91 results on '"Itzhaki O"'
Search Results
2. Macrophage-Recognized Molecules of Apoptotic Cells are Expressed at Higher Levels in AKR Lymphoma of Aged as Compared to Young Mice
- Author
-
Itzhaki, O., Skutelsky, E., Kaptzan, T., Siegal, A., Michowitz, M., Sinai, J., Huszar, M., Nafar, S., Leibovici, J., Back, Nathan, editor, Cohen, Irun R., editor, Kritchevsky, David, editor, Lajtha, Abel, editor, Paoletti, Rodolfo, editor, Keisari, Yona, editor, and Ofek, Itzhak, editor
- Published
- 2002
- Full Text
- View/download PDF
3. Self-care behaviors of patients with implanted left-ventricular assist device in Israel: a comparative study before and during COVID-19 pandemic
- Author
-
Melnikov, S, primary, Jaarsma, T, additional, Yaari, V, additional, Abuhazira, M, additional, Ben Avraham, B, additional, Itzhaki, O, additional, Shaul, A, additional, and Ben Gal, T, additional
- Published
- 2022
- Full Text
- View/download PDF
4. P1108: HIGH COMPLETE RESPONSE RATE FOLLOWING POINT-OF-CARE ANTI CD19 CAR T-CELL THERAPY IN PATIENTS WITH RELAPSED/REFRACTORY FOLLICULAR LYMPHOMA
- Author
-
Fried, S., primary, Besser, M. J., additional, Shkury, E., additional, Yerushalmi, R., additional, Shem-Tov, N., additional, Danylesko, I., additional, Jacoby, E., additional, Itzhaki, O., additional, Shouval, R., additional, Kedmi, M., additional, Shimoni, A., additional, Nagler, A., additional, and Avigdor, A., additional
- Published
- 2022
- Full Text
- View/download PDF
5. Epigenetic Profiling and Response to CD19 Chimeric Antigen Receptor T-Cell Therapy in B-Cell Malignancies
- Author
-
Garcia-Prieto, C. A., Villanueva, L., Bueno-Costa, A., Davalos, V., Gonzalez-Navarro, E. A., Juan, M., Urbano-Ispizua, A., Delgado, J., Ortiz-Maldonado, V., Del Bufalo, F., Locatelli, Franco, Quintarelli, C., Sinibaldi, M., Soler, M., Castro De Moura, M., Ferrer, G., Urdinguio, R. G., Fernandez, A. F., Fraga, M. F., Bar, D., Meir, A., Itzhaki, O., Besser, M. J., Avigdor, A., Jacoby, E., Esteller, M., Locatelli F. (ORCID:0000-0002-7976-3654), Garcia-Prieto, C. A., Villanueva, L., Bueno-Costa, A., Davalos, V., Gonzalez-Navarro, E. A., Juan, M., Urbano-Ispizua, A., Delgado, J., Ortiz-Maldonado, V., Del Bufalo, F., Locatelli, Franco, Quintarelli, C., Sinibaldi, M., Soler, M., Castro De Moura, M., Ferrer, G., Urdinguio, R. G., Fernandez, A. F., Fraga, M. F., Bar, D., Meir, A., Itzhaki, O., Besser, M. J., Avigdor, A., Jacoby, E., Esteller, M., and Locatelli F. (ORCID:0000-0002-7976-3654)
- Abstract
Background: Chimeric antigen receptor (CAR) T cells directed against CD19 (CART19) are effective in B-cell malignancies, but little is known about the molecular factors predicting clinical outcome of CART19 therapy. The increasingly recognized relevance of epigenetic changes in cancer immunology prompted us to determine the impact of the DNA methylation profiles of CART19 cells on the clinical course. Methods: We recruited 114 patients with B-cell malignancies, comprising 77 patients with acute lymphoblastic leukemia and 37 patients with non-Hodgkin lymphoma who were treated with CART19 cells. Using a comprehensive DNA methylation microarray, we determined the epigenomic changes that occur in the patient T cells upon transduction of the CAR vector. The effects of the identified DNA methylation sites on clinical response, cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, event-free survival, and overall survival were assessed. All statistical tests were 2-sided. Results: We identified 984 genomic sites with differential DNA methylation between CAR-untransduced and CAR-transduced T cells before infusion into the patient. Eighteen of these distinct epigenetic loci were associated with complete response (CR), adjusting by multiple testing. Using the sites linked to CR, an epigenetic signature, referred to hereafter as the EPICART signature, was established in the initial discovery cohort (n = 79), which was associated with CR (Fisher exact test, P <. 001) and enhanced event-free survival (hazard ratio [HR] = 0.36; 95% confidence interval [CI] = 0.19 to 0.70; P =. 002; log-rank P =. 003) and overall survival (HR = 0.45; 95% CI = 0.20 to 0.99; P =. 047; log-rank P =. 04;). Most important, the EPICART profile maintained its clinical course predictive value in the validation cohort (n = 35), where it was associated with CR (Fisher exact test, P <. 001) and enhanced overall survival (HR = 0.31; 95% CI = 0.11 to 0.84; P =. 02; log-rank P
- Published
- 2022
6. Adoptive transfer of short term cultured tumor infiltrating lymphocytes (young TIL) in metastatic melanoma patients
- Author
-
Shapira-Frommer, R., Besser, M., Ami, E. B., Zippel, D., Apter, S., Markel, G., Treves, A., Shimoni, A., Yrushalmi, R., Itzhaki, O., Nun, A. B., Nave, R., Kutchuk, I., Nagler, A., and Schachter, J.
- Published
- 2012
- Full Text
- View/download PDF
7. BASELINE CLINICAL AND PET-CT TUMOR BURDEN PARAMETERS DO NOT PREDICT OUTCOME OF RELAPSE/REFRACTORY AGGRESSIVE B CELL LYMPHOMA PATIENTS TREATED WITH ANTI-CD19 CAR T-CELLS
- Author
-
Avigdor, A., primary, Davidson, T., additional, Shimoni, A., additional, Yerushalmi, R., additional, Shem-Tov, N., additional, Danylesko, I., additional, Itzhaki, O., additional, Toren, A., additional, Jacoby, E., additional, Besser, M., additional, and Nagler, A., additional
- Published
- 2019
- Full Text
- View/download PDF
8. POINT‐OF‐CARE ANTI‐BCMA CAR T‐CELL THERAPY INDUCES ENCOURAGING RESPONSE RATES IN HIGH‐RISK RELAPSED/REFRACTORY MULTIPLE MYELOMA.
- Author
-
Magen, H., Fried, S., Itzhaki, O., Shem‐Tov, N., Danylesko, I., Yerushalmi, R., Marcus, R., Shouval, R., Nevo, L., Shapira‐Frommer, R., Nagler, A., Shimoni, A., Shkury, E., and Avigdor, A.
- Subjects
MULTIPLE myeloma ,T cells ,CHIMERIC antigen receptors ,CYTOKINE release syndrome ,POINT-of-care testing - Abstract
B Introduction: b Anti-BCMA chimeric antigen receptor (CAR) T-cell therapy showed excellent efficacy in patients with relapsed/refractory multiple myeloma (R/R MM). We report outcomes of phase 1b/2 single-center clinical trial of autologous POC anti-BCMA CAR T-cell therapy in patients (pts) with R/R MM treated with >=3 prior therapies (NCT05243212). POINT-OF-CARE ANTI-BCMA CAR T-CELL THERAPY INDUCES ENCOURAGING RESPONSE RATES IN HIGH-RISK RELAPSED/REFRACTORY MULTIPLE MYELOMA. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
9. Serum CEACAM1 Elevation Correlates with Melanoma Progression and Failure to Respond to Adoptive Cell Transfer Immunotherapy
- Author
-
Ortenberg, R., primary, Sapoznik, S., additional, Zippel, D., additional, Shapira-Frommer, R., additional, Itzhaki, O., additional, Kubi, A., additional, Zikich, D., additional, Besser, M. J., additional, Schachter, J., additional, and Markel, G., additional
- Published
- 2015
- Full Text
- View/download PDF
10. Macrophage-Recognized Molecules of Apoptotic Cells are Expressed at Higher Levels in AKR Lymphoma of Aged as Compared to Young Mice
- Author
-
Itzhaki, O., primary, Skutelsky, E., additional, Kaptzan, T., additional, Siegal, A., additional, Michowitz, M., additional, Sinai, J., additional, Huszar, M., additional, Nafar, S., additional, and Leibovici, J., additional
- Full Text
- View/download PDF
11. Adoptive transfer of short-term cultured tumor-infiltrating lymphocytes (young TIL) in metastatic melanoma patients.
- Author
-
Shapira-Frommer, R., primary, Besser, M., additional, Kuchuk, I., additional, Nave, R., additional, Zippel, D., additional, Treves, A., additional, Nagler, A., additional, Apter, S., additional, Shimoni, A., additional, Yerushalmi, R., additional, Ben-Ami, E., additional, Ben-Nun, A., additional, Markel, G., additional, Itzhaki, O., additional, Catane, R., additional, and Schachter, J., additional
- Published
- 2011
- Full Text
- View/download PDF
12. 537 The role of microRNA molecules in the regulation of aggressive features in melanoma
- Author
-
Greenberg, E., primary, Itzhaki, O., additional, Hershkovitz, L., additional, Nemlich, Y., additional, Ortenberg, R., additional, Besser, M.J., additional, Keisari, Y., additional, Schachter, J., additional, Shomron, N., additional, and Markel, G., additional
- Published
- 2010
- Full Text
- View/download PDF
13. 449 Inhibition of vascular-like network formation of highly aggressive melanoma
- Author
-
Itzhaki, O., primary, Greenberg, E., additional, Besser, M.J., additional, Nemlich, Y., additional, Ortenberg, R., additional, Schachter, J., additional, Treves, A., additional, and Markel, G., additional
- Published
- 2010
- Full Text
- View/download PDF
14. Elevated apoptosis in tumors of aged as compared to those of young mice is more pronounced in primary than in metastatic tumors of AKR lymphoma
- Author
-
Itzhaki, O., primary, Kaptzan, T., additional, Skutelsky, E., additional, Sinai, J., additional, Schiby, G., additional, Huszar, M., additional, and Leibovici, J., additional
- Published
- 2008
- Full Text
- View/download PDF
15. Anti-metastatic therapeutic modalities based on the aged cancer patient as a model – a suggestion
- Author
-
Leibovici, J., primary, Kaptzan, T., additional, Itzhaki, O., additional, Skutelsky, E., additional, Sinai, J., additional, Siegal, A., additional, and Michowitz, M., additional
- Published
- 2008
- Full Text
- View/download PDF
16. Preclinical evaluation of adoptive cell therapy for patients with metastatic renal cell carcinoma
- Author
-
Markel G, Cohen-Sinai T, Mj, Besser, Oved K, Itzhaki O, Seidman R, Fridman E, Aj, Treves, Keisari Y, Zohar Dotan, Ramon J, and Schachter J
17. Adoptive cell therapy for metastatic melanoma patients: clinical and pre-clinical development.
- Author
-
Schachter, J., Treves, A., Shapira-Frommer, R., Barak, D., Itzhaki, O., Hardan, I., Papa, M., Catane, R., Shalmon-Zifroni, B., and Besser, M.
