as in routine clinical practice.
- Published
- 2021
- Full Text
- View/download PDF
14. Strategy to prevent epitope masking in CAR.CD19+ B-cell leukemia blasts
- Author
-
Quintarelli, C., Guercio, M., Manni, S., Boffa, I., Sinibaldi, M., DI Cecca, S., Caruso, S., Abbaszadeh, Z., Camera, A., Cembrola, B., Ciccone, R., Orfao, A., Martin-Martin, L., Gutierrez-Herrero, S., Herrero-Garcia, M., Cazzaniga, G., Nunes, V., Songia, S., Marcatili, P., Marin, F. I., Ruella, M., Bertaina, V., Vinti, L., Del Bufalo, F., Algeri, M., Merli, P., De Angelis, B., Locatelli, Franco, Locatelli F. (ORCID:0000-0002-7976-3654), Quintarelli, C., Guercio, M., Manni, S., Boffa, I., Sinibaldi, M., DI Cecca, S., Caruso, S., Abbaszadeh, Z., Camera, A., Cembrola, B., Ciccone, R., Orfao, A., Martin-Martin, L., Gutierrez-Herrero, S., Herrero-Garcia, M., Cazzaniga, G., Nunes, V., Songia, S., Marcatili, P., Marin, F. I., Ruella, M., Bertaina, V., Vinti, L., Del Bufalo, F., Algeri, M., Merli, P., De Angelis, B., Locatelli, Franco, and Locatelli F. (ORCID:0000-0002-7976-3654)
- Abstract
Chimeric antigen receptor T-cells (CAR T-cells) for the treatment of relapsing/refractory B-cell precursor acute lymphoblastic leukemia have led to exciting clinical results. However, CAR T-cell approaches revealed a potential risk of CD19-/CAR+ leukemic relapse due to inadvertent transduction of leukemia cells. BackgroundMethods We evaluated the impact of a high percentage of leukemia blast contamination in patient-derived starting material (SM) on CAR T-cell drug product (DP) manufacturing. In vitro as well as in vivo models were employed to identify characteristics of the construct associated with better profile of safety in case of inadvertent B-cell leukemia transduction during CAR T-cell manufacturing. Results The presence of large amounts of CD19+ cells in SM did not affect the transduction level of DPs, as well as the CAR T-cell rate of expansion at the end of standard production of 14 days. DPs were deeply characterized by flow cytometry and molecular biology for Ig-rearrangements, showing that the level of B-cell contamination in DPs did not correlate with the percentage of CD19+ cells in SM, in the studied patient cohort. Moreover, we investigated whether CAR design may affect the control of CAR+ leukemia cells. We provided evidences that CAR.CD19 short linker (SL) prevents complete epitope masking in CD19+CAR+ leukemia cells and we demonstrated in vitro and in vivo that CD19 +CAR(SL)+leukemic cells are killed by CAR.CD19 T-cells. Conclusions Taken together, these data suggest that a VL-VH SL may result in a safe CAR-T product, even when manufacturing starts from biological materials characterized by heavy contamination of leukemia blasts.
- Published
- 2021
15. Comparative assessment of telomere length before and after hematopoietic SCT: role of grafted cells in determining post-transplant telomere status
- Author
-
Ruella, M, Rocci, A, Ricca, I, Carniti, C, Bodoni, C Labetti, Ladetto, M, Caracciolo, D, Boccadoro, M, Carlo-Stella, C, Corradini, P, and Tarella, C
- Published
- 2010
- Full Text
- View/download PDF
16. Haploidentical cellular therapy in elderly patients with acute myeloid leukemia: Description of its use in high risk patients
- Author
-
Cignetti, A., Ruella, M., Elia, A., Tassi, V., Redoglia, V., Gottardi, D., and Tarella, C.
- Published
- 2013
- Full Text
- View/download PDF
17. Perspectives in immunotherapy: meeting report from the Immunotherapy Bridge (29-30 November, 2017, Naples, Italy)
- Author
-
Ascierto, P.A. (Paolo Antonio), Brugarolas, J. (James), Buonaguro, L. (Luigi), Butterfield, L.H. (Lisa H.), Carbone, D. (David), Daniele, B. (Bruno), Ferris, R. (Robert), Fox, B.A. (Bernard A.), Galon, J. (Jerome), Gridelli, C. (Cesare), Kaufman, H.L. (Howard L.), Klebanoff, C.A. (Christopher A.), Melero, I. (Ignacio), Nathan, P. (Paul), Paulos, C.M. (Chrystal M.), Ruella, M. (Marco), Sullivan, R. (Ryan), Zarour, H. (Hassane), and Puzanov, I. (Igor)
- Subjects
Adoptive cell transfer combination therapy ,Immunotherapy ,Cancer vaccines ,Chekpoint inhibitors ,Biomarkers - Abstract
Immunotherapy represents the third important wave in the history of the systemic treatment of cancer after chemotherapy and targeted therapy and is now established as a potent and effective treatment option across several cancer types. The clinical success of anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4, first, and anti-programmed death (PD)-1/PD-ligand (L)1 agents in melanoma and other cancers a few years later, has encouraged increasing focus on the development of other immunotherapies (e.g. monoclonal antibodies with other immune targets, adoptive cell transfer, and vaccines), with over 3000 immuno-oncology trials ongoing, involving hundreds of research institutes across the globe. The potential use of these different immunotherapeutic options in various combinations with one another and with other treatment modalities is an area of particular promise. The third Immunotherapy Bridge meeting (29-30 November, 2017, Naples, Italy) focused on recent advances in immunotherapy across various cancer types and is summarised in this report.
- Published
- 2018
18. EVITA: PHASE I/II STUDY OF EVEROLIMUS PLUS ITACITINIB IN RELAPSED/REFRACTORY CLASSICAL HODGKIN LYMPHOMA
- Author
-
Svoboda, J., primary, Barta, S., additional, Nasta, S., additional, Landsburg, D., additional, Gerson, J., additional, Ruella, M., additional, Waite, T., additional, King, C., additional, Emanuel, S.A., additional, Ballard, H., additional, and Schuster, S., additional
- Published
- 2019
- Full Text
- View/download PDF
19. Outcomes with bendamustine lymphodepletion and brexucabtagene autoleucel for mantle cell lymphoma.
- Author
-
Chong, E. A., Gerson, J. N., Nasta, S. D., Landsburg, D. J., Barta, S. K., Svoboda, J., Weber, E., Ghilardi, G., Ruella, M., Porter, D. L., Frey, N. V., Chong, E. R., and Schuster, S. J.
- Subjects
MANTLE cell lymphoma ,CYTOKINE release syndrome - Abstract
14/16 (88%) patients developed CRS; 11/14 (79%) had grade 1-2 CRS and 3 (19%) had grade 3 or greater CRS. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded per ASTCT consensus criteria. 5 (31%) patients had ICANS; 2 (12.5%) had grade 1-2 ICANS, and 3 (19%) had grade 3 or greater ICANS. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
20. Factors influencing the occurrence of secondary malignancies in high-risk lymphoma patients following high-dose therapy and autograft
- Author
-
Tarella C, Passera R, Magni M, Rossi A, Benedetti F, Gueli A, Ruella M, Patti C, Parvis G, Gallamini A, Cortelazzo S, Corradini P, Zoli V, Mule A, Crocchiolo R, Bosa M, Barbui A, Di Nicola M, Caracciolo D, Sorio M, Gianni AM, Rambaldi A., CICERI , FABIO, Tarella, C, Passera, R, Magni, M, Rossi, A, Benedetti, F, Gueli, A, Ruella, M, Patti, C, Parvis, G, Ciceri, Fabio, Gallamini, A, Cortelazzo, S, Corradini, P, Zoli, V, Mule, A, Crocchiolo, R, Bosa, M, Barbui, A, Di Nicola, M, Caracciolo, D, Sorio, M, Gianni, Am, and Rambaldi, A.
