203 results on '"Gee AP"'
Search Results
2. Flow cytometric cell sorting combined with molecular chimerism analysis to detect minimal recurrent leukemia: good news and bad news
- Author
-
Jr, LSLamb, Robbins, NF, Abhyankar, S, Joyce, M, Stetler-Stevenson, M, Henslee-Downey, PJ, and Gee, AP
- Published
- 1997
- Full Text
- View/download PDF
3. Donor leukocyte infusion for treatment of graft rejection post partially mismatched related donor bone marrow transplant
- Author
-
Godder, KT, Abhyankar, SH, Lamb, LS, Best, RG, Geier, SS, Pati, AR, Gee, AP, and Henslee-Downey, PJ
- Published
- 1998
- Full Text
- View/download PDF
4. Immunotherapy of high-risk acute leukemia with a recipient (autologous) vaccine expressing transgenic human CD40L and IL-2 after chemotherapy and allogeneic stem cell transplantation.
- Author
-
Rousseau, R, Biagi, E, Dutour, A, Yvon, E, Brown, M, Lin, T, Mei, Z, Grilley, B, Popek, E, Heslop, H, Gee, A, Krance, R, Popat, U, Carrum, G, Margolin, J, Brenner, M, Rousseau, RF, Yvon, ES, Brown, MP, Heslop, HE, Gee, AP, Krance, RA, Margolin, JF, Brenner, MK, BIAGI, ETTORE, Rousseau, R, Biagi, E, Dutour, A, Yvon, E, Brown, M, Lin, T, Mei, Z, Grilley, B, Popek, E, Heslop, H, Gee, A, Krance, R, Popat, U, Carrum, G, Margolin, J, Brenner, M, Rousseau, RF, Yvon, ES, Brown, MP, Heslop, HE, Gee, AP, Krance, RA, Margolin, JF, Brenner, MK, and BIAGI, ETTORE
- Abstract
CD40L generates immune responses in leukemia-bearing mice, an effect that is potentiated by IL-2. We studied the feasibility, safety, and immunologic efficacy of an IL-2- and CD40L-expressing recipient-derived tumor vaccine consisting of leukemic blasts admixed with skin fibroblasts transduced with adenoviral vectors encoding human IL-2 (hIL-2) and hCD40L. Ten patients (including 7 children) with high-risk acute myeloid (n = 4) or lymphoblastic (n = 6) leukemia in cytologic remission (after allogeneic stem cell transplantation [n = 9] or chemotherapy alone [n = 1]) received up to 6 subcutaneous injections of the IL-2/CD40L vaccine. None of the patients were receiving immunosuppressive drugs. No severe adverse reactions were noted. Immunization produced a 10- to 890-fold increase in the frequencies of major histocompatibility complex (MHC)-restricted T cells reactive against recipient-derived blasts. These leukemia-reactive T cells included both T-cytotoxic/T-helper 1 (Th1) and Th2 subclasses, as determined from their production of granzyme B, interferon-gamma, and interleukin-5. Two patients produced systemic IgG antibodies that bound to their blasts. Eight patients remained disease free for 27 to 62 months after treatment (5-year overall survival, 90%). Thus, even in heavily treated patients, including recipients of allogeneic stem cell transplants, recipient-derived antileukemia vaccines can induce immune responses reactive against leukemic blasts. This approach may be worthy of further study, particularly in patients with a high risk of relapse.