- Published
- 2006
- Full Text
- View/download PDF
18. Pre-treatment Pulmonary Function Testing Has Limited Utility In B-cell Lymphoma Treated with CD19 CAR T-cells.
- Author
-
Sdayoor I, Shouval R, Fried S, Marcus R, Danylesko I, Yerushalmi R, Shem-Tov N, Itzhaki O, Jacoby E, Kedmi M, Nagler A, Shimoni A, Segel MJ, and Avigdor A
- Abstract
Pulmonary function tests (PFT) are recommended for hematopoietic cell transplantation (HCT) evaluation. However, their prognostic value in chimeric antigen receptor T-cell (CAR-T) therapy remains unclear. We assessed the predictive significance of PFTs and pulmonary comorbidity classifications, per the Hematopoietic Cell Transplantation-Comorbidity Index (HCT-CI), in B-cell lymphoma patients undergoing autologous CD19-CAR-T therapy. Single-center retrospective analysis encompassing 192 patients with relapsed/refractory (R/R) B-cell lymphoma, treated with commercial and point-of-care CD19-directed CAR-T therapy. Pre-therapy PFT were conducted, and patients were stratified into 3 HCT-CI-based pulmonary comorbidity grades, utilizing forced expiratory volume in 1 second (FEV1) and single-breath diffusing capacity for carbon monoxide (DLCO). Outcomes and toxicities were evaluated using univariate and multivariable Cox regression, logistic regression, Kaplan-Meier method, and spline models. Pulmonary comorbidity measures were not correlated with overall response rates or immune-toxicities, including cytokine release syndrome grade>2 and immune effector cell-associated neurotoxicity grade>2. Categorical FEV1, DLCO, and pulmonary comorbidity level did not correlate with overall survival (p=0.3, p=0.4, p=0.6, respectively) or progression free survival (PFS) (p=0.058, p>0.9, p=0.2, respectively). FEV1 as continuous measure was associated with reduced PFS in a multivariable model (HR 0.87 [95% CI 0.78-0.96], p=0.007). Spline modeling demonstrated a linear correlation between FEV1 and PFS. Categorical FEV1, DLCO, and pulmonary comorbidity level failed to predict therapy efficacy or toxicity. FEV1 as continuous measure was the sole PFT measure associated with PFS, independent of OS or severe toxicities., (Copyright © 2025 American Society of Hematology.)
- Published
- 2025
- Full Text
- View/download PDF
19. Point of care CD19 chimeric antigen receptor (CAR) T-cells for relapsed/refractory acute myeloid leukemia (AML) with aberrant CD19 antigen expression.
- Author
-
Danylesko I, Shem-Tov N, Yerushalmi R, Jacoby E, Toren A, Shouval R, Itzhaki O, Avigdor A, Shimoni A, and Nagler A
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Child, T-Lymphocytes immunology, T-Lymphocytes transplantation, Cytokine Release Syndrome etiology, Cytokine Release Syndrome therapy, Young Adult, Treatment Outcome, Antigens, CD19 immunology, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute genetics, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects, Receptors, Chimeric Antigen therapeutic use, Receptors, Chimeric Antigen genetics, Hematopoietic Stem Cell Transplantation
- Abstract
Relapsed/refractory (r/r) acute myeloid leukemia (AML) is associated with poor prognosis. CD19 is a B-cell marker, is aberrantly expressed in AML, mostly with t(8; 21)(q22; q22.1). Here we report the results of a phase 2 study giving point of care produced CD19 CAR T- cells for r/r AML with aberrant expression of CD19 (NCT04257175). Lymphodepletion included fludarabine and cyclophosphamide The response was evaluated by bone marrow (BM) aspiration on day 28. Six patients (5 adults and 1 child) were included. Median number of previous chemotherapy lines was 4 (range, 3-8) and four patients received CAR T-cells 8-18 months post allogeneic hematopoietic stem cell transplantation (allo-HSCT). Cytokine release syndrome (CRS) of any grade occurred in all patients, and 1 patient had grade 3 CRS. Immune effector cell-associated neurotoxicity syndrome (ICANS) occurred in 2 patients at low grades. Tocilizumab was administered to 2 patients and corticosteroids to 3 patients. Four patients achieved a complete remission (CR), while 2/6 progressed (PD). Three patients (2 with CR and 1 with PD) underwent allo-HSCT (it was the second transplant in 2) 2-5 months post CAR T-cells infusion. The median duration of response in patients achieving CR was 8.5 (range; 3-14) months. However, all patients eventually died within 5 (1-18) months. In conclusion, CD19 CAR T- cell treatment for AML is feasible and safe. However, the response is short and should be followed by allo-HSCT. Hopefully, future long term results will be improved by combining the CAR T- cell therapy with the emerging novel effective anti-leukemic compounds., Competing Interests: Declaration of competing interest The authors declare that they have no relevant conflict of interest and no competing financial interests., (Copyright © 2024. Published by Elsevier Masson SAS.)
- Published
- 2024
- Full Text
- View/download PDF
20. CD-19 CAR-T cells for polyrefractory rheumatoid arthritis.
- Author
-
Lidar M, Rimar D, David P, Jacoby E, Shapira-Frommer R, Itzhaki O, Slobodin G, Stern MD, Eshed I, Sanalla T, Marcus R, Shimoni A, Yerushalmi R, Shem Tov N, Danylesko I, and Avigdor A
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2024
- Full Text
- View/download PDF
21. Anti-CD19 chimeric antigen receptor T-cell therapy has less efficacy in Richter transformation than in <I>de novo</I> large B-cell lymphoma and transformed low-grade B-cell lymphoma.
- Author
-
Benjamini O, Fried S, Shouval R, Flynn JR, Beyar-Katz O, Leslie LA, Zucherman T, Yerushalmi R, Shem-Tov N, Palomba ML, Danylesko I, Sdayoor I, Malka H, Itzhaki O, Suh H, Devlin SM, Marcus R, Dahi PB, Jacoby E, Shah GL, Sauter CS, Ip A, Perales MA, Nagler A, Shimoni A, Scordo M, and Avigdor A
- Subjects
- Humans, Male, Middle Aged, Female, Aged, Retrospective Studies, Adult, Treatment Outcome, Aged, 80 and over, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Leukemia, Lymphocytic, Chronic, B-Cell mortality, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Neoplasm Grading, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Lymphoma, Large B-Cell, Diffuse therapy, Lymphoma, Large B-Cell, Diffuse immunology, Lymphoma, Large B-Cell, Diffuse mortality, Antigens, CD19 immunology, Receptors, Chimeric Antigen immunology
- Abstract
The activity of anti-CD19 chimerci antigen receptor (CAR) T-cell therapy in chronic lymphocytic leukemia (CLL) with Richter's transformation (RT) to aggressive large B-cell lymphoma (LBCL) is largely unknown. In a multicenter retrospective study, we report the safety and efficacy of CAR T-cell therapy in patients with RT (N=30) compared to patients with aggressive B-cell lymphoma (N=283) and patients with transformed indolent non-Hodgkin lymphoma (iNHL) (N=141) between April 2016 and January 2023. Two-thirds of patients received prior therapy for CLL before RT and 89% of them received B-cell receptor and B-cell lymphoma 2 inhibitors. Toxicities of CAR T-cell therapy in RT were similar to other lymphomas, with no fatalities related to cytokine release syndrome or immune effector-cell associated neurotoxicity synderome. The 100-day overall response rate and complete response rates in patients with RT were 57% and 47%, respectively. With a median follow-up of 19 months, the median overall survival (OS) was 9.9 months in patients with RT compared to 18 months in de novo LBCL and not reached in patients with transformed iNHL. The OS at 12 months was 45% in patients with RT compared with 62% and 75% in patients with de novo LBCL and transformed iNHL, respectively. In a multivariate analysis, worse OS was associated with RT histology, elevated lactate dehydrogenase, and more prior lines of therapy. CAR T-cell therapy can salvage a proportion of patients with CLL and RT exposed to prior targeted agents; however, efficacy in RT is inferior compared to de novo LBCL and transformed iNHL.