- Published
- 2010
21. A recent update of 3 consecutive prospective trials with high-dose therapy and autograft, without or with rituximab, as primary treatment for advanced-stage follicular lymphoma shows a sizeable group of patients surviving in continuous complete remission
- Author
-
Tarella C, Ladetto M, Benedetti F, Vitolo U, Pulsoni A, Patti C, Callea V, Rambaldi A, Piccin A, Devizzi L, Musso M, Iannitto E, Spedini P, Liberati AM, Gallamini A, Rodeghiero F, Gini G, De Crescenzo A, Di Raimondo F, Levis A, Chisesi T, Petrone T, Scalabrini DR, Rossi G, Carella AM, Parvis G, Majolino I, Passera R, Ruella M, Pileri A, Gianni AM, Corradini P., CICERI , FABIO, Tarella, C, Ladetto, M, Benedetti, F, Vitolo, U, Pulsoni, A, Patti, C, Callea, V, Rambaldi, A, Piccin, A, Devizzi, L, Musso, M, Iannitto, E, Spedini, P, Liberati, Am, Ciceri, Fabio, Gallamini, A, Rodeghiero, F, Gini, G, De Crescenzo, A, Di Raimondo, F, Levis, A, Chisesi, T, Petrone, T, Scalabrini, Dr, Rossi, G, Carella, Am, Parvis, G, Majolino, I, Passera, R, Ruella, M, Pileri, A, Gianni, Am, and Corradini, P.
- Published
- 2010
22. Kinase inhibitor ibrutinib to prevent cytokine-release syndrome after anti-CD19 chimeric antigen receptor T cells for B-cell neoplasms
- Author
-
Ruella, M, primary, Kenderian, S S, additional, Shestova, O, additional, Klichinsky, M, additional, Melenhorst, J J, additional, Wasik, M A, additional, Lacey, S F, additional, June, C H, additional, and Gill, S, additional
- Published
- 2016
- Full Text
- View/download PDF
23. Pregnancy complications predict thrombotic events in young women with essential thrombocythemia
- Author
-
Randi, M. L., Bertozzi, I., Rumi, E., Elena, C., Finazzi, G., Vianelli, N., Polverelli, N., Ruggeri, M., Vannucchi, A. M., Antonioli, E., Lussana, F., Tieghi, A., Iurlo, A., Elli, E., Ruella, M., Fabris, F., Cazzola, M., and Barbui, T.
- Subjects
JAK2 V617F MUTATION ,Adult ,Adolescent ,BUDD-CHIARI-SYNDROME ,WORLD-HEALTH-ORGANIZATION ,MYELOPROLIFERATIVE DISORDERS ,POLYCYTHEMIA-VERA ,ORAL-CONTRACEPTIVES ,RISK, SURVIVAL ,Mutation, Missense ,Hemorrhage ,Comorbidity ,Young Adult ,Pregnancy ,Humans ,Point Mutation ,Thrombophilia ,Retrospective Studies ,RISK ,Pregnancy Complications, Hematologic ,Pregnancy Outcome ,Anticoagulants ,Thrombosis ,Janus Kinase 2 ,Stillbirth ,Prognosis ,Abortion, Spontaneous ,Pregnancy Complications ,Stroke ,SURVIVAL ,Female ,Thrombocythemia, Essential - Abstract
Although Philadelphia-negative myeloproliferative neoplasms (MPNs) occur typically in middle to advanced age, any age group may be affected, posing a challenge for their management during pregnancy when they occur in young females. There is a high incidence of thromboembolic events and pregnancy complications in patients with myeloproliferative neoplasms, and a possible relationship between these complications is a matter of concern. The aim of this article was to correlate thrombosis and pregnancy outcome in 158 females with ET experiencing 237 pregnancies. Seven patients had a thrombotic event before their first pregnancy, one of them ended (14.3%) in a miscarriage. Among the 151 patients with no history of thrombosis before they became pregnant, 40 (26.5%) had a miscarriage (P = NS). Eighteen patients (11.4%) developed major thrombotic complications (12 splanchnic vein, 1 cerebral vein, 2 coronary syndromes, and 3 strokes) after at least one pregnancy (4 uneventful and 14 complicated). The occurrence of thrombosis was significantly more frequent (P0.001) in patients with a history of pregnancy complications (28%) than in those experiencing a normal pregnancy and delivery (3.7%). Pregnancy complications in women with ET are associated with a higher risk of subsequent thromboses, so pregnant women with this neoplasm who miscarry need to be carefully monitored.
- Published
- 2013
24. EZH2 INHIBITION ENHANCES CAR T ANTITUMOR EFFECT BY INDUCING LYMPHOMA IMMUNOGENICITY AND ENHANCING T CELL FUNCTION.
- Author
-
Isshiki, Y., Porazzi, P., Melnick, A., Ruella, M., and Béguelin, W.
- Subjects
CELL physiology ,T cells ,IMMUNE response ,LYMPHOMAS - Abstract
B Introduction: b Despite recent successes with CAR T-cell therapy against CD19 in patients with B-celllymphomas, only ~30%-40% of DLBCL and FL patients maintain a durable complete long-term remission, and over half of the patients will ultimately relapse. To investigate CAR T-cell-mediated tumor killing in human cells in combination with EZH2i tazemetostat, we used the GCB-DLBCL cell lines SUDHL4 (EZH2 SP Y641F sp ), OCI-Ly18 and Toledo (WT EZH2). [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
25. Post-transplant evaluation of bone marrow hematopoietic function in patients receiving high-dose Yttrium-90-Ibritumomab Tiuxetan (Zevalin) with autograft
- Author
-
Guidetti, A., Carlo Stella, C., Ruella, M., Devizzi, L., Sia, D., Giacomini, A, Locatelli, S. L., Testi, A., Magni, M., Di Nicola, M., Matteucci, P., Buttiglieri, S., Risso, A., Tarella, C., and Gianni, A. M.
- Published
- 2009
26. Anti-CD123 chimeric antigen receptor redirected T cells for relapsed B-cell acute lymphoblastic leukemia
- Author
-
Ruella, M., primary, Shestova, O., additional, Kenderian, S., additional, Barrett, D., additional, Grupp, S., additional, Scholler, J., additional, Lacey, S., additional, Kalos, M., additional, June, C.H., additional, and Gill, S., additional
- Published
- 2014
- Full Text
- View/download PDF
27. Lymphocyte transformation and autoimmune disorders
- Author
-
Tarella, C., primary, Gueli, A., additional, Ruella, M., additional, and Cignetti, A., additional
- Published
- 2013
- Full Text
- View/download PDF
28. Bone marrow-derived cell mobilization by G-CSF to enhance osseointegration of bone substitute in high tibial osteotomy
- Author
-
Marmotti, A., primary, Castoldi, F., additional, Rossi, R., additional, Marenco, S., additional, Risso, A., additional, Ruella, M., additional, Tron, A., additional, Borrè, A., additional, Blonna, D., additional, and Tarella, C., additional
- Published
- 2012
- Full Text
- View/download PDF
29. Comparative assessment of telomere length before and after hematopoietic SCT: role of grafted cells in determining post-transplant telomere status
- Author
-
Ruella, M, primary, Rocci, A, additional, Ricca, I, additional, Carniti, C, additional, Bodoni, C Labetti, additional, Ladetto, M, additional, Caracciolo, D, additional, Boccadoro, M, additional, Carlo-Stella, C, additional, Corradini, P, additional, and Tarella, C, additional
- Published
- 2009
- Full Text
- View/download PDF
30. C011 Marked telomere loss following autologous stem cell transplantation may be predictive for the onset of secondary myelodysplastic syndrome/acute leukemia
- Author
-
Ricca, I., primary, Rocci, A., additional, Zanni, M., additional, Dellacasa, C., additional, Ruella, M., additional, Compagno, M., additional, Caracciolo, D., additional, Ferrero, D., additional, Boccadoro, M., additional, Ladetto, M., additional, and Tarella, C., additional
- Published
- 2007
- Full Text
- View/download PDF
31. The long-term outcome of high-risk follicular lymphoma patients receiving intensive therapy with autograft as primary treatment: Results from three consecutive prospective trials performed in Italy over the past 18 years
- Author
-
Tarella, C., Marco LADETTO, Ruella, M., Passera, R., Pileri, A., Gianni, A. M., and Corradini, P.