- Published
- 2006
5. Effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: the TIME randomized trial.
- Author
-
Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis SG, Forder JR, Anderson RD, Hatzopoulos AK, Penn MS, Perin EC, Chambers J, Baran KW, Raveendran G, Lambert C, Lerman A, Simon DI, Vaughan DE, Lai D, and Gee AP
- Abstract
Context: While the delivery of cell therapy after ST-segment elevation myocardial infarction (STEMI) has been evaluated in previous clinical trials, the influence of the timing of cell delivery on the effect on left ventricular function has not been analyzed.Objectives: To determine the effect of intracoronary autologous bone marrow mononuclear cell (BMC) delivery after STEMI on recovery of global and regional left ventricular function and whether timing of BMC delivery (3 days vs 7 days after reperfusion) influences this effect.Design, Setting, and Patients: A randomized, 2 × 2 factorial, double-blind, placebo-controlled trial, Timing In Myocardial infarction Evaluation (TIME) enrolled 120 patients with left ventricular dysfunction (left ventricular ejection fraction [LVEF] ≤ 45%) after successful primary percutaneous coronary intervention (PCI) of anterior STEMI between July 17, 2008, and November 15, 2011, as part of the Cardiovascular Cell Therapy Research Network sponsored by the National Heart, Lung, and Blood Institute.Interventions: Intracoronary infusion of 150 × 106 BMCs or placebo (randomized 2:1) within 12 hours of aspiration and cell processing administered at day 3 or day 7 (randomized 1:1) after treatment with PCI.Main Outcome Measures: The primary end points were change in global (LVEF) and regional (wall motion) left ventricular function in infarct and border zones at 6 months measured by cardiac magnetic resonance imaging and change in left ventricular function as affected by timing of treatment on day 3 vs day 7. The secondary end points included major adverse cardiovascular events as well as changes in left ventricular volumes and infarct size.Results: The mean (SD) patient age was 56.9 (10.9) years and 87.5% of participants were male. At 6 months, there was no significant increase in LVEF for the BMC group (45.2% [95% CI, 42.8% to 47.6%] to 48.3% [95% CI, 45.3% to 51.3%) vs the placebo group (44.5% [95% CI, 41.0% to 48.0%] to 47.8% [95% CI, 43.4% to 52.2%]) (P = .96). There was no significant treatment effect on regional left ventricular function observed in either infarct or border zones. There were no significant differences in change in global left ventricular function for patients treated at day 3 (−0.9% [95% CI, −6.6% to 4.9%], P = .76) or day 7 (1.1% [95% CI, −4.7% to 6.9%], P = .70). The timing of treatment had no significant effect on regional left ventricular function recovery. Major adverse events were rare among all treatment groups.Conclusion: Among patients with STEMI treated with primary PCI, the administration of intracoronary BMCs at either 3 days or 7 days after the event had no significant effect on recovery of global or regional left ventricular function compared with placebo.Trial Registration: clinicaltrials.gov Identifier: NCT00684021. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
6. Effect of transendocardial delivery of autologous bone marrow mononuclear cells on functional capacity, left ventricular function, and perfusion in chronic heart failure: the FOCUS-CCTRN trial.
- Author
-
Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, Silva GV, Lai D, Thomas JD, Kronenberg MW, Martin AD, Anderson RD, Traverse JH, Penn MS, Anwaruddin S, Hatzopoulos AK, Gee AP, Taylor DA, Cogle CR, and Smith D
- Abstract
Context: Previous studies using autologous bone marrow mononuclear cells (BMCs) in patients with ischemic cardiomyopathy have demonstrated safety and suggested efficacy.Objective: To determine if administration of BMCs through transendocardial injections improves myocardial perfusion, reduces left ventricular end-systolic volume (LVESV), or enhances maximal oxygen consumption in patients with coronary artery disease or LV dysfunction, and limiting heart failure or angina.Design, Setting, and Patients: A phase 2 randomized double-blind, placebo-controlled trial of symptomatic patients (New York Heart Association classification II-III or Canadian Cardiovascular Society classification II-IV) with a left ventricular ejection fraction of 45% or less, a perfusion defect by single-photon emission tomography (SPECT), and coronary artery disease not amenable to revascularization who were receiving maximal medical therapy at 5 National Heart, Lung, and Blood Institute-sponsored Cardiovascular Cell Therapy Research Network (CCTRN) sites between April 29, 2009, and April 18, 2011.Intervention: Bone marrow aspiration (isolation of BMCs using a standardized automated system performed locally) and transendocardial injection of 100 million BMCs or placebo (ratio of 2 for BMC group to 1 for placebo group).Main Outcome Measures: Co-primary end points assessed at 6 months: changes in LVESV assessed by echocardiography, maximal oxygen consumption, and reversibility on SPECT. Phenotypic and functional analyses of the cell product were performed by the CCTRN biorepository core laboratory.Results: Of 153 patients who provided consent, a total of 92 (82 men; average age: 63 years) were randomized (n = 61 in BMC group and n = 31 in placebo group). Changes in LVESV index (-0.9 mL/m(2) [95% CI, -6.1 to 4.3]; P = .73), maximal oxygen consumption (1.0 [95% CI, -0.42 to 2.34]; P = .17), and reversible defect (-1.2 [95% CI, -12.50 to 10.12]; P = .84) were not statistically significant. There were no differences found in any of the secondary outcomes, including percent myocardial defect, total defect size, fixed defect size, regional wall motion, and clinical improvement.Conclusion: Among patients with chronic ischemic heart failure, transendocardial injection of autologous BMCs compared with placebo did not improve LVESV, maximal oxygen consumption, or reversibility on SPECT.Trial Registration: clinicaltrials.gov Identifier: NCT00824005. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
7. Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: the LateTIME randomized trial.