- Published
- 2024
- Full Text
- View/download PDF
22. Improved CAR-T cell activity associated with increased mitochondrial function primed by galactose.
- Author
-
Gross G, Alkadieri S, Meir A, Itzhaki O, Aharoni-Tevet Y, Ben Yosef S, Zenab A, Shbiro L, Toren A, Yardeni T, and Jacoby E
- Subjects
- Animals, Mice, Humans, Receptors, Chimeric Antigen metabolism, Receptors, Chimeric Antigen immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Antigens, CD19 metabolism, T-Lymphocytes metabolism, T-Lymphocytes immunology, Xenograft Model Antitumor Assays, Galactose metabolism, Mitochondria metabolism, Immunotherapy, Adoptive methods
- Abstract
CD19 CAR-T cells have led to durable remissions in patients with refractory B-cell malignancies; nevertheless, most patients eventually relapse in the long term. Many interventions aimed at improving current products have been reported, with a subset of them focusing on a direct or indirect link to the metabolic state of the CAR-T cells. We assessed clinical products from an ongoing clinical trial utilizing CD19-28z CAR-T cells from patients with acute lymphoblastic leukemia. CAR-T clinical products leading to a complete response had significantly higher mitochondrial function (by oxygen consumption rate) irrespective of mitochondrial content. Next, we replaced the carbon source of the media from glucose to galactose to impact cellular metabolism. Galactose-containing media increased mitochondrial activity in CAR-T cells, and improved in in-vitro efficacy, without any consistent phenotypic change in memory profile. Finally, CAR-T cells produced in galactose-based glucose-free media resulted in increased mitochondrial activity. Using an in-vivo model of Nalm6 injected mice, galactose-primed CAR-T cells significantly improved leukemia-free survival compared to standard glucose-cultured CAR-T cells. Our results prove the significance of mitochondrial metabolism on CAR-T cell efficacy and suggest a translational pathway to improve clinical products., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
23. Genetic Modification of Tumor-Infiltrating Lymphocytes, Peripheral T Cells, and T-Cell Model Cell Lines.
- Author
-
Weinstein-Marom H, Blokon-Kogan D, Levi-Mann M, Katzman C, Shalev S, Zaitsev M, Besser MJ, Shapira-Frommer R, Gross G, Itzhaki O, and Nissim L
- Subjects
- Transfection, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Cell Line, CD8-Positive T-Lymphocytes, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating metabolism, T-Lymphocytes metabolism
- Abstract
Genetic modification of tumor-infiltrating lymphocytes (TILs) or circulating T cells has become an important avenue in cancer therapy. Here we describe a comprehensive method for establishing and expanding TIL cultures and genetically modifying them with a gene of interest (GOI) via retroviral transduction or mRNA transfection. The method includes all the important steps starting with TIL extraction from tumors through to the maintenance of the genetically modified TILs. The protocol includes instructions for retroviral transduction and mRNA transfection of circulating T cells or T-cell lines. The GOIs most commonly introduced into the target cells are chimeric antigen receptors (CARs); genetic adjuvants, such as membrane-bound interleukins; and antitumor T-cell receptors (TCRs)., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
24. Point-of-care anti-CD19 chimeric antigen receptor T-cell therapy for relapsed/refractory follicular lymphoma.
- Author
-
Fried S, Shkury E, Itzhaki O, Sdayoor I, Yerushalmi R, Shem-Tov N, Danylesko I, Jacoby E, Shouval R, Kedmi M, Marcus R, Nagler A, Shimoni A, and Avigdor A
- Subjects
- Humans, Point-of-Care Systems, Immunotherapy, Adoptive adverse effects, Cell- and Tissue-Based Therapy, Antigens, CD19, Receptors, Chimeric Antigen, Lymphoma, Follicular therapy, Lymphoma, Follicular drug therapy
- Abstract
Patients with relapsed/refractory follicular lymphoma (R/R-FL) often require multiple treatment lines. We performed a phase 1b/2 single-center clinical trial of autologous point-of-care anti-CD19 chimeric antigen receptor (CAR) T-cells in R/R-FL patients treated patients with ≥ 2 treatment lines. All 26 patients enrolled received CAR T-cell infusion at a median of 11 days after leukapheresis. Seventy-seven percent of patients had POD24. At enrollment, disease stage was III-IV in 85% of the patients, 77% had high-risk FLIPI score, and 77% had progressive disease. Grade III-IV cytokine release and immune effector cell-associated neurotoxicity syndromes occurred in 12% and 16% of the patients, respectively. Overall response rate at 1-month was 88%. The median follow-up was 15.4 months. One-year overall and progression-free survival were 100% and 63%, respectively. In conclusion, point-of-care CAR T-cell, manufactured within 11 days, induced a high response rate with an acceptable safety profile in patients with high-risk R/R-FL.
- Published
- 2023
- Full Text
- View/download PDF
25. Improved CAR-T cell activity associated with increased mitochondrial function primed by galactose.
- Author
-
Gross G, Alkadieri S, Meir A, Itzhaki O, Aharony-Tevet Y, Yosef SB, Zenab A, Shbiro L, Toren A, Yardeni T, and Jacoby E
- Abstract
CD19 CAR-T cells have led to durable remissions in patients with refractory B-cell malignancies; nevertheless, most patients eventually relapse in the long term. Many interventions aimed at improving current products have been reported, with a subset of them focusing on a direct or indirect link to the metabolic state of the CAR-T cells. We assessed clinical products from an ongoing clinical trial utilizing CD19-28z CAR-T cells from patients with acute lymphoblastic leukemia. CAR-T clinical products leading to a complete response had significantly higher mitochondrial function (by oxygen consumption rate) irrespective of mitochondrial content. Next, we replaced the carbon source of the media from glucose to galactose to impact cellular metabolism. Galactose-containing media increased mitochondrial activity in CAR-T cells, and improved in vitro efficacy, without any consistent phenotypic change in memory profile. Finally, CAR-T cells produced in galactose-based glucose-free media resulted in increased mitochondrial activity. Using an in vivo model of Nalm6 injected mice, galactose-primed CAR-T cells significantly improved leukemia-free survival compared to standard glucose-cultured CAR-T cells. Our results prove the significance of mitochondrial metabolism on CAR-T cell efficacy and suggest a translational pathway to improve clinical products., Competing Interests: Competing Interests statement: The authors report no financial conflict of interests. This work was funded by the Dotan center for hematologic malignancies grant (EJ) and NIH grant 5R01CA259635 (TY).
- Published
- 2023
- Full Text
- View/download PDF
26. MYC Induces Immunotherapy and IFNγ Resistance Through Downregulation of JAK2.
- Author
-
Markovits E, Harush O, Baruch EN, Shulman ED, Debby A, Itzhaki O, Anafi L, Danilevsky A, Shomron N, Ben-Betzalel G, Asher N, Shapira-Frommer R, Schachter J, Barshack I, Geiger T, Elkon R, Besser MJ, and Markel G
- Subjects
- Humans, Down-Regulation, Immunotherapy, T-Lymphocytes pathology, Interferon-gamma genetics, Janus Kinase 2 genetics, Melanoma genetics, Melanoma therapy, Melanoma pathology
- Abstract
Immunotherapy has revolutionized the treatment of advanced melanoma. Because the pathways mediating resistance to immunotherapy are largely unknown, we conducted transcriptome profiling of preimmunotherapy tumor biopsies from patients with melanoma that received PD-1 blockade or adoptive cell therapy with tumor-infiltrating lymphocytes. We identified two melanoma-intrinsic, mutually exclusive gene programs, which were controlled by IFNγ and MYC, and the association with immunotherapy outcome. MYC-overexpressing melanoma cells exhibited lower IFNγ responsiveness, which was linked with JAK2 downregulation. Luciferase activity assays, under the control of JAK2 promoter, demonstrated reduced activity in MYC-overexpressing cells, which was partly reversible upon mutagenesis of a MYC E-box binding site in the JAK2 promoter. Moreover, silencing of MYC or its cofactor MAX with siRNA increased JAK2 expression and IFNγ responsiveness of melanomas, while concomitantly enhancing the effector functions of T cells coincubated with MYC-overexpressing cells. Thus, we propose that MYC plays a pivotal role in immunotherapy resistance through downregulation of JAK2., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
27. Point-of-care CAR T-cell therapy as salvage strategy for out-of-specification tisagenlecleucel.
- Author
-
Fried S, Shouval R, Varda-Bloom N, Besser MJ, Yerushalmi R, Shem-Tov N, Danylesko I, Jacoby E, Teihman S, Itzhaki O, Fein JA, Kedmi M, Shimoni A, Nagler A, and Avigdor A
- Subjects
- Adult, Humans, Antigens, CD19, Neoplasm Recurrence, Local etiology, Point-of-Care Systems, Receptors, Antigen, T-Cell, Immunotherapy, Adoptive, Lymphoma, Large B-Cell, Diffuse pathology
- Abstract
Tisagenlecleucel (tisa-cel) is an anti-CD19 chimeric antigen receptor (CAR) T-cell therapy approved for patients with relapsed/refractory large B-cell lymphoma. Outcomes of patients with out-of-commercial specification (OOS) CAR T products are not well characterized. We therefore assessed 37 adult patients who underwent leukapheresis for tisa-cel therapy in a single center. In nine (24%) patients, manufactured tisa-cel was considered OOS. Three of them (33%) received tisa-cel after institutional review board approval; 2/9 (22%) did not receive tisa-cel due to disease progression; and 4/9 (44%) received academic point-of-care (POC) CAR T-cell as salvage therapy, at a median of 35 days following OOS notification. Three of those four patients achieved a complete response. In univariate analysis, risk factors for OOS were ≥ 4 prior therapies or previous bendamustine exposure. In conclusion, we report high OOS incidence of 24% in real-life setting. Forty-four percent of those patients received POC CAR T-cell as salvage therapy.