32. Adoptive cellular therapy
- Author
-
Ruella, M., Rotolo, A., Katherine Cummins, and June, C. H.
33. Induction of Resistance to Chimeric Antigen Receptor (CAR) T cell Therapy by Transduction of a Single Leukemic B Cell
- Author
-
Ruella, M, primary, Xu, J, additional, Barrett, DM, additional, Kulikovskaya, I, additional, Nazimuddin, F, additional, Bhoj, VG, additional, Orlando, E, additional, Fry, TJ, additional, Bitter, H, additional, Maude, SL, additional, Levine, BL, additional, Nobles, CL, additional, Bushman, FD, additional, Young, RM, additional, Scholler, J, additional, Gill, SI, additional, June, CH, additional, Grupp, SA, additional, Lacey, SF, additional, and Melenhorst, JJ, additional
- Full Text
- View/download PDF
34. Strategy to prevent epitope masking in CAR.CD19+ B-cell leukemia blasts
- Author
-
Pietro Merli, Gianni Cazzaniga, Franco Locatelli, Vittorio Nunes, Zeinab Abbaszadeh, Alberto Orfao, Matilde Sinibaldi, Mattia Algeri, Frederikke Isa Marin, Maria Herrero-Garcia, Stefano Di Cecca, Paolo Marcatili, Biagio De Angelis, Concetta Quintarelli, Sara Gutiérrez-Herrero, Luciana Vinti, Lourdes Martín-Martín, Roselia Ciccone, Biancamaria Cembrola, Simona Songia, Simona Caruso, Iolanda Boffa, Simona Manni, Valentina Bertaina, Marika Guercio, Antonio Camera, Francesca Del Bufalo, Marco Ruella, Quintarelli, C, Guercio, M, Manni, S, Boffa, I, Sinibaldi, M, DI Cecca, S, Caruso, S, Abbaszadeh, Z, Camera, A, Cembrola, B, Ciccone, R, Orfao, A, Martin-Martin, L, Gutierrez-Herrero, S, Herrero-Garcia, M, Cazzaniga, G, Nunes, V, Songia, S, Marcatili, P, Marin, F, Ruella, M, Bertaina, V, Vinti, L, Del Bufalo, F, Algeri, M, Merli, P, De Angelis, B, Locatelli, F, Cancer Research UK, Associazione Italiana per la Ricerca sul Cancro, Asociación Española Contra el Cáncer, and Ministero della Salute
- Subjects
0301 basic medicine ,Cancer Research ,receptor ,receptors ,Epitope ,Epitopes ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Immunology and Allergy ,hematologic neoplasms ,RC254-282 ,Receptors, Chimeric Antigen ,biology ,medicine.diagnostic_test ,Chemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Leukemia ,Oncology ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,030220 oncology & carcinogenesis ,translational medical research ,Molecular Medicine ,immunotherapy ,Immunology ,cell engineering ,adoptive ,CD19 ,Flow cytometry ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,Leukemia, B-Cell ,medicine ,Animals ,Humans ,Pharmacology ,Immune Cell Therapies and Immune Cell Engineering ,medicine.disease ,Chimeric antigen receptor ,In vitro ,Disease Models, Animal ,030104 developmental biology ,B-cell leukemia ,chimeric antigen ,Cancer research ,biology.protein ,human activities ,hematologic neoplasm - Abstract
[Abstract]: Chimeric antigen receptor T-cells (CAR T-cells) for the treatment of relapsing/refractory B-cell precursor acute lymphoblastic leukemia have led to exciting clinical results. However, CAR T-cell approaches revealed a potential risk of CD19-/CAR+ leukemic relapse due to inadvertent transduction of leukemia cells. [Methods]: We evaluated the impact of a high percentage of leukemia blast contamination in patient-derived starting material (SM) on CAR T-cell drug product (DP) manufacturing. In vitro as well as in vivo models were employed to identify characteristics of the construct associated with better profile of safety in case of inadvertent B-cell leukemia transduction during CAR T-cell manufacturing. [Results]: The presence of large amounts of CD19+ cells in SM did not affect the transduction level of DPs, as well as the CAR T-cell rate of expansion at the end of standard production of 14 days. DPs were deeply characterized by flow cytometry and molecular biology for Ig-rearrangements, showing that the level of B-cell contamination in DPs did not correlate with the percentage of CD19+ cells in SM, in the studied patient cohort. Moreover, we investigated whether CAR design may affect the control of CAR+ leukemia cells. We provided evidences that CAR.CD19 short linker (SL) prevents complete epitope masking in CD19+CAR+ leukemia cells and we demonstrated in vitro and in vivo that CD19 +CAR(SL)+leukemic cells are killed by CAR.CD19 T-cells. [Conclusions]: Taken together, these data suggest that a VL-VH SL may result in a safe CAR-T product, even when manufacturing starts from biological materials characterized by heavy contamination of leukemia blasts., The experimental work was supported by grants awarded by Ricerca Finalizzata GR-2013 02359212 (CQ), GR-2016-02364546 (BDA), RF-2016- 02364388 (FL), Accelerator Award-Cancer Research UK/AIRC/AECC-INCAR project (FL and AO), Associazione Italiana Ricerca per la Ricerca sul Cancro (AIRC)-Special Project 5×1000 no. 9962 (FL), AIRC IG 2018 id. 21724 (FL), Ricerca Corrente (FL, CQ, BDA), Ministero dell’Università e della Ricerca (Grant PRIN 2017 to FL); Italian Healthy Ministry project on CAR T RCR-2019-23669115 (coordinator FL), Independent Research grant AIFA (FL PI: 2016 call).
- Published
- 2021
35. The aging effect of chemotherapy on cultured human mesenchymal stem cells
- Author
-
Corrado Tarella, Alessandra Risso, Tiziana Spatola, Stefano Buttiglieri, Lorenzo Silengo, Marco Ruella, Enrico Vittorio Avvedimento, Buttiglieri, S., Ruella, M., Risso, A., Spatola, T., Silengo, L., Avvedimento, VITTORIO ENRICO, and Tarella, C.