- Author
-
Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, Forder JR, Byrne BJ, Hatzopoulos AK, Penn MS, Perin EC, Baran KW, Chambers J, Lambert C, Raveendran G, Simon DI, Vaughan DE, Simpson LM, Gee AP, and Taylor DA
- Abstract
Context: Clinical trial results suggest that intracoronary delivery of autologous bone marrow mononuclear cells (BMCs) may improve left ventricular (LV) function when administered within the first week following myocardial infarction (MI). However, because a substantial number of patients may not present for early cell delivery, the efficacy of autologous BMC delivery 2 to 3 weeks post-MI warrants investigation.Objective: To determine if intracoronary delivery of autologous BMCs improves global and regional LV function when delivered 2 to 3 weeks following first MI.Design, Setting, and Patients: A randomized, double-blind, placebo-controlled trial (LateTIME) of the National Heart, Lung, and Blood Institute-sponsored Cardiovascular Cell Therapy Research Network of 87 patients with significant LV dysfunction (LV ejection fraction [LVEF] ≤45%) following successful primary percutaneous coronary intervention (PCI) between July 8, 2008, and February 28, 2011.Interventions: Intracoronary infusion of 150 × 10(6) autologous BMCs (total nucleated cells) or placebo (BMC:placebo, 2:1) was performed within 12 hours of bone marrow aspiration after local automated cell processing.Main Outcome Measures: Changes in global (LVEF) and regional (wall motion) LV function in the infarct and border zone between baseline and 6 months, measured by cardiac magnetic resonance imaging. Secondary end points included changes in LV volumes and infarct size.Results: A total of 87 patients were randomized (mean [SD] age, 57 [11] years; 83% men). Harvesting, processing, and intracoronary delivery of BMCs in this setting was feasible. Change between baseline and 6 months in the BMC group vs placebo for mean LVEF (48.7% to 49.2% vs 45.3% to 48.8%; between-group mean difference, -3.00; 95% CI, -7.05 to 0.95), wall motion in the infarct zone (6.2 to 6.5 mm vs 4.9 to 5.9 mm; between-group mean difference, -0.70; 95% CI, -2.78 to 1.34), and wall motion in the border zone (16.0 to 16.6 mm vs 16.1 to 19.3 mm; between-group mean difference, -2.60; 95% CI, -6.03 to 0.77) were not statistically significant. No significant change in LV volumes and infarct volumes was observed; both groups decreased by a similar amount at 6 months vs baseline.Conclusion: Among patients with MI and LV dysfunction following reperfusion with PCI, intracoronary infusion of autologous BMCs vs intracoronary placebo infusion, 2 to 3 weeks after PCI, did not improve global or regional function at 6 months.Trial Registration: clinicaltrials.gov Identifier: NCT00684060. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
8. Gene transfer: regulatory issues and their impact on the clinical investigator and the good manufacturing production facility.
- Author
-
Grilley, Bj and Gee, Ap
- Subjects
- *
GENETIC transformation , *GENETIC recombination , *NUCLEIC acids , *MICROBIAL genetics , *RECOMBINANT DNA - Abstract
Summary The first human gene-transfer study was submitted to the Recombinant DNA Advisory Committee (RAC) in 1988, thus initiating a new era in clinical research. As per the RAC Website (last updated 22nd November 2002), almost 550 human gene-transfer studies have been submitted to the RAC. However, there are currently no licensed gene-therapy products available in the USA. The natural evolution of the review process to accommodate these novel protocols, as well as the death of Jesse Gelsinger in 1999, have led to significant changes in the initial and ongoing review of gene-transfer studies. However, the basic framework of the review process remains unchanged. Gene-transfer protocols require oversight by the Food and Drug Administration (FDA), the Recombinant DNA Advisory Committee (RAC), the Institutional Biosafety Committee (IBC), and the Institutional Review Board (IRB). Such oversight includes both initial review of the protocol and ongoing review of the study through the review of annual reports, adverse events, and proposed amendments to the study. In addition to such review of the protocol, the product itself is required by the FDA to be prepared under current good manufacturing practices (cGMP). This article discusses both regulatory oversight and current GMP issues in depth. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