- Published
- 2022
- Full Text
- View/download PDF
28. Impact of cryopreservation on CAR T production and clinical response.
- Author
-
Brezinger-Dayan K, Itzhaki O, Melnichenko J, Kubi A, Zeltzer LA, Jacoby E, Avigdor A, Shapira Frommer R, and Besser MJ
- Abstract
Adoptive cell therapy with chimeric antigen receptor (CAR) T cells has become an efficient treatment option for patients with hematological malignancies. FDA approved CAR T products are manufactured in centralized facilities from fresh or frozen leukapheresis and the cryopreserved CAR T infusion product is shipped back to the patient. An increasing number of clinical centers produce CAR T cells on-site, which enables the use of fresh and cryopreserved PBMCs and CAR T cells. Here we determined the effect of cryopreservation on PBMCs and CD19 CAR T cells in a cohort of 118 patients treated with fresh CAR T cells and in several patients head-to-head. Cryopreserved PBMCs, obtained from leukapheresis products, contained less erythrocytes and T cells, but were sufficient to produce CAR T cells for therapy. There was no correlation between the recovery of PBMCs and the transduction efficacy, the number of CAR T cells obtained by the end of the manufacturing process, the in vitro reactivity, or the response rate to CAR T therapy. We could show that CAR T cells cryopreserved during the manufacturing process, stored and resumed expansion at a later time point, yielded sufficient cell numbers for treatment and led to complete remissions. Phenotype analysis including T cell subtypes, chemokine receptor and co-inhibitory/stimulatory molecules, revealed that fresh CAR T cells expressed significantly more TIM-3 and contained less effector T cells in comparison to their frozen counterparts. In addition, fresh CAR T infusion products demonstrated increased in vitro anti-tumor reactivity, however cryopreserved CAR T cells still showed high anti-tumor potency and specificity. The recovery of cryopreserved CAR T cells was similar in responding and non-responding patients. Although fresh CAR T infusion products exhibit higher anti-tumor reactivity, the use of frozen PBMCs as staring material and frozen CAR T infusion products seems a viable option, as frozen products still exhibit high in vitro potency and cryopreservation did not seem to affect the clinical outcome., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Brezinger-Dayan, Itzhaki, Melnichenko, Kubi, Zeltzer, Jacoby, Avigdor, Shapira Frommer and Besser.)
- Published
- 2022
- Full Text
- View/download PDF
29. Intravenous Iron Supplementation for the Treatment of Chemotherapy-Induced Anemia: A Systematic Review and Meta-Analysis of Randomized Controlled Trials.
- Author
-
Buchrits S, Itzhaki O, Avni T, Raanani P, and Gafter-Gvili A
- Abstract
Background: The pathophysiology of cancer-related anemia is multifactorial, including that of chemotherapy-induced anemia (CIA). The guidelines are not consistent in their approach to the use of intravenous (IV) iron in patients with cancer as part of the clinical practice. Materials and methods: All randomized controlled trials that compared IV iron with either no iron or iron taken orally for the treatment of CIA were included. We excluded trials if erythropoiesis-stimulating agents (ESAs) were used. The primary outcome was the percentage of patients requiring a red blood cell (RBC) transfusion during the study period. The secondary outcomes included the hematopoietic response (an increase in the Hb level by more than 1 g/dL or an increase above 11 g/dL), the iron parameters and adverse events. For the dichotomous data, risk ratios (RRs) with 95% confidence intervals (Cis) were estimated and pooled. For the continuous data, the mean differences were calculated. A fixed effect model was used, except in the event of significant heterogeneity between the trials (p < 0.10; I2 > 40%), in which we used a random effects model. Results: A total of 8 trials published between January 1990 and July 2021 that randomized 1015 patients fulfilled the inclusion criteria. Of these, 553 patients were randomized to IV iron and were compared with 271 patients randomized to oral iron and 191 to no iron. IV iron decreased the percentage of patients requiring a blood transfusion compared with oral iron (RR 0.72; 95% CI 0.55−0.95) with a number needed to treat of 20 (95% CI 11−100). IV iron increased the hematopoietic response (RR 1.23; 95% CI 1.01−1.5). There was no difference with respect to the risk of adverse events (RR 0.97; 95% CI 0.88−1.07; 8 trials) or severe adverse events (RR 1.09; 95% CI 0.76−1.57; 8 trials). Conclusions: IV iron resulted in a decrease in the need for RBC transfusions, with no difference in adverse events in patients with CIA. IV iron for the treatment of CIA should be considered in clinical practice.
- Published
- 2022
- Full Text
- View/download PDF
30. Parameters of long-term response with CD28-based CD19 chimaeric antigen receptor-modified T cells in children and young adults with B-acute lymphoblastic leukaemia.
- Author
-
Jacoby E, Bielorai B, Hutt D, Itzhaki O, Adam E, Bar D, Besser MJ, and Toren A
- Subjects
- Acute Disease, Antigens, CD19, CD28 Antigens, Child, Humans, Immunotherapy, Adoptive methods, Neoplasm Recurrence, Local therapy, T-Lymphocytes, Young Adult, Lymphoma, B-Cell, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Chimeric Antigen
- Abstract
CD28-based CD19 chimaeric antigen receptor-modified (CAR-)Tcells were recently FDA-approved for adult acute lymphoblastic leukaemia (ALL). We report long-term outcome of 37 children and young adults treated with autologous CD19 CAR-T cells. The complete remission rate was 86%, of which 71% were polymerase chain reaction (PCR) minimal residual disease (MRD)-negative, 14% were MRD-negative by flow cytometry, and 14% were PCR MRD-positive. 26 patients proceeded to subsequent haematopoietic stem cell transplant (HSCT). 11 patients had a CD19-postive relapse (eight post HSCT and three without) and one had a CD19-negative relapse. All relapse events occurred within two years from cell therapy. With a median follow-up of three years, the median event-free survival (EFS) is 17 months and the median overall survival (OS) is not reached. The three-year EFS is 41% and OS is 56%. Patients with >5% blasts in the bone marrow prior to lymphodepletion had an inferior EFS. All patients with a PCR MRD-positive result at day 28 had relapsed after CAR-T-cell therapy. A prior HSCT did not significantly affect outcome, but a consolidative transplant after achieving remission improved long-term results. Overall, prelymphodepletion disease burden and molecular MRD negativity following CAR-T cells are predictors of long-term outcome following CD19 CAR-T-cell therapy for ALL., (© 2022 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
31. Molecular and Functional Signatures Associated with CAR T Cell Exhaustion and Impaired Clinical Response in Patients with B Cell Malignancies.
- Author
-
Beider K, Itzhaki O, Schachter J, Grushchenko-Polaq AH, Voevoda-Dimenshtein V, Rosenberg E, Ostrovsky O, Devillers O, Shapira Frommer R, Zeltzer LA, Toren A, Jacoby E, Shimoni A, Avigdor A, Nagler A, and Besser MJ
- Subjects
- Antigens, CD19, B-Lymphocytes, Humans, Immunotherapy, Adoptive, Leukemia, B-Cell genetics, Leukemia, B-Cell therapy, Receptors, Chimeric Antigen
- Abstract
Despite the high rates of complete remission following chimeric antigen receptor (CAR) T cell therapy, its full capacity is currently limited by the generation of dysfunctional CAR T cells. Senescent or exhausted CAR T cells possess poor targeting and effector functions, as well as impaired cell proliferation and persistence in vivo. Strategies to detect, prevent or reverse T cell exhaustion are therefore required in order to enhance the effectiveness of CAR T immunotherapy. Here we report that CD19 CAR T cells from non-responding patients with B cell malignancies show enrichment of CD8
+ cells with exhausted/senescent phenotype and display a distinct transcriptional signature with dysregulation of genes associated with terminal exhaustion. Furthermore, CAR T cells from non-responding patients exhibit reduced proliferative capacity and decreased IL-2 production in vitro, indicating functional impairment. Overall, our work reveals potential mediators of resistance, paving the way to studies that will enhance the efficacy and durability of CAR T therapy in B cell malignancies.- Published
- 2022
- Full Text
- View/download PDF
32. Epigenetic Profiling and Response to CD19 Chimeric Antigen Receptor T-Cell Therapy in B-Cell Malignancies.
- Author
-
Garcia-Prieto CA, Villanueva L, Bueno-Costa A, Davalos V, González-Navarro EA, Juan M, Urbano-Ispizua Á, Delgado J, Ortiz-Maldonado V, Del Bufalo F, Locatelli F, Quintarelli C, Sinibaldi M, Soler M, Castro de Moura M, Ferrer G, Urdinguio RG, Fernandez AF, Fraga MF, Bar D, Meir A, Itzhaki O, Besser MJ, Avigdor A, Jacoby E, and Esteller M
- Subjects
- Antigens, CD19, Cell- and Tissue-Based Therapy, Epigenesis, Genetic, Humans, Immunotherapy, Adoptive adverse effects, Receptors, Antigen, T-Cell genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Chimeric Antigen
- Abstract
Background: Chimeric antigen receptor (CAR) T cells directed against CD19 (CART19) are effective in B-cell malignancies, but little is known about the molecular factors predicting clinical outcome of CART19 therapy. The increasingly recognized relevance of epigenetic changes in cancer immunology prompted us to determine the impact of the DNA methylation profiles of CART19 cells on the clinical course., Methods: We recruited 114 patients with B-cell malignancies, comprising 77 patients with acute lymphoblastic leukemia and 37 patients with non-Hodgkin lymphoma who were treated with CART19 cells. Using a comprehensive DNA methylation microarray, we determined the epigenomic changes that occur in the patient T cells upon transduction of the CAR vector. The effects of the identified DNA methylation sites on clinical response, cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, event-free survival, and overall survival were assessed. All statistical tests were 2-sided., Results: We identified 984 genomic sites with differential DNA methylation between CAR-untransduced and CAR-transduced T cells before infusion into the patient. Eighteen of these distinct epigenetic loci were associated with complete response (CR), adjusting by multiple testing. Using the sites linked to CR, an epigenetic signature, referred to hereafter as the EPICART signature, was established in the initial discovery cohort (n = 79), which was associated with CR (Fisher exact test, P < .001) and enhanced event-free survival (hazard ratio [HR] = 0.36; 95% confidence interval [CI] = 0.19 to 0.70; P = .002; log-rank P = .003) and overall survival (HR = 0.45; 95% CI = 0.20 to 0.99; P = .047; log-rank P = .04;). Most important, the EPICART profile maintained its clinical course predictive value in the validation cohort (n = 35), where it was associated with CR (Fisher exact test, P < .001) and enhanced overall survival (HR = 0.31; 95% CI = 0.11 to 0.84; P = .02; log-rank P = .02)., Conclusions: We show that the DNA methylation landscape of patient CART19 cells influences the efficacy of the cellular immunotherapy treatment in patients with B-cell malignancy., (© The Author(s) 2021. Published by Oxford University Press.)