- Subjects
Adult ,Senescence ,Cancer Research ,Telomerase ,Bone Marrow Cells ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,Immunophenotyping ,Adipocytes ,Genetics ,medicine ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Clonogenic assay ,Molecular Biology ,Cells, Cultured ,Cellular Senescence ,Tumor Stem Cell Assay ,Etoposide ,Antibiotics, Antineoplastic ,Osteoblasts ,Dose-Response Relationship, Drug ,Tumor Suppressor Proteins ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Hematology ,Telomere ,Antineoplastic Agents, Phytogenic ,Neoplasm Proteins ,DNA-Binding Proteins ,medicine.anatomical_structure ,Doxorubicin ,Immunology ,Cancer research ,Bone marrow ,Stem cell ,Protein Processing, Post-Translational - Abstract
Various agents, including chemotherapeutic drugs, can induce cell senescence. However, the mechanisms involved in the aging pathway, particularly the stress that chemotherapy imposes on telomeres, are still undefined. To address these issues, human mesenchymal stem cells (MSCs) were assessed as target cells to investigate the initiation of the aging process by chemotherapy. The MSCs were obtained from bone marrow (BM) cells from normal adults and grown in the presence of platelet lysates. Cultured MSCs were identified for immunophenotype, and for growth and differentiation properties. The MSCs were exposed to 10 nM doxorubicin and 500 ng/mL etoposide, sublethal doses that induce DNA double-stranded breaks. Telomere length (TL) was assessed by flow-fluorescence in situ hybridization and Southern blotting. Initial TL shortening was detectable in MSCs at 5 days after drug exposure, with progressive reduction compared with untreated cells at 7, 14, 21, and 28 days in culture. After a single exposure, MSCs were unable to regain the lost telomere sequences for up to 28 days in culture. The ATM phosphorylation was documented early after drug exposure, while no telomerase activation was observed. Chemotherapy-induced TL shortening was associated with reduced clonogenic activity in vitro and accelerated adipose differentiation. Analogous behavior in the differentiation pattern was observed in naturally aged MSCs. These results indicate that cultured MSCs represent a useful cellular model to investigate novel drugs that may favor or, conversely, might prevent TL loss in human stem cells. The TL shortening is a permanent signature of previous chemotherapy-mediated DNA damage, and predicts impaired proliferative and differentiation potential.
- Published
- 2011
36. A Recent Update of Three Consecutive Prospective Trials with High-Dose Therapy and Autograft, without or with Rituximab, as Primary Treatment for Advanced-Stage Follicular Lymphoma (FL) Shows a Sizeable Group of Patients Surviving in Continuous Complete Remission up to 16 Years After the End of Treatment: Should We Still Consider FL An Incurable Disease ?
- Author
-
Delia Rota Scalabrini, Umberto Vitolo, Corrado Tarella, Francesco Rodeghiero, Ignazio Majolino, Alessandro Levis, Roberto Passera, Giuseppe Rossi, Alessandro Massimo Gianni, Angelo Michele Carella, Andrea Gallamini, Maurizio Musso, Teodoro Chisesi, Alberto De Crescenzo, Guido Gini, Alessandro Pulsoni, Pierangelo Spedini, Fabio Benedetti, Emilio Iannitto, Guido Parvis, Francesco Di Raimondo, Tommasina Perrone, Marco Ruella, Paolo Corradini, Michele Magni, Andrea Piccin, Alessandro Rambaldi, Fabio Ciceri, Caterina Patti, Anna Marina Liberati, Vincenzo Callea, Alessandro Pileri, Marco Ladetto, Tarella, C, Ladetto, M, Benedetti, F, Vitolo, U, Pulsoni, A, Patti, C, Callea, V, Rambaldi, A, Piccin, A, Magni, M, Musso, M, Iannitto, E, Spedini, P, Liberati, Am, Ciceri, Fabio, Gallamini, A, Rodeghiero, F, Gini, G, De Crescenzo, A, Di Raimondo, F, Levis, A, Chisesi, T, Perrone, T, Scalabrini, Dr, Rossi, G, Carella, Am, Parvis, G, Majolino, I, Passera, R, Ruella, M, Pileri, A, Gianni, A, and Corradini, P.
- Subjects
medicine.medical_specialty ,Chemotherapy ,Acute leukemia ,Cyclophosphamide ,business.industry ,medicine.medical_treatment ,Immunology ,Follicular lymphoma ,Phases of clinical research ,Cell Biology ,Hematology ,medicine.disease ,Debulking ,Biochemistry ,Gastroenterology ,Surgery ,Regimen ,Internal medicine ,medicine ,Rituximab ,business ,medicine.drug - Abstract
Abstract 882 Background. Three consecutive trials have been performed in Italy over the last 18 years, to verify the efficacy of the use of High-Dose Sequential Chemotherapy (HDS) and autograft as first-line therapy for high-risk Follicular Lymphoma (FL) Patients and Methods. The HDS regimen has been previously described (Corradini P et al, Blood 1997; Tarella C et al, Stem Cells 1998). Briefly, it consists of intensive debulking (2 APO courses +/− 2 DHAP courses) followed by the high-dose phase, including the sequential administration of etoposide (2 g/sqm), methotrexate (MTX) (8 g/sqm) and cyclophosphamide (CY) (7 g/sqm). PBPC collection is scheduled after the last course to maximize the “in vivo purging effect” operated by high-dose chemotherapy. The final autologous stem cell transplant (auto-SCT) conclude the program, two conditioning regimen have been employed, either the BEAM schedule or the Mitoxantrone/L-PAM combination. In the most recent schedule, Rituximab was included in place of MTX. In details, 2 Rituximab doses were administered before CY, after CY and after auto-SCT, with the aim of further improving disease control and the in-vivo purging. The first trial was a single Center phase II study exploring both feasibility and efficacy of the HDS program as first line therapy in advanced-stage indolent lymphoma (1991-1998, 26 FL patients) (Tarella C et al, Leukemia 2000); a subsequent multicenter phase 2 trial was then started at national level (GITMO, Gruppo Italiano Trapianto Midollo Osseo), to verify the efficacy of HDS in advanced-stage FL in a multicenter setting (1996-1999, 92 patients) (Ladetto M et al, Blood 2002); lastly, a muticenter phase 3 study was performed together with GITMO and IIL (Intergruppo Italiano Linfomi) Centers, comparing Rituximab supplemented HDS (R-HDS) vs. CHOP-R in aaIPI 2-3 FL (2000-2005, 68 patients in the R-HDS arm) (Ladetto M et al, Blood 2008). Overall, 186 patients have been treated with HDS, updated results have been obtained for 168 of them. They all had a diagnosis of FL (grade 1-2: 71%) and always presented with advanced stage, their median age was 48 yrs., LDH was high in 48%, BM involved in 77%. Results. 140 patients out of 168 (83%) attained Complete Remission (CR); there were 6 early toxic deaths (3.6%); 8 patients had Partial Remission (4.8%) and 14 had no response (8.3%), soon followed by disease progression. So far 14 patients (8.3%) developed secondary myelodysplasia or acute leukemia (sMDS/AL), and 7 patients (4.2%) had a secondary solid neoplasia. As of July 2008, 50 of 168 patients died, due to: i. early toxicity (6 patients); ii. disease progression (25 patients, 15%); iii. second neoplasia (12 patients, 7.1%); iv. other causes (7 patients, 4.2%). Thus, at a median follow-up of 10 yrs., 118 patients (70.2%) are alive, and 80 (48%) are in their 1st continuous CR (CCR), and most of them are also in molecular remission. The actuarial OS and DFS curves are reported in Figures 1A and B. The latest relapse has been recorded at 8 yrs since HDS. So far, 50 patients (30%) are presently in their 1st CCR between 8 and 16 yrs after HDS. Conclusions. i. advanced stage FL treated upfront with the intensive HDS regimen had a prolonged survival, with median survival not yet reached after 10 yrs. of follow-up; ii. main causes of death were disease progression and both early and late toxic side effects; iii. approximately half of the patients are long-term survivors without any sign of disease recurrence. This suggest that a prolonged PFS and possibly the disease eradication should be pursued also in advanced-stage FL. Future studies will verify whether these therapeutic goals may be achieved with chemo-immunotherapeutic schemes at least as effective but less toxic and laborious than HDS program with autograft. Disclosures: Tarella: Roche: Honoraria, research financial support. Ladetto:Roche: research financial support. Vitolo:Roche: Lecture fees. Rambaldi:Roche: Honoraria. Corradini:Roche: Honoraria.