9. In vitro interactions between γδT cells, DC, and CD4[sup +] T cells; implications for the immunotherapy of leukemia.
- Author
-
Ye, Z, Haley, S, Gee, AP, Henslee-Downey, PJ, and Lamb Jr, LS
- Subjects
T cells ,DENDRITIC cells ,ANTIGEN presenting cells ,LYMPHOID tissue ,IMMUNOTHERAPY ,LEUKEMIA - Abstract
Background: γδT cells contribute to immune defense against infectious organisms and some malignancies, but the process of activation and proliferation of these cells is not well understood. It is known that the immune response of γδT cells is not MHC-dependent, but is likely based on direct recognition of surface peptides and non-peptide ligands. This study examined whether DCs and CD4[sup +] T cells can participate in the activation of γδT cells. Method: Peripheral blood γδT cells were co-cultured with CD34-derived autologous DCs and CD4[sup +] T cells using contact-dependent cultures and transwell systems. Proliferation, immunophenotyping, and cytotoxicity assays determined the extent of γδT cell proliferation and cytotoxicity. Results: Human γδT cells expanded 221.3 ± 76-fold in cultures with DCs, and 165.7 ± 76.6-fold with CD4[sup +] T-cells alone. Proliferation was enhanced (1949.8 ± 261.3-fold) when γδT cells were cultured with both DC and CD4[sup +] T cells. Proliferation was contact-dependent, and resulted in the expansion of Vδ1+ or Vδ2+ cells cytotoxic against several leukemic cell-lines, but not against allogeneic PHA-induced lymphoid blasts. Ligation of the T-cell receptor with anti-pan-δ Ab significantly up-regulated cytotoxicity against K562, KBM-5 and KG1a, and normal BM, but not against Molt-4, allogeneic EBV-transfected B cells and allogeneic PHA-blasts. Minimal cytotoxic activity was shown against allogeneic marrow colony-forming units granulocyte-macrophage and erythrocyte colony-forming units. Conclusion: DCs can participate in the activation of γδT cells against specific autologous targets, and cytotoxicity can be enhanced by further stimulation via the T-cell receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
10. Product Release Assays.
- Author
-
Gee, AP
- Subjects
- *
FIRE assay , *CHEMISTRY , *CELLS , *BIOLOGY , *ENDOTOXINS - Abstract
Provides an overview of the types of assays used in cell manufacturing. Features of sterility testing; Assays for cell characterization; Endotoxin testing.
- Published
- 1999
- Full Text
- View/download PDF
11. Influence of T Cell Depletion Method on Circulating γδ T Cell Reconstitution and Potential Role in the Graft-Versus-Leukemia Effect.
- Author
-
Lamb Jr, LS, Gee, AP, Hazlett, LJ, Musk, P, Parrish, RS, O'Hanlon, TP, Geier, SS, Folk, RS, Harris, WG, McPherson, K, Lee, C, and Henslee-Downey, PJ
- Subjects
- *
T cells , *GRAFT versus host disease , *LEUKEMIA , *ANTINEOPLASTIC agents , *BONE marrow transplantation - Abstract
Background Our laboratory previously reported that leukemia patients who developed ≥ 10% γδ + T cells during the first six months after receiving an anti-TCRαβ T-cell-depleted (TCD) graft from a partially mismatched related donor (PMRD) had a disease-free survival (DFS) advantage. These γδ + T cells were Vδ1 + CD3 + CD4 - CD8 - CD69 + HLADR + and are cytotoxic to K562 cells. Methods In order to determine whether the anti-αβ TCD regimen was associated with these findings, we compared the reconstitution of γδ + T cells from patients who received TCD PMRD grafts using the anti-tCRαβ MAb T10B9-1A31 (previously reported) with similar patients who received grafts using the anti-CD3 MAb OKT3. Results Increased cytotoxic Vδ1+ T cells were seen in 10 of 43 T10B9 TCD patients compared to 7 of 100 in the OKT3 TCD group (23% versus 7%, p = 0.010 ). T10B9 patients with increased γδ + T cells also exhibited a higher range of increased γδ + T cells and the length of time the γδ + T cells remained high was longer when compared to OKT3 patients. Patients with increased γδ + T cells whose grafts were T-cell depleted with T10B9 showed a significant decrease in relapse ( p = 0.038 ). Similar rates and reduction in relapse were seen in OKT3 TCD patients, although significance was not reached due to the small number of patients with increased γδ + T cells. Estimated 3 year disease-free survival was significantly improved in T10B9 patients with increased γδ + T cells (0.79 versus 0.31, p = 0.009 ), a trend also seen in OKT3 patients ( p = 0.091 ). Discussion These observations indicate that Vδ1 + CD4 - CD8 - cytotoxic T cells are associated with lower relapse rates and improved survival, and thus may have a role in a graft-versus-leukemia effect. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