- Published
- 2022
- Full Text
- View/download PDF
33. Post-transplantation erythrocytosis in kidney transplant recipients-A retrospective cohort study.
- Author
-
Hofstetter L, Rozen-Zvi B, Schechter A, Raanani P, Itzhaki O, Rahamimov R, and Gafter-Gvili A
- Subjects
- Adult, Female, Glomerular Filtration Rate, Graft Rejection, Humans, Kidney physiopathology, Male, Middle Aged, Retrospective Studies, Treatment Outcome, Kidney Transplantation adverse effects, Polycythemia etiology
- Abstract
Objectives: To characterize risk factors for the development of post-transplant erythrocytosis (PTE), and its long-term effect on mortality, graft failure, and thrombosis., Methods: Retrospective study including all kidney transplant recipients in Rabin Medical Center (RMC) during the years 2005-2014. The primary outcome was a composite outcome of all-cause mortality or graft failure at the end of follow-up. Secondary outcomes included death censored graft loss, venous thromboembolism, major adverse cardiovascular events, and mortality. A matched control group was also evaluated. Univariate and multivariate time-varying Cox model analyses were conducted for outcome evaluation., Results: A total of 1304 patients were included, 169 of whom were diagnosed with PTE (12.9%). PTE was associated with male gender, higher glomerular filtration rate (GFR), and polycystic kidney disease. PTE was found to be associated with a reduced risk of the primary outcome (HR 0.355, CI 95% 0.151-0.89, P = .027) in a univariate time-varying Cox analysis, but was not associated with the composite outcome in a multivariate analysis. There was no difference in the primary outcome when the PTE group was compared with the matched control., Conclusion: PTE was not found to be associated with long-term outcomes of graft failure and poor survival., (© 2021 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
34. microRNA expression patterns in tumor infiltrating lymphocytes are strongly associated with response to adoptive cell transfer therapy.
- Author
-
Galore-Haskel G, Greenberg E, Yahav I, Markovits E, Ortenberg R, Shapira-Fromer R, Itzhaki O, Schachter J, Besser MJ, and Markel G
- Subjects
- Adult, Aged, Female, Follow-Up Studies, Humans, Male, Melanoma genetics, Melanoma immunology, Melanoma therapy, Middle Aged, Prognosis, Survival Rate, Tumor Cells, Cultured, Adoptive Transfer methods, Biomarkers, Tumor genetics, Gene Expression Regulation, Neoplastic, Lymphocytes, Tumor-Infiltrating immunology, Melanoma pathology, MicroRNAs genetics
- Abstract
Adoptive cell transfer (ACT) using autologous tumor infiltrating lymphocytes (TILs) was previously shown to yield clinical response in metastatic melanoma patients as an advanced line. Unfortunately, there is no reliable marker for predicting who will benefit from the treatment. We analyzed TIL samples from the infusion bags used for treatment of 57 metastatic melanoma patients and compared their microRNA profiles. The discovery cohort included six responding patients and seven patients with progressive disease, as defined by RECIST1.1. High throughput analysis with NanoString nCounter demonstrated significantly higher levels of miR-34a-5p and miR-22-3p among TIL from non-responders. These results were validated in TIL infusion bag samples from an independent cohort of 44 patients, using qRT-PCR of the individual microRNAs. Using classification trees, a data-driven predictive model for response was built, based on the level of expression of these microRNAs. Patients that achieved stable disease were classified with responders, setting apart the patients with progressive disease. Moreover, the expression levels of miR-34a-5p in the infused TIL created distinct survival groups, which strongly supports its role as a potential biomarker for TIL-ACT therapy. Indeed, when tested against autologous melanoma cells, miR
Low TIL cultures exhibited significantly higher cytotoxic activity than miRHigh TIL cultures, and expressed features of terminally exhausted effectors. Finally, overexpression of miR-34a-5p or miR-22-3p in TIL inhibited their cytotoxic ability in vitro. Overall, we show that a two-microRNA signature correlates with failure of TIL-ACT therapy and survival in melanoma patients.- Published
- 2021
- Full Text
- View/download PDF
35. Comparison of non-myeloablative lymphodepleting preconditioning regimens in patients undergoing adoptive T cell therapy.
- Author
-
Nissani A, Lev-Ari S, Meirson T, Jacoby E, Asher N, Ben-Betzalel G, Itzhaki O, Shapira-Frommer R, Schachter J, Markel G, and Besser MJ
- Subjects
- Adult, Clinical Trials, Phase II as Topic, Cyclophosphamide adverse effects, Cytokines blood, Female, Humans, Length of Stay, Male, Melanoma immunology, Melanoma metabolism, Melanoma secondary, Middle Aged, Myeloablative Agonists adverse effects, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Recovery of Function, Skin Neoplasms immunology, Skin Neoplasms metabolism, Skin Neoplasms pathology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Time Factors, Treatment Outcome, Vidarabine pharmacology, Cyclophosphamide therapeutic use, Immunotherapy, Adoptive adverse effects, Lymphocyte Depletion adverse effects, Melanoma therapy, Myeloablative Agonists therapeutic use, Skin Neoplasms therapy, T-Lymphocytes transplantation, Transplantation Conditioning adverse effects, Vidarabine analogs & derivatives, Whole-Body Irradiation adverse effects
- Abstract
Background: Adoptive cell therapy with T cells genetically engineered to express a chimeric antigen receptor (CAR-T) or tumor-infiltrating T lymphocytes (TIL) demonstrates impressive clinical results in patients with cancer. Lymphodepleting preconditioning prior to cell infusion is an integral part of all adoptive T cell therapies. However, to date, there is no standardization and no data comparing different non-myeloablative (NMA) regimens., Methods: In this study, we compared NMA therapies with different doses of cyclophosphamide or total body irradiation (TBI) in combination with fludarabine and evaluated bone marrow suppression and recovery, cytokine serum levels, clinical response and adverse events., Results: We demonstrate that a cumulative dose of 120 mg/kg cyclophosphamide and 125 mg/m
2 fludarabine (120Cy/125Flu) and 60Cy/125Flu preconditioning were equally efficient in achieving deep lymphopenia and neutropenia in patients with metastatic melanoma, whereas absolute lymphocyte counts (ALCs) and absolute neutrophil counts were significantly higher following 200 cGyTBI/75Flu-induced NMA. Thrombocytopenia was most profound in 120Cy/125Flu patients. 30Cy/75Flu-induced preconditioning in patients with acute lymphoblastic leukemia resulted in a minor ALC decrease, had no impact on platelet counts and did not yield deep neutropenia. Following cell infusion, 120Cy/125Flu patients with objective tumor response had significantly higher ALC and significant lower inflammatory indexes, such as neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR). Receiver-operating characteristics curve analysis 7 days after cell infusion was performed to determine the cut-offs, which distinguish between responding and non-responding patients in the 120Cy/125Flu cohort. NLR≤1.79 and PLR≤32.7 were associated with clinical response and overall survival. Cytokine serum levels did not associate with clinical response in patients with TIL. Patients in the 120Cy/125Flu cohort developed significantly more acute NMA-related adverse events, including thrombocytopenia, febrile neutropenia and cardiotoxicity, and stayed significantly longer in hospital compared with the 60Cy/125Flu and TBI/75Flu cohorts., Conclusions: Bone marrow depletion and recovery were equally affected by 120Cy/125Flu and 60Cy/125Flu preconditioning; however, toxicity and consequently duration of hospitalization were significantly lower in the 60Cy/125Flu cohort. Patients in the 30Cy/75Flu and TBI/75Flu groups rarely developed NMA-induced adverse events; however, both regimens were not efficient in achieving deep bone marrow suppression. Among the regimens, 60Cy/125Flu preconditioning seems to achieve maximum effect with minimum toxicity., Competing Interests: Competing interests: No, there are no competing interests., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
36. The significance of acute kidney injury in Clostridioides difficile infection.
- Author
-
Avni T, Hammud H, Itzhaki O, Gafter-Gvili A, Rozen-Zvi B, Ben-Zvi H, Bishara J, and Atamna A
- Subjects
- Adult, Clostridioides, Humans, Retrospective Studies, Risk Factors, Acute Kidney Injury etiology, Clostridium Infections complications, Renal Insufficiency, Chronic complications, Sepsis complications
- Abstract
Objective: Clostridioides difficile infection (CDI) may present as sepsis with acute kidney injury (AKI). Herein, we aimed to evaluate the clinical outcomes of patients with AKI complicating CDI., Methods: All consecutive adult patients hospitalized in Rabin Medical Center between 1 January 2013 and 31 December 2018 with laboratory confirmed CDI, were included in the study. Subjects were divided into two groups: patients with AKI and controls. Primary outcome was all-cause mortality at 30 days after the CDI episode. Secondary outcomes included number of patients with deteriorating renal functions at 90 days, 90-day all-cause mortality, length of hospital stay and readmission rates. A multivariable analysis adjusted for other risk factors for mortality and renal function deterioration was conducted. An analysis of subgroups based on baseline kidney function and AKI stage was also performed. Results are reported as odds ratios (OR) with 95% confidence intervals (95% CI)., Results: A total of 527 patients were included, amongst them 140 patients with AKI and 387 controls. Patients with AKI were significantly older, had more comorbidities, and more of them had chronic kidney disease (CKD) at any stage at baseline. On multivariable analysis, 30 days all-cause mortality was significantly higher in patients with AKI, OR 1.67, 95% CI 1.05-2.66. Mortality was also significantly associated with advanced age and baseline CKD. Among patients alive at 90 days, deterioration of renal function was significantly more common in patients with AKI (27/63 (42.8%) vs 22/191 (11.5%), P = .000). In a multivariable analysis, deterioration of renal function at 90 days was associated with AKI at presentation (OR 4.67, 95% CI 1.05-20.6). Early (at discharge) renal function recovery was not associated with protection from further deterioration of renal function at day 90., Conclusions: CDI patients with AKI have an increased risk of mortality and further deterioration of renal function. Early renal function recovery does not infer protection from further deterioration of renal function at 3 months. Caution and nephrology follow-up should be considered after discharge for all patients who developed AKI during CDI, regardless of discharge creatinine levels., (© 2020 John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