- Published
- 2009
37. Prevalence and Prognostication of CD5+ Mature T-Cell Lymphomas.
- Author
-
Elghawy O, Cao M, Xu J, Landsburg DJ, Svoboda J, Nasta SD, Chong EA, Schuster SJ, Thomas CJ, Carter JS, Tavakkoli M, Ruella M, and Barta SK
- Abstract
Background: T-cell lymphomas (TCLs) are a group of heterogenous cancers with poor rates and duration of response. There remains a great challenge in risk stratification of these cancers. Cluster of differentiation (CD) 5 has shown prognostic implication in many subtypes of B-cell lymphoma; however, its role in TCLs is not known. Methods: We performed a single-institution retrospective analysis of newly diagnosed patients with TCL. CD5 positivity was determined based on positive results via immunohistochemistry and/or flow cytometry. We used univariate and multivariable analysis of biological factors to assess their association with survival outcomes. Results: A total of 194 patients with TCL spanning 14 subtypes were identified. CD5 positivity was noted in 63% of patients, with the highest proportion of CD5 expression in TFH TCL (93.9%), PTCL-NOS (82.9%), and ATLL (77.8%) ( p = 0.00004). Older age at diagnosis ( p = 0.001), stage III or IV disease ( p = 0.05), and bone marrow involvement ( p = 0.003) were also associated with CD5 expression. Complete response rates were numerically lower in patients with CD5+ TCL across all subtypes. OS/PFS was not statistically associated with CD5 status in the overall cohort; however there was significantly decreased OS in CD5+ TFH TCL ( p = 0.04) and CD5+ ATLL ( p = 0.04) patients. Conclusions: This study represents the first to examine CD5 expression as a prognostic biomarker for outcomes in TCL. The frequent expression of CD5 in the most common nodal TCL in the Western world underpins its potential as an attractive target for cellular therapies. Confirmation of these findings in a larger cohort and investigation of potential pathophysiological mechanisms explaining our observations are planned.
- Published
- 2024
- Full Text
- View/download PDF
38. Directed evolution-based discovery of ligands for in vivo restimulation of CAR-T cells.
- Author
-
Grzywa T, Mehta N, Cossette B, Romanov A, Paruzzo L, Ramasubramanian R, Cozzone A, Morgan D, Sukaj I, Bergaggio E, Tannir R, Kadauke S, Myers R, Yousefpour P, Ghilardi G, Schuster S, Neeser A, Frey N, Goncalves B, Zhang L, Abraham W, Suh H, Ruella M, Grupp S, Chiarle R, Wittrup KD, Ma L, and Irvine DJ
- Abstract
Chimeric antigen receptor (CAR) T cell therapy targeting CD19 elicits remarkable clinical efficacy in B-cell malignancies, but many patients relapse due to failed expansion and/or progressive loss of CAR-T cells. We recently reported a strategy to potently restimulate CAR-T cells in vivo, enhancing their functionality by administration of a vaccine-like stimulus comprised of surrogate peptide ligands for a CAR linked to a lymph node-targeting amphiphilic PEG-lipid (termed CAR-T-vax). Here, we demonstrate a general strategy to generate and optimize peptide mimotopes enabling CAR-T-vax generation for any CAR. Using the clinical CD19 CAR FMC63 as a test case, we employed yeast surface display to identify peptide binders to soluble IgG versions of FMC63, which were subsequently affinity matured by directed evolution. CAR-T vaccines using these optimized mimotopes triggered marked expansion of both murine CD19 CAR-T cells in a syngeneic model and human CAR-T cells in a humanized mouse model of B cell acute lymphoblastic leukemia (B-ALL), and enhanced control of leukemia progression. This approach thus enables vaccine boosting to be applied to any clinically-relevant CAR-T cell product.
- Published
- 2024
- Full Text
- View/download PDF
39. Targeting TGFβ-activated kinase-1 activation in microglia reduces CAR T immune effector cell-associated neurotoxicity syndrome.
- Author
-
Vinnakota JM, Biavasco F, Schwabenland M, Chhatbar C, Adams RC, Erny D, Duquesne S, El Khawanky N, Schmidt D, Fetsch V, Zähringer A, Salié H, Athanassopoulos D, Braun LM, Javorniczky NR, Ho JNHG, Kierdorf K, Marks R, Wäsch R, Simonetta F, Andrieux G, Pfeifer D, Monaco G, Capitini C, Fry TJ, Blank T, Blazar BR, Wagner E, Theobald M, Sommer C, Stelljes M, Reicherts C, Jeibmann A, Schittenhelm J, Monoranu CM, Rosenwald A, Kortüm M, Rasche L, Einsele H, Meyer PT, Brumberg J, Völkl S, Mackensen A, Coras R, von Bergwelt-Baildon M, Albert NL, Bartos LM, Brendel M, Holzgreve A, Mack M, Boerries M, Mackall CL, Duyster J, Henneke P, Priller J, Köhler N, Strübing F, Bengsch B, Ruella M, Subklewe M, von Baumgarten L, Gill S, Prinz M, and Zeiser R
- Subjects
- Animals, Mice, Humans, Immunotherapy, Adoptive methods, p38 Mitogen-Activated Protein Kinases metabolism, NF-kappa B metabolism, Lymphoma, B-Cell immunology, Antigens, CD19 immunology, Female, T-Lymphocytes immunology, Signal Transduction, MAP Kinase Kinase Kinases metabolism, MAP Kinase Kinase Kinases genetics, Microglia immunology, Microglia metabolism, Neurotoxicity Syndromes etiology, Neurotoxicity Syndromes immunology, Receptors, Chimeric Antigen immunology
- Abstract
Cancer immunotherapy with chimeric antigen receptor (CAR) T cells can cause immune effector cell-associated neurotoxicity syndrome (ICANS). However, the molecular mechanisms leading to ICANS are not well understood. Here we examined the role of microglia using mouse models and cohorts of individuals with ICANS. CD19-directed CAR (CAR19) T cell transfer in B cell lymphoma-bearing mice caused microglia activation and neurocognitive deficits. The TGFβ-activated kinase-1 (TAK1)-NF-κB-p38 MAPK pathway was activated in microglia after CAR19 T cell transfer. Pharmacological TAK1 inhibition or genetic Tak1 deletion in microglia using Cx3cr1
CreER :Tak1fl/fl mice resulted in reduced microglia activation and improved neurocognitive activity. TAK1 inhibition allowed for potent CAR19-induced antilymphoma effects. Individuals with ICANS exhibited microglia activation in vivo when studied by translocator protein positron emission tomography, and imaging mass cytometry revealed a shift from resting to activated microglia. In summary, we prove a role for microglia in ICANS pathophysiology, identify the TAK1-NF-κB-p38 MAPK axis as a pathogenic signaling pathway and provide a rationale to test TAK1 inhibition in a clinical trial for ICANS prevention after CAR19 T cell-based cancer immunotherapy., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