37. Genetic Modification of Tumor-Infiltrating Lymphocytes via Retroviral Transduction.
- Author
-
Weinstein-Marom H, Gross G, Levi M, Brayer H, Schachter J, Itzhaki O, and Besser MJ
- Subjects
- Antigens, CD19 immunology, Cell Line, Tumor, Humans, Immunotherapy, Adoptive methods, Interferon-gamma immunology, K562 Cells, Lymphocyte Activation immunology, Melanoma immunology, Receptors, Chimeric Antigen immunology, Lymphocytes, Tumor-Infiltrating immunology, Retroviridae immunology
- Abstract
Adoptive T cell therapy (ACT) holds great promise for cancer treatment. One approach, which has regained wide interest in recent years, employs antitumor T cells isolated from tumor lesions ("tumor-infiltrating lymphocytes" or TIL). It is now appreciated that a considerable proportion of anti-melanoma TIL recognize new HLA-binding peptides resulting from somatic mutations, which occurred during tumor progression. The clinical efficacy of TIL can potentially be improved via their genetic modification, designed to enhance their survival, homing capacity, resistance to suppression, tumor killing ability and additional properties of clinical relevance. Successful implementation of such gene-based strategies critically depends on efficient and reproducible protocols for gene delivery into clinical TIL preparations. Here we describe an optimized protocol for the retroviral transduction of TIL. As the experimental system we employed anti-melanoma TIL cultures prepared from four patients, recombinant retrovirus encoding an anti-CD19 chimeric antigen receptor (CAR) as a model gene of interest and CD19+ and CD19- human cell lines serving as target cells. Transduction on day 7 of the rapid expansion protocol (REP) resulted in 69 ± 8% CAR positive TIL. Transduced, but not untransduced TIL, from the four patients responded robustly to CD19+, but not CD19- cell lines, as judged by substantial secretion of IFN-γ following co-culture. In light of the rekindled interest in antitumor TIL, this protocol can be incorporated into a broad range of gene-based approaches for improving the in-vivo survival and functionality of TIL in the clinical setting., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Weinstein-Marom, Gross, Levi, Brayer, Schachter, Itzhaki and Besser.)
- Published
- 2021
- Full Text
- View/download PDF
38. Feasibility of leukapheresis for CAR T-cell production in heavily pre-treated pediatric patients.
- Author
-
Hutt D, Bielorai B, Baturov B, Z'orbinski I, Ilin N, Adam E, Itzhaki O, Besser MJ, Toren A, and Jacoby E
- Subjects
- Adolescent, Adult, Child, Feasibility Studies, Female, Humans, Male, Receptors, Chimeric Antigen, Young Adult, Leukapheresis methods
- Abstract
Background: Autologous CD19 chimeric-antigen receptor (CAR) T-cells are an effective salvage therapy for patients with relapsed or refractory B cell malignancies. The essential first step in the production is the collection of mature lymphocytes through leukapheresis. It is a challenging procedure given the fact patients are heavily pretreated and the special considerations of pediatric apheresis., Methods: We analyzed the data of leukapheresis outcome for CAR T production in a phase 1b/2 clinical trial enrolling 34 children, adolescents and young adults with relapsed or refractory B-cell malignancies., Results: All patients underwent a single leukapheresis. Given a short production time for CAR T-cells, most patients received bridging therapy prior to apheresis. Leukapheresis was performed using peripheral venous access in the majority (82%) of patients, and the remainder required arterial line or central venous access. T-cell collection efficiency (CE) was variable with a median of 18%. No apheresis-related adverse events were noted, and all procedures were successful but two: one resulting in lower than target dose (1 × 10
6 CAR + cells/kg) and the other in failure of CAR T-cell production., Conclusions: Collection of sufficient T-cells in heavily pretreated pediatric patients via a single apheresis procedure is feasible even with relatively low T-cell CE., Competing Interests: Declaration of Competing Interest DH and EJ received honoraria from Novartis. All other authors have no conflicts of interest., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
39. Gamma-Delta CAR-T Cells Show CAR-Directed and Independent Activity Against Leukemia.
- Author
-
Rozenbaum M, Meir A, Aharony Y, Itzhaki O, Schachter J, Bank I, Jacoby E, and Besser MJ
- Subjects
- Animals, Genetic Engineering methods, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Transduction, Genetic methods, Xenograft Model Antitumor Assays, Cell Culture Techniques methods, Immunotherapy, Adoptive methods, Leukemia, Receptors, Antigen, T-Cell, gamma-delta immunology, Receptors, Chimeric Antigen immunology
- Abstract
Autologous T cells engineered to express a chimeric antigen receptor (CAR) against the CD19 antigen are in the frontline of contemporary hemato-oncology therapies, leading to high remission rates in B-cell malignancies. Although effective, major obstacles involve the complex and costly individualized manufacturing process, and CD19 target antigen loss or modulation leading to resistant and relapse following CAR therapy. A potential solution for these limitations is the use of donor-derived γδT cells as a CAR backbone. γδT cells lack allogenecity and are safely used in haploidentical transplants. Moreover, γδT cells are known to mediate natural anti-tumor responses. Here, we describe a 14-day production process initiated from peripheral-blood mononuclear cells, leading to a median 185-fold expansion of γδ T cells with high purity (>98% CD3+ and >99% γδTCR+). CAR transduction efficacy of γδ T cells was equally high when compared to standard CAR-T cells (60.5 ± 13.2 and 65.3 ± 18.3%, respectively). CD19-directed γδCAR-T cells were effective against CD19+ cell lines in vitro and in vivo, showing cytokine production, direct target killing, and clearance of bone marrow leukemic cells in an NSG model. Multiple injections of γδCAR-T cells and priming of mice with zoledronate lead to enhanced tumor reduction in vivo . Unlike standard CD19 CAR-T cells, γδCAR-T cells were able to target CD19 antigen negative leukemia cells, an effect that was enhanced after priming the cells with zoledronate. In conclusion, γδCAR-T cell production is feasible and leads to highly pure and efficient effector cells. γδCAR-T cell may provide a promising platform in the allogeneic setting, and may target leukemic cells also after antigen loss., (Copyright © 2020 Rozenbaum, Meir, Aharony, Itzhaki, Schachter, Bank, Jacoby and Besser.)
- Published
- 2020
- Full Text
- View/download PDF
40. Comprehensive single institute experience with melanoma TIL: Long term clinical results, toxicity profile, and prognostic factors of response.
- Author
-
Besser MJ, Itzhaki O, Ben-Betzalel G, Zippel DB, Zikich D, Kubi A, Brezinger K, Nissani A, Levi M, Zeltzer LA, Ben-Nun A, Asher N, Shimoni A, Nagler A, Markel G, Shapira-Frommer R, and Schachter J
- Subjects
- CD8-Positive T-Lymphocytes drug effects, Cyclophosphamide administration & dosage, Cyclophosphamide adverse effects, Female, Follow-Up Studies, Humans, Interleukin-2 administration & dosage, Interleukin-2 adverse effects, Lymphocytes, Tumor-Infiltrating pathology, Male, Middle Aged, Prognosis, Progression-Free Survival, Skin Neoplasms drug therapy, Skin Neoplasms pathology, Vidarabine administration & dosage, Vidarabine adverse effects, Vidarabine analogs & derivatives, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Lymphocytes, Tumor-Infiltrating drug effects, Melanoma drug therapy, Melanoma pathology
- Abstract
Adoptive cell transfer (ACT) of tumor-infiltrating lymphocytes (TIL) mediates objective responses in 30% to 50% of patients with metastatic melanoma according to multiple, small phase 2 trials. Here we report the long-term clinical results, intent-to-treat analysis, predictors of response and toxicity profile in a large patient cohort. A total of 179 refractory melanoma patients were enrolled in the ACT trial. TIL were administered in combination with high-dose bolus interleukin-2 following preconditioning with cyclophosphamide and fludarabine. Patients were followed-up for a median of 7.2 years. A total of 107 (60%) of 179 enrolled patients were treated. The main reason for the drop out of the study was clinical deterioration. Of 103 evaluated patients, 29 patients (28%) achieved an objective response (OR), including complete remission (8%) or partial response (20%). Sixteen pateints exhibited stable disease. Predictors of response were performance status, time of TIL in culture and CD8 frequency in the infusion product. The absolute lymphocyte count 1 and 2 weeks after TIL infusion was the most predictive parameter of response. With a medium follow-up time of 7.2 years, OR patients reached a median overall survival (OS) of 58.45 months and a median progression-free survival (PFS) of 15.43 months, as compared with nonresponders, with 6.73 months OS and 2.60 months PFS. By 6 years, 50% of OR patients were alive and 43% had no documented progression. TIL ACT can yield durable objective responses, even as salvage therapy in highly advanced metastatic melanoma patients., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
41. Immune co-culture cell microarray - a feasible tool for high-throughput functional investigation of lymphocyte-cancer interactions.