40. Immunologic Predictors of Vaccine Responsiveness in Patients With Lymphoma and Chronic Lymphocytic Leukemia.
- Author
-
Chong EA, Kumashie KG, Chong ER, Fabrizio J, Gupta A, Svoboda J, Barta SK, Walsh KM, Napier EB, Lundberg RK, Nasta SD, Gerson JN, Landsburg DJ, Gonzalez J, Gaano A, Weirick ME, McAllister CM, Awofolaju M, John GN, Kammerman SC, Novacek J, Pajarillo R, Lundgreen KA, Tanenbaum N, Gouma S, Drapeau EM, Adamski S, D'Andrea K, Pattekar A, Hicks A, Korte S, Sharma H, Herring S, Williams JC, Hamilton JT, Bates P, Hensley SE, Prak ETL, Greenplate AR, Wherry EJ, Schuster SJ, Ruella M, and Vella LA
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Retrospective Studies, Prospective Studies, SARS-CoV-2 immunology, Adult, Antibodies, Viral blood, Antibodies, Viral immunology, Vaccination, Immunoglobulin M blood, Lymphoma immunology, Lymphoma therapy, Aged, 80 and over, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell therapy, COVID-19 Vaccines immunology, COVID-19 Vaccines administration & dosage, COVID-19 immunology, COVID-19 prevention & control
- Abstract
Patients with B-cell lymphomas have altered cellular components of vaccine responses due to malignancy and therapy, and the optimal timing of vaccination relative to therapy remains unknown. Severe acute respiratory syndrome coronavirus 2 vaccines created an opportunity for new insights in vaccine timing because patients were challenged with a novel antigen across multiple phases of treatment. We studied serologic messenger RNA vaccine response in retrospective and prospective cohorts with lymphoma and chronic lymphocytic leukemia, paired with clinical and research immune parameters. Reduced serologic response was observed more frequently during active treatment, but nonresponse was also common within observation and posttreatment groups. Total immunoglobulin A and immunoglobulin M correlated with successful vaccine response. In individuals treated with anti-CD19-directed chimeric antigen receptor-modified T cells, nonresponse was associated with reduced B and T follicular helper cells. Predictors of vaccine response varied by disease and therapeutic group, and therefore further studies of immune health during and after cancer therapies are needed to individualize vaccine timing., Competing Interests: Potential conflicts of interest. E. A. C. receives research support from Genentech/Roche and AbbVie and has served as a consultant for Beigene, AstraZeneca, and TG Therapeutics. J. S. has served as a consultant for ATARA, AstraZeneca, Celgene Corporation, Adaptive, and Genmab, and receives research support from AstraZeneca, Merck, Incyte, Bristol-Myers Squibb, Pharmacyclics, TG Therapeutics, Seattle Genetics, and Adaptive. S. K. B. received honoraria from Acrotech, Seagen, Kyowa Kirin, and Daiichi Sankyo. S. D. N. receives research support from Roche, Rafael, ATARA, Pharmacyclics, and Takeda/Millennium; is a data monitoring committee member for Merck; and has served as a consultant for Epizyme and Morphosys. D. J. L. received research funding from Curis and Triphase; is a data and safety monitoring board member for Karyopharm; and has served as a consultant for Morphosys/Incyte, Epizyme, and ADC Therapeutics. J. N. G. received research funding from Loxo and has served as a consultant for Genentech, AbbVie, and Kite. E. L. P. received funding from Roche Diagnostics Corporation for the evaluation of serologic tests for SARS-CoV-2. E. J. W. is consulting or is an advisor for Merck, Marengo, Janssen, Related Sciences, Synthekine, and Surface Oncology; is a founder of Surface Oncology, Danger Bio, and Arsenal Biosciences; and is an inventor on a patent (US patent number 10,370,446) submitted by Emory University that covers the use of PD-1 blockade to treat infections and cancer. S. J. S. reports research funding from Acerta, Celgene, Genentech/Roche, Merck, Novartis, Pharmacyclics, and TG Therapeutics; received honoraria/consulting fees from Acerta, AstraZeneca, Celgene, Incyte, Janssen, Loxo Oncology, Morphosys, and Nordic Nanovector; is a steering committee member for Celgene, Nordic Nanovector, and Novartis; and has a patent for combination therapies of CAR T cells and PD-1 inhibitors. M. R. holds patents related to CAR T cells; has served as a consultant for nanoString, BMS, GSK, Bayer, Sana Therapeutics, and AbClon; receives research funding from AbClon, nanoString, viTToria biotherapeutics, Oxford Nanoimaging, and Beckman Coulter; and is the scientific founder of viTToria Biotherapeutics. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2024. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
41. CD5 deletion enhances the antitumor activity of adoptive T cell therapies.
- Author
-
Patel RP, Ghilardi G, Zhang Y, Chiang YH, Xie W, Guruprasad P, Kim KH, Chun I, Angelos MG, Pajarillo R, Hong SJ, Lee YG, Shestova O, Shaw C, Cohen I, Gupta A, Vu T, Qian D, Yang S, Nimmagadda A, Snook AE, Siciliano N, Rotolo A, Inamdar A, Harris J, Ugwuanyi O, Wang M, Carturan A, Paruzzo L, Chen L, Ballard HJ, Blanchard T, Xu C, Abdel-Mohsen M, Gabunia K, Wysocka M, Linette GP, Carreno B, Barrett DM, Teachey DT, Posey AD, Powell DJ Jr, Sauter CT, Pileri S, Pillai V, Scholler J, Rook AH, Schuster SJ, Barta SK, Porazzi P, and Ruella M
- Subjects
- Animals, Mice, Humans, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen genetics, Cell Line, Tumor, CRISPR-Cas Systems, Female, Immunotherapy, Adoptive methods, CD5 Antigens immunology, T-Lymphocytes immunology, T-Lymphocytes transplantation
- Abstract
Most patients treated with US Food and Drug Administration (FDA)-approved chimeric antigen receptor (CAR) T cells eventually experience disease progression. Furthermore, CAR T cells have not been curative against solid cancers and several hematological malignancies such as T cell lymphomas, which have very poor prognoses. One of the main barriers to the clinical success of adoptive T cell immunotherapies is CAR T cell dysfunction and lack of expansion and/or persistence after infusion. In this study, we found that CD5 inhibits CAR T cell activation and that knockout (KO) of CD5 using CRISPR-Cas9 enhances the antitumor effect of CAR T cells in multiple hematological and solid cancer models. Mechanistically, CD5 KO drives increased T cell effector function with enhanced cytotoxicity, in vivo expansion, and persistence, without apparent toxicity in preclinical models. These findings indicate that CD5 is a critical inhibitor of T cell function and a potential clinical target for enhancing T cell therapies.
- Published
- 2024
- Full Text
- View/download PDF
42. Bendamustine as Lymphodepletion for Brexucabtagene Autoleucel Therapy of Mantle Cell Lymphoma.
- Author
-
Chong EA, Chong ER, Therwhanger D, Nasta SD, Landsburg DJ, Barta SK, Svoboda J, Gerson JN, Ghilardi G, Paruzzo L, Fraietta JA, Weber E, Stefano N, Porter DL, Frey NV, Garfall AL, Ruella M, and Schuster SJ
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Retrospective Studies, Vidarabine analogs & derivatives, Vidarabine therapeutic use, Vidarabine administration & dosage, Adult, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating administration & dosage, Antigens, CD19 immunology, Treatment Outcome, Bendamustine Hydrochloride therapeutic use, Bendamustine Hydrochloride administration & dosage, Lymphoma, Mantle-Cell drug therapy, Immunotherapy, Adoptive methods
- Abstract
Brexucabtagene autoleucel (brexu-cel) is an autologous CD19-directed chimeric antigen receptor (CAR) T-cell therapy approved for treatment of relapsed/refractory mantle cell lymphoma (MCL). During a fludarabine shortage, we used bendamustine as an alternative to standard cyclophosphamide/fludarabine (cy/flu) lymphodepletion (LD) prior to brexu-cel. We assessed MCL patient outcomes as well as CAR T-cell expansion and persistence after brexu-cel following bendamustine or cy/flu LD at our center. This was a retrospective single institution study that utilized prospectively banked blood and tissue samples. Clinical efficacy was assessed by 2014 Lugano guidelines. CAR T-cell expansion and persistence in peripheral blood were assessed on day 7 and at ≥month 6 for patients with available samples. Seventeen patients received bendamustine and 5 received cy/flu. For the bendamustine cohort, 14 (82%) received bridging therapy and 4 (24%) had CNS involvement. Fifteen patients (88%) developed CRS with 4 (24%) ≥grade 3 events. Six (35%) patients developed ICANS with 4 (24%) events ≥grade 3. No patient had ≥grade 3 cytopenias at day 90. Best objective (BOR) and complete response (CRR) rates were 82% and 65%, respectively. At 24.5 months median follow-up, 12-month progression-free survival (PFS) was 45%, 24-month PFS was 25%, and median duration of response was 19 months. Median OS was not reached. BOR was 25% (1/4) for patients with CNS involvement. CAR transgene expansion after bendamustine LD was observed on day 7 in all (4/4) patients tested and persisted at ≥6 months (2/2), regardless of response. Bendamustine LD before brexu-cel for MCL is feasible and safe with a lower frequency and shorter duration of cytopenias than reported for cy/flu. Both CAR T-cell expansion and persistence were observed after bendamustine LD. Outcomes appear comparable to the real world outcomes reported with cy/flu LD., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