- Author
-
Baruch EN, Ortenberg R, Avivi C, Anafi L, Dick-Necula D, Stossel C, Moshkovits Y, Itzhaki O, Besser MJ, Schachter J, Barshack I, and Markel G
- Subjects
- Coculture Techniques, Humans, Lymphocytes, Lymphocytes, Tumor-Infiltrating, Reproducibility of Results, Neoplasms
- Abstract
Omics analyses often result in dozens to hundreds of potential targets, requiring validation for their biological relevance. Current high-throughput functional investigation methods are frequently labor-intensive, expensive, and display low reproducibility. The Immune Co-Culture Cell Microarray (ICCM) is a formalin-fixed paraffin-embedded cell block microarray based on co-cultures of patient-derived tumor-infiltrating lymphocytes and their autologous melanoma cells. Each ICCM slide represents the same experiment and can be stained using standard immunohistochemistry and immunofluorescence techniques. Functional dynamics assessment of both proteins and microRNAs using ICCM stained slides demonstrated similar findings to flow cytometry assays and to previously published patient-derived biopsy reports., (© 2020 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2020
- Full Text
- View/download PDF
42. Head-to-head comparison of in-house produced CD19 CAR-T cell in ALL and NHL patients.
- Author
-
Itzhaki O, Jacoby E, Nissani A, Levi M, Nagler A, Kubi A, Brezinger K, Brayer H, Zeltzer LA, Rozenbaum M, Vernitsky H, Markel G, Toren A, Avigdor A, Schachter J, and Besser MJ
- Subjects
- Adolescent, Adult, Age Factors, Antigens, CD19 genetics, Antigens, CD19 metabolism, Drug Resistance, Neoplasm, Female, Humans, Immunotherapy methods, Lymphocytes, Tumor-Infiltrating immunology, Lymphoma, Non-Hodgkin immunology, Lymphoma, Non-Hodgkin pathology, Male, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Receptors, Chimeric Antigen genetics, Treatment Outcome, Young Adult, Antigens, CD19 immunology, Immunotherapy, Adoptive methods, Leukocytes, Mononuclear immunology, Lymphoma, Non-Hodgkin therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Chimeric Antigen immunology
- Abstract
Background: CD19 chimeric antigen receptor T (CAR-T) cells demonstrate remarkable remission rates in pediatric and adult patients with refractory or relapsed (r/r) acute lymphoblastic leukemia (ALL) and non-Hodgkin's lymphoma (NHL). In 2016, we initiated a clinical trial with in-house produced CD19 CAR-T cells with a CD28 co-stimulatory domain. We analyzed, for the first time, differences in production features and phenotype between ALL and NHL patients., Methods: Non-cryopreserved CAR-T cells were produced from patients' peripheral blood mononuclear cells within 9 to 10 days. 93 patients with r/r ALL and NHL were enrolled under the same study. CAR-T cells of ALL and NHL patients were produced simultaneously, allowing the head-to-head comparison., Results: All patients were heavily pretreated. Three patients dropped out from the study due to clinical deterioration (n=2) or production failure (n=1). Cells of ALL patients (n=37) expanded significantly better and contained more CAR-T cells than of NHL patients (n=53). Young age had a positive impact on the proliferation capacity. The infusion products from ALL patients contained significantly more naïve CAR-T cells and a significantly higher expression of the chemokine receptor CXCR3. PD-1, LAG-3, TIM-3, and CD28 were equally expressed. 100% of ALL patients and 94% of NHL patients received the target dose of 1×10e6 CAR-T/kg. The overall response rate was 84% (30/36) in ALL and 62% (32/52) in NHL. We further compared CAR-T cell infusion products to tumor infiltrating lymphocytes (TIL), another common type of T cell therapy, mainly clinically effective in solid tumors. CAR-T cells contained significantly more naïve T cells and central memory T cells and significantly less CCR5 compared to TIL infusion products., Conclusions: The in-house production of CAR-T cells is highly efficient and fast. Clinical response rate is high. CAR-T cells can be successfully produced for 99% of patients in just 9 to 10 days. Cells derived from ALL patients demonstrate a higher proliferation rate and contain higher frequencies of CAR-T cells and naïve T cells than of NHL patients. In addition, understanding the differences between CAR-T and TIL infusion products, may provide an angle to develop CAR-T cells for the treatment of solid tumors in the future., Trial Registration Number: ClinicalTrials.gov; CAR-T: NCT02772198, First posted: May 13, 2016; TIL: NCT00287131, First posted: February 6, 2006., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
43. Tumor-infiltrating lymphocytes from human prostate tumors reveal anti-tumor reactivity and potential for adoptive cell therapy.
- Author
-
Yunger S, Bar El A, Zeltzer LA, Fridman E, Raviv G, Laufer M, Schachter J, Markel G, Itzhaki O, and Besser MJ
- Abstract
Advanced prostate cancer remains incurable and is the second leading cause of mortality in men. Immunotherapy based on the adoptive transfer of tumor-infiltrating lymphocytes (TIL) has demonstrated promising clinical results in patients with metastatic melanoma and lately also in other solid tumors. However, the ability to obtain TIL from patients with prostate cancer, considered poorly immunogenic, remains unknown. In this study, we investigate the feasibility of isolating and expanding TIL from primary prostate tumors. We collected tumor specimens from eight patients with diagnosed prostate adenocarcinoma undergoing radical prostatectomy and were able to successfully expand multiple autologous TIL cultures from all patients. Twenty-eight prostate-TIL cultures were further expanded using a standard rapid expansion procedure under Good Manufacturing Practice conditions. TIL cultures were phenotypically characterized for T cell subset composition, differentiation status and co-inhibitory/stimulatory markers such as PD-1, TIM-3, LAG-3, and CD28 and were found to have in general similarity to TIL obtained from patients with melanoma and lung carcinoma previously treated at our center. All analyzed TIL cultures were functional as determined by the capability to produce high level of IFNγ upon stimuli. Most importantly, co-culture assays of prostate-TIL with autologous tumors demonstrated anti-tumor reactivity. In conclusion, these findings demonstrate that functional and anti-tumor reactive TIL can be obtained, despite the immunosuppressive microenvironment of the cancer, thus this study supports the development of TIL therapy for prostate cancer patients., (© 2019 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2019
- Full Text
- View/download PDF
44. Tissue Harvesting for Adoptive Tumor Infiltrating Lymphocyte Therapy in Metastatic Melanoma.
- Author
-
Zippel D, Friedman-Eldar O, Rayman S, Hazzan D, Nissan A, Schtrechman G, Markel G, Schachter J, Itzhaki O, and Besser MJ
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor, Female, Humans, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Male, Melanoma metabolism, Melanoma pathology, Middle Aged, Neoplasm Metastasis, Neoplasm Staging, Programmed Cell Death 1 Receptor antagonists & inhibitors, Young Adult, Immunotherapy, Adoptive methods, Lymphocytes, Tumor-Infiltrating immunology, Melanoma immunology, Melanoma therapy
- Abstract
Background/aim: Adoptive transfer of tumor-infiltrating lymphocytes (TILs) combined with non-myeloablative chemotherapy (NMA) has been shown to prolong survival in patients with metastatic disease., Materials and Methods: Tissue harvesting was performed form a variety of sites. TILs were isolated, expanded and infused with bolus high-dose IL-2., Results: Between 2008 and 2018, 242 lesions were resected for TILs harvesting from a range of sites form 196 patients without mortality and with minimal morbidity. Of those harvested, 75 were unable to complete therapy because of clinical deterioration during the wait period. Of 121 evaluable treated patients, there was no effect of metastatic site biopsied on the mean fold TIL expansion. Those receiving prior ipilimumab had a higher TIL fold expansion but a lower TIL fold expansion than those exposed to anti-PD1 therapy., Conclusion: Harvesting may be safely performed with successful TIL expansion from most sites. Prior check point inhibitory immunotherapy may potentially influence TIL fold expansion., (Copyright© 2019, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
45. Combined Expression of Genetic Adjuvants Via mRNA Electroporation Exerts Multiple Immunostimulatory Effects on Antitumor T Cells.
- Author
-
Weinstein-Marom H, Levin N, Pato A, Shmuel N, Sharabi-Nov A, Peretz T, Eisenberg G, Lotem M, Itzhaki O, Besser MJ, and Gross G
- Subjects
- Adjuvants, Immunologic administration & dosage, Cells, Cultured, Humans, Interferon-gamma, Melanoma therapy, CD8-Positive T-Lymphocytes transplantation, Electroporation, Gene Transfer Techniques, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating transplantation, RNA, Messenger administration & dosage
- Abstract
Adoptive transfer of tumor-infiltrating lymphocytes (TILs) or gene-modified T cells expressing antitumor TCRs or chimeric antigen receptors often yields a high rate of clinical response in several types of cancer. New approaches for enhancing the functional properties of antitumor T cells could improve the clinical outcome of these treatments. To this end, we created 3 classes of genes, each designed to operate autonomously upon expression in T cells. We recently reported on the enhancing effects of constitutively active toll-like receptor 4 (caTLR4), membrane (mem) interleukin-2, memIL-12, and memIL-15, and self-oligomerizing, constitutively active CD40 (caCD40). Here, we evaluated their combined effects on peripheral blood CD8 T cells and different antimelanoma TIL cultures following mRNA electroporation. Expression in CD8 T cells induced transient production of interferon-γ and prolonged and robust upregulation of CD25, CD69, 4-1BB, and OX40. The adjuvants enhanced cytolytic activity of TILs and production of interferon-γ and TNF-α in the presence of autologous, but not mismatched, melanoma for at least 3 days after electroporation. Expression of the 3 adjuvants in young TILs from different patients markedly increased the expression of CD25, OX40, 4-1BB, CD127, and CD28 and exhibited cooperative and, at times, synergistic effects. Furthermore, predefined mixtures of mRNA encoding these adjuvants markedly enhanced the specific antitumor response of selected TILs and killing of autologous melanoma cells by young TILs. Our findings suggest that combinations of these new genetic adjuvants can substantially improve the functional properties of antitumor T cells, offering a new tool of unique versatility in adoptive cell therapy.