43. CAR-T Therapy for All: Addressing the Access Gap.
- Author
-
Ruella M
- Subjects
- Humans, Receptors, Chimeric Antigen, Health Services Accessibility, Immunotherapy, Adoptive
- Published
- 2024
- Full Text
- View/download PDF
44. CAR T-cell Resistance to Oncogenic Transformation.
- Author
-
Ruella M and June CH
- Subjects
- Humans, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell metabolism, Neoplasms immunology, Neoplasms therapy, Immunotherapy, Adoptive methods, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen genetics, Cell Transformation, Neoplastic immunology, Cell Transformation, Neoplastic genetics, T-Lymphocytes immunology
- Abstract
In this commentary, we discuss the investigation into reports of T-cell malignancies following chimeric antigen receptor T-cell therapy. We argue that although these cases should be thoroughly examined, current data suggest that such risks with autologous chimeric antigen receptor T cells are remarkably low compared with other cancer treatments. We also emphasize the importance of continued research, transparent reporting, and participation in postauthorization safety studies., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
45. Recent Advances in Immune-Based Therapies for Acute Myeloid Leukemia.
- Author
-
Restelli C, Ruella M, Paruzzo L, Tarella C, Pelicci PG, and Colombo E
- Subjects
- Humans, Cancer Vaccines therapeutic use, Cancer Vaccines immunology, Tumor Microenvironment immunology, Tumor Microenvironment drug effects, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors pharmacology, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute therapy, Immunotherapy methods, Immunotherapy trends
- Abstract
Despite advancements, acute myeloid leukemia (AML) remains unconquered by current therapies. Evidence of immune evasion during AML progression, such as HLA loss and T-cell exhaustion, suggests that antileukemic immune responses contribute to disease control and could be harnessed by immunotherapy. In this review, we discuss a spectrum of AML immunotherapy targets, encompassing cancer cell-intrinsic and surface antigens as well as targeting in the leukemic milieu, and how they can be tailored for personalized approaches. These targets are overviewed across major immunotherapy modalities applied to AML: immune checkpoint inhibitors, antibody-drug conjugates, therapeutic vaccines, bispecific/trispecific antibodies, and chimeric antigen receptor (CAR)-T and CAR-NK cells. Significance: Immune therapies in AML treatment show evolving promise. Ongoing research aims to customize approaches for varied patient profiles and clinical scenarios. This review covers immune surveillance mechanisms, therapy options like checkpoint inhibitors, antibodies, CAR-T/NK cells, and vaccines, as well as resistance mechanisms and microenvironment considerations., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
46. The BTLA-HVEM axis restricts CAR T cell efficacy in cancer.
- Author
-
Guruprasad P, Carturan A, Zhang Y, Cho JH, Kumashie KG, Patel RP, Kim KH, Lee JS, Lee Y, Kim JH, Chung J, Joshi A, Cohen I, Shestov M, Ghilardi G, Harris J, Pajarillo R, Angelos M, Lee YG, Liu S, Rodriguez J, Wang M, Ballard HJ, Gupta A, Ugwuanyi OH, Hong SJA, Bochi-Layec AC, Sauter CT, Chen L, Paruzzo L, Kammerman S, Shestova O, Liu D, Vella LA, Schuster SJ, Svoboda J, Porazzi P, and Ruella M
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Mice, Knockout, Neoplasms immunology, Neoplasms therapy, Signal Transduction, T-Lymphocytes, Regulatory immunology, Immunotherapy, Adoptive methods, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Receptors, Chimeric Antigen genetics, Receptors, Immunologic metabolism, Receptors, Immunologic genetics, Receptors, Tumor Necrosis Factor, Member 14 metabolism, Receptors, Tumor Necrosis Factor, Member 14 immunology, Receptors, Tumor Necrosis Factor, Member 14 genetics, Tumor Microenvironment immunology
- Abstract
The efficacy of T cell-based immunotherapies is limited by immunosuppressive pressures in the tumor microenvironment. Here we show a predominant role for the interaction between BTLA on effector T cells and HVEM (TNFRSF14) on immunosuppressive tumor microenvironment cells, namely regulatory T cells. High BTLA expression in chimeric antigen receptor (CAR) T cells correlated with poor clinical response to treatment. Therefore, we deleted BTLA in CAR T cells and show improved tumor control and persistence in models of lymphoma and solid malignancies. Mechanistically, BTLA inhibits CAR T cells via recruitment of tyrosine phosphatases SHP-1 and SHP-2, upon trans engagement with HVEM. BTLA knockout thus promotes CAR signaling and subsequently enhances effector function. Overall, these data indicate that the BTLA-HVEM axis is a crucial immune checkpoint in CAR T cell immunotherapy and warrants the use of strategies to overcome this barrier., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
47. Delimitation of the risk area of the vertebral artery during the paramedian suboccipital approach.
- Author
-
Villamil F, Caffaratti G, Ruella M, Laplace LD, Calandri I, Darakdjian M, Nuñez M, Mormandi R, and Cervio A
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Retrospective Studies, Aged, Prospective Studies, Adolescent, Young Adult, Neurosurgical Procedures methods, Neuronavigation methods, Vertebral Artery anatomy & histology, Vertebral Artery diagnostic imaging
- Abstract
Objective: The V3 segment of the vertebral artery (V3-VA) is at risk during diverse approaches to the craniovertebral junction. Our objective is to present a system of anatomic and topographic landmarks to identify the V3-VA during the paramedian suboccipital approach (PMSOA) with the help of minimal or basic tools., Material and Methods: The first was a retrospective analysis of the angiotomography (CTA) of 50 patients over 18-years old, and 9 anatomical dissections. A series of lines were defined between the different bony landmarks. Within this lines the risk area of the vertebral artery (RAsV3-VA) and the risk point of the vertebral artery (RPsV3-VA) were defined. The second stage was a prospective study, where the previously defined measurements were carried out by using neuronavigation in 10 patients (20 sides) operated with the PMSO approach in order to confirm the presence of the V3 segment in the RAsV3-VA and RPsV3-VA., Results: In the first stage, the V3 segment was found in the middle third of the X line in 96,6% of the cases. The distance between the inion and the UCP (percentile 5) was 20 mm and to the LCP (percentile 95) was 40 mm. In the range between the UCP and the LCP, in the middle third of the inion-mastoid line (RAsV3-VA), we found 90% of the V3-VA. The measurements taken during the second stage revealed that the artery was in the middle third of the X line in 97% of the cases. 85% of the patients presented the total of the V3s-VA on the RAsV3-VA and in 85% there was a direct relationship with the V3 segment and the RPV3s-VA., Conclusion: We propose an easy-to-implement system to delimit the risk area of the V3-VA during the PMSOA. We believe that these landmarks provide a practical, reliable, costless and useful tool that could decrease the risk of lesion of the V3-VA during this approach without the need of using., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
48. Streamlined measurement of chimeric antigen receptor T-cell concentration, size, viability and two-color phenotyping during manufacturing.
- Author
-
Pajarillo R, Paruzzo L, Carturan A, Ugwuanyi O, White G, Guruprasad P, Ballard HJ, Patel RP, Zhang Y, Lee YG, Hong SJA, Dittami GM, and Ruella M
- Subjects
- Humans, Antigens, CD19 immunology, Antigens, CD19 metabolism, Phenotype, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism, Immunophenotyping methods, Cell Size, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism, Cell Survival, Flow Cytometry methods, Immunotherapy, Adoptive methods, T-Lymphocytes immunology, T-Lymphocytes metabolism
- Abstract
Background Aims: The successful development of CD19-targeted chimeric antigen receptor (CAR) T-cell therapies has led to an exponential increase in the number of patients recieving treatment and the advancement of novel CAR T products. Therefore, there is a strong need to develop streamlined platforms that allow rapid, cost-effective, and accurate measurement of the key characteristics of CAR T cells during manufacturing (i.e., cell number, cell size, viability, and basic phenotype)., Methods: In this study, we compared the novel benchtop cell analyzer Moxi GO II (ORFLO Technologies), which enables simultaneous evaluation of all the aforementioned parameters, with current gold standards in the field: the Multisizer Coulter Counter (cell counter) and the BD LSRFortessa (flow cytometer)., Results: Our results demonstrated that the Moxi GO II can accurately measure cell number and cell size (i.e., cell volume) while simultaneously assessing simple two-color flow cytometry parameters, such as CAR T-cell viability and CD4 or CAR expression., Conclusions: These measurements are comparable with those of gold standard instruments, demonstrating that the Moxi GO II is a promising platform for quickly monitoring CAR T-cell growth and phenotype in research-grade and clinical samples., Competing Interests: Declaration of Competing Interest MR holds patents related to CAR T cells that are licensed to Novartis, Tmunity (Kite) and viTToria biotherapeutics. MR has served as a consultant for nanoString, BMS, GSK, GLG, GuidePoint, Sana, Bayer and AbClon. MR receives research funding from AbClon, Beckman Coulter, Oxford Nano Imaging, Curiox and viTToria biotherapeutics. MR is the scientific founder of viTToria biotherapeutics. The Ruella laboratory was provided a Moxi GO II device and supplies to perform this study. GD is an employee of ORFLO Technologies. All other authors declare no competing interests., (Copyright © 2024 International Society for Cell & Gene Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
49. Efficacy and safety of bendamustine for lymphodepletion before lisocabtagene maraleucel.
- Author
-
Ghilardi G, Paruzzo L, Patel V, Svoboda J, Chong ER, Fardella E, Chong EA, Gabrielli G, Nasta SD, Landsburg DJ, Carter J, Pajarillo R, Barta SK, White G, Weber E, Napier E, Porter DL, Garfall AL, Schuster SJ, and Ruella M
- Subjects
- Humans, Middle Aged, Male, Female, Aged, Retrospective Studies, Adult, Lymphoma, Large B-Cell, Diffuse drug therapy, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Biological Products therapeutic use, Biological Products adverse effects, Aged, 80 and over, Treatment Outcome, Bendamustine Hydrochloride therapeutic use, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects
- Abstract
Bendamustine has been retrospectively shown to be an effective and safe lymphodepletion regimen prior to the anti-CD19 chimeric antigen receptor T cell (CART) products tisagenlecleucel and axicabtagene ciloleucel, as well as the anti-BCMA CART products idecabtagene vicleucel and ciltacabtagene autoleucel. However, bendamustine as lymphodepletion prior to lisocabtagene maraleucel (liso-cel), a 4-1BB co-stimulated, fixed CD4:CD8 ratio anti-CD19 CART product, has not been described yet. Thus, we studied a cohort of sequentially-treated patients with large B-cell lymphomas who received bendamustine lymphodepletion before liso-cel at the University of Pennsylvania between 5/2021 and 12/2023 (n = 31). Patients were evaluated for toxicities and responses. Of note, 7 patients (22.6%) would have dnot met the inclusion criteria for the registrational liso-cel clinical trials, mostly due to older age. Overall and complete response rates were 76.9% and 73.1%, respectively. At a median follow-up of 6.3 months, the 6-month progression-free and overall survival were 59.9% and 91.1%, respectively. Rates of cytokine-release syndrome (CRS) and neurotoxicity (ICANS) of any grade were 9.7% and 9.7%, respectively, with no grade ≥ 3 events. No infections were reported during the first 30 days following liso-cel infusion. Neutropenia ≥ grade 3 was observed in 29.0% of patients; thrombocytopenia ≥ grade 3 occurred in 9.7%. In conclusion, bendamustine lymphodepletion before liso-cel appears to be a strategy that can drive tumor responses while ensuring a mild toxicity profile., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
50. CAR T-Cell Immunotherapy in Minority Patients with Lymphoma.
- Author
-
Ghilardi G, Williamson S, Pajarillo R, Paruzzo L, Chen L, Grady C, Doucette A, Nemecek E, Gabrielli G, Barta SK, Svoboda J, Chong EA, Yelton R, Nasta SD, Landsburg DJ, Ugwuanyi O, Chen AI, Schachter L, White G, Ballard HJ, Weber E, Curley C, Porter DL, Garfall AL, Hwang WT, Guerra CE, Maziarz RT, Schuster SJ, and Ruella M
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Minority Groups statistics & numerical data, Receptors, Chimeric Antigen immunology, Antigens, CD19 immunology, Antigens, CD19 therapeutic use, Immunotherapy, Adoptive adverse effects
- Abstract
Background: Administration of anti-CD19 chimeric antigen receptor T-cell (CART19) immunotherapy for large B-cell lymphomas (LBCLs), a subset of non-Hodgkin lymphoma (NHL), involves high costs and access to specialized tertiary care centers. We investigated whether minority health populations (MHPs) have equal access to CART19 and whether their outcomes are similar to those of non-MHPs., Methods: We analyzed the prevalence and clinical outcomes of patients treated with commercial CART19 at two geographically and socioeconomically different institutions: the Abramson Cancer Center (ACC, Philadelphia, Pennsylvania) and the Knight Cancer Institute (KCI, Portland, Oregon)., Results: In the ACC catchment area, 8956 patients were diagnosed with NHL between 2015 and 2019 (latest available data from the state registry), including 17.9% MHPs. In the ACC, between 2018 and 2022 (CART became available in 2018), 1492 patients with LBCL were treated, and 194 received CART19. The proportion of MHPs was 15.7% for the entire LBCL cohort but only 6.7% for the CART19 cohort. During the same time, in the KCI catchment area, 4568 patients were diagnosed with NHL, including 4.2% MHPs. In the KCI, 396 patients with LBCL were treated, and 47 received CART19. The proportion of MHPs was 6.6% for the entire LBCL cohort and 4.2% for the CART19 cohort. The 3-month response, survival, and toxicities after CART19 infusion showed similar results, although the number of patients who were treated was limited., Conclusions: This study shows that the access of MHPs to tertiary centers for LBCL care was preserved but appeared reduced for commercial CART19 immunotherapy. Although clinical outcomes of MHPs seemed similar to those of non-MHPs, the small sample size precludes drawing firm conclusions. Further studies are needed. (Funded by the Laffey McHugh Foundation and others.).
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.