- Published
- 2019
- Full Text
- View/download PDF
46. Locally produced CD19 CAR T cells leading to clinical remissions in medullary and extramedullary relapsed acute lymphoblastic leukemia.
- Author
-
Jacoby E, Bielorai B, Avigdor A, Itzhaki O, Hutt D, Nussboim V, Meir A, Kubi A, Levy M, Zikich D, Zeltzer LA, Brezinger K, Schachter J, Nagler A, Besser MJ, and Toren A
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Female, Hematopoietic Stem Cell Transplantation methods, Humans, Immunotherapy, Adoptive adverse effects, Male, Middle Aged, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, Receptors, Antigen, T-Cell immunology, Remission Induction methods, Salvage Therapy adverse effects, Salvage Therapy methods, Survival Rate, Treatment Outcome, Young Adult, Antigens, CD19 immunology, Immunotherapy, Adoptive methods, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
Autologous CD19 chimeric-antigen receptor (CAR) T cells demonstrated remarkable remission rates in relapsed and refractory acute lymphoblastic leukemia (R/R ALL). Here, we report results from a phase 1b/2 study of in-house produced CD19 CAR with a CD28 costimulatory domain. Twenty-one patients with R/R ALL were enrolled, and 20 infused. The median age was 11 years (range, 5-48). Patients had a median of 4 prior regimens, including blinatumomab in 6 and prior stem-cell transplantation in 10. In total 8 patients had extramedullary (EM) leukemic involvement, and prior to lymphodepletion and CAR 7 had active lesions, a group underrepresented in previous trials. In vivo expansion of CAR T cells was observed in 18 patients. In total 16 patients developed cytokine release syndrome, and 11 patients developed neurotoxicity, with no toxic deaths. All responding patients were referred to an allogeneic hematopoietic stem-cell transplantation. The remission rate was 90%, including resolution of all refractory EM sites. Four responding patients relapsed, 3 who had a PCR-MRD positive remission at 28 days following CAR-T cells and 1 patient 21 months after an MRD-negative response. The estimated 1-year event-free survival and overall survival are 73% and 90%, respectively. Patients with R/R EM ALL may also benefit from CAR-T cell therapy., (© 2018 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
47. Potent Activation of Human T Cells by mRNA Encoding Constitutively Active CD40.
- Author
-
Levin N, Weinstein-Marom H, Pato A, Itzhaki O, Besser MJ, Eisenberg G, Peretz T, Lotem M, and Gross G
- Subjects
- Adjuvants, Immunologic, Humans, Immunotherapy, Adoptive methods, Melanoma immunology, RNA, Messenger, Skin Neoplasms immunology, Tumor Cells, Cultured, CD40 Antigens immunology, Lymphocyte Activation immunology, Lymphocytes, Tumor-Infiltrating immunology, T-Lymphocytes immunology
- Abstract
New strategies for augmenting the actual performance of therapeutic T cells in vivo are needed for improving clinical outcome of adoptive cell therapy. Cumulative findings suggest that CD40 plays an intrinsic role in T cell costimulation. Recently, we demonstrated the ability of truncated, auto-oligomerizing CD40 derivatives to induce strong activation of APCs in a ligand-independent manner. We reasoned that constitutively active CD40 (caCD40) can similarly exert enhancing effects on human antitumor T cells. To test this assumption, we transfected human T cells with in vitro-transcribed caCD40 mRNA. In polyclonal T cells, caCD40 triggered IFN-γ secretion and upregulated CD25 and 4-1BB. In antimelanoma tumor-infiltrating lymphocytes (TILs), caCD40 induced massive production of IFN-γ, exerting a pronounced synergistic effect when coexpressed with constitutively active TLR4 devoid of its extracellular ligand binding. In unselected "young" TILs, caCD40 reproducibly increased surface expression of CD25, OX40, 4-1BB, CD127, and CD28. Three days post-mRNA electroporation of CD8 TILs, caCD40 elevated IFN-γ and TNF-α production and cytolytic activity in the presence of autologous but not HLA-I-mismatched melanoma. Enhanced killing of autologous melanoma by young TILs was observed 4 d posttransfection. These findings suggest that caCD40 can function as a potent T cell adjuvant and provide essential guidelines for similar manipulation of other key members of the TNFR family., (Copyright © 2018 by The American Association of Immunologists, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
48. Establishment of adoptive cell therapy with tumor infiltrating lymphocytes for non-small cell lung cancer patients.
- Author
-
Ben-Avi R, Farhi R, Ben-Nun A, Gorodner M, Greenberg E, Markel G, Schachter J, Itzhaki O, and Besser MJ
- Subjects
- Aged, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Follow-Up Studies, Humans, Lung Neoplasms immunology, Lung Neoplasms pathology, Lymphocyte Activation, Male, Prognosis, Carcinoma, Non-Small-Cell Lung therapy, Cell- and Tissue-Based Therapy, Immunotherapy, Adoptive, Lung Neoplasms therapy, Lymphocytes, Tumor-Infiltrating immunology, T-Lymphocytes immunology
- Abstract
Adoptive cell therapy (ACT) of tumor infiltration lymphocytes (TIL) yields promising clinical results in metastatic melanoma patients, who failed standard treatments. Due to the fact that metastatic lung cancer has proven to be susceptible to immunotherapy and possesses a high mutation burden, which makes it responsive to T cell attack, we explored the feasibility of TIL ACT in non-small cell lung cancer (NSCLC) patients. Multiple TIL cultures were isolated from tumor specimens of five NSCLC patients undergoing thoracic surgery. We were able to successfully establish TIL cultures by various methods from all patients within an average of 14 days. Fifteen lung TIL cultures were further expanded to treatment levels under good manufacturing practice conditions and functionally and phenotypically characterized. Lung TIL expanded equally well as 103 melanoma TIL obtained from melanoma patients previously treated at our center, and had a similar phenotype regarding PD1, CD28, and 4-1BB expressions, but contained a higher percent of CD4 T cells. Lung carcinoma cell lines were established from three patients of which two possessed TIL cultures with specific in vitro anti-tumor reactivity. Here, we report the successful pre-clinical production of TIL for immunotherapy in the lung cancer setting, which may provide a new treatment modality for patients with metastatic NSCLC. The initiation of a clinical trial is planned for the near future.
- Published
- 2018
- Full Text
- View/download PDF
49. Selection of Shared and Neoantigen-Reactive T Cells for Adoptive Cell Therapy Based on CD137 Separation.
- Author
-
Seliktar-Ofir S, Merhavi-Shoham E, Itzhaki O, Yunger S, Markel G, Schachter J, and Besser MJ
- Abstract
Adoptive cell therapy (ACT) of autologous tumor infiltrating lymphocytes (TIL) is an effective immunotherapy for patients with solid tumors, yielding objective response rates of around 40% in refractory patients with metastatic melanoma. Most clinical centers utilize bulk, randomly isolated TIL from the tumor tissue for ex vivo expansion and infusion. Only a minor fraction of the administered T cells recognizes tumor antigens, such as shared and mutation-derived neoantigens, and consequently eliminates the tumor. Thus, there are many ongoing effects to identify and select tumor-specific TIL for therapy; however, those approaches are very costly and require months, which is unreasonable for most metastatic patients. CD137 (4-1BB) has been identified as a co-stimulatory marker, which is induced upon the specific interaction of T cells with their target cell. Therefore, CD137 can be a useful biomarker and an important tool for the selection of tumor-reactive T cells. Here, we developed and validated a simple and time efficient method for the selection of CD137-expressing T cells for therapy based on magnetic bead separation. CD137 selection was performed with clinical grade compliant reagents, and TIL were expanded in a large-scale manner to meet cell numbers required for the patient setting in a GMP facility. For the first time, the methodology was designed to comply with both clinical needs and limitations, and its feasibility was assessed. CD137-selected TIL demonstrated significantly increased antitumor reactivity and were enriched for T cells recognizing neoantigens as well as shared tumor antigens. CD137-based selection enabled the enrichment of tumor-reactive T cells without the necessity of knowing the epitope specificity or the antigen type. The direct implementation of the CD137 separation method to the cell production of TIL may provide a simple way to improve the clinical efficiency of TIL ACT.
- Published
- 2017
- Full Text
- View/download PDF
50. Adoptive Cell Therapy for Metastatic Melanoma.
- Author
-
Merhavi-Shoham E, Itzhaki O, Markel G, Schachter J, and Besser MJ
- Subjects
- Animals, Humans, Melanoma immunology, Melanoma pathology, Randomized Controlled Trials as Topic, Immunotherapy, Adoptive methods, Melanoma therapy
- Abstract
Adoptive cell therapy (ACT) of tumor-infiltrating lymphocytes (TILs) is a powerful form of immunotherapy by inducing durable complete responses that significantly extend the survival of melanoma patients. Mutation-derived neoantigens were recently identified as key factors for tumor recognition and rejection by TILs. The isolation of T-cell receptor (TCR) genes directed against neoantigens and their retransduction into peripheral T cells may provide a new form of ACT.Genetic modifications of T cells with chimeric antigen receptors (CARs) have demonstrated remarkable clinical results in hematologic malignancies, but are so far less effective in solid tumors. Only very limited reports exist in melanoma. Progress in CAR T-cell engineering, including neutralization of inhibitory signals or additional safety switches, may open opportunities also in melanoma.We review clinical results and latest developments of adoptive therapies with TILs, T-cell receptor, and CAR-modified T cells and discuss future directions for the treatment of melanoma.
- 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.