12. Debulking Blood Stem Cell Collections by Density Gradient Centrifugation in a Closed-Vessel System.
- Author
-
Przepiorka, D, Lu, J-G, Anderlini, P, Körbling, M, Donato, M, Champlin, R, Gee, AP, and van Vlasselaer, P
- Subjects
BLOOD cells ,STEM cells ,HEMAPHERESIS ,CELLS ,BONE marrow - Abstract
Background Blood stem cells collected by apheresis are largely mononuclear cells in nature, so manipulation of blood stem-cell components freequently requires more time and reagents than for a marrow harvest. Reducing the nucleated cell number in mobilized blood stemcell collections, while preserving hematopoietic progenitor content, would make such manipulations simpler and less costly, but only if debulking procedures were not complex. Methods We evaluated separation of light-density cells and enrichment of CD34 + cells from mobilized peripheral blood stem-cell collections by density gradient centrifugation over buoyant density solution 60 (BDS 60) in a single, closed vessel. Results Fifteen apheresis products from five normal volunteers and eight cancer patients contained 3.44 (range, 1.19-5.51) × 10 10 nucleated cells. Following processing and washing, there was a median 29% recovery of nucleated cells, 79% recovery of CD34 + cells, 2.49-fold enrichment of CD34 + cells, 0.96-log depletion of CD3 + cells, 0.48-log depletion of CD56 + cells, and 0.72-log depletion of CD19 + cells. Results of processing were affected by the variability in composition of the apheresis products. The enrichment of CD34 + cells varied by donor type, and there was a logarithmic relationship between the preprocessing percentage of CD19 + cells and the log reduction in CD19 + cells. Recovery of cells after thawing and washing was acceptable for processed cells cryopreserved at concentrations over the range of 0.01-1.5 × 10 8 /mL. Discussion These results demonstrate a simple method by which an apheresis product of 1-5 × 10 10 cells can be debulked effectively in a single, closed vessel. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
13. What a cell-processing laboratory can and can't do for cellular therapy.
- Author
-
Gee, AP
- Subjects
- *
THERAPEUTICS , *CLINICAL medicine , *CELLULAR therapy , *ORGANOTHERAPY , *MEDICAL research - Abstract
Reviews the role of a cell-processing facility (CPF) in cellular therapy. Responsibility for bridging the gap between basic and clinical research; Components of a cellular-therapy batch record; Information on pre-clinical tasks for the CPF.
- Published
- 2002
- Full Text
- View/download PDF
14. Immune therapy for EBV infections after hemopoietic stem-cell transplant.
- Author
-
Heslop, HE, Bollard, CM, Gottschalk, S, Kuehnle, I, Huls, MH, Gee, AP, Brenner, MK, and Rooney, CM
- Subjects
EPSTEIN-Barr virus ,ONCOGENIC DNA viruses ,THERAPEUTICS ,STEM cells ,CELL transplantation - Abstract
Discusses research on immune therapy for Epstein-Barr virus (EBV) infections after hemopoietic stem-cell transplant. Treatment received by patients on the prophylaxis study; Susceptibility of tumor cells to immunotherapeutic approaches in other EBV-associated primary malignancies; Information on an alternate means of prophylaxis.
- Published
- 2002
- Full Text
- View/download PDF
15. Cell sorting for therapeutic applications — points to consider.
- Author
-
Gee, AP and Durett, AG
- Subjects
- *
FLOW cytometry , *FLUORIMETRY , *CYTOLOGICAL techniques - Abstract
Discusses the development of a fluorescence-activated cell sorter (FACS)-based procedure for the preparation of therapeutic cells. Advantage of FACS over alternative methods for cell enrichment; Classical disadvantage with FACS; Measurement of viability; Regulatory issues.
- Published
- 2002
- Full Text
- View/download PDF
16. Are You Feeling Ablated?
- Author
-
Gee, AP
- Subjects
- *
SCIENCE career counseling , *MEDICAL career counseling , *PSYCHOLOGICAL burnout , *BUREAUCRACY , *ASSOCIATIONS, institutions, etc. - Abstract
Focuses on the environment for scientific and medical achievements as of 1999. Reasons for the burnout experienced by people in medicine; Impact of bureaucracy in the medical fields; Role of professional society ISHAGE.
- Published
- 1999
- Full Text
- View/download PDF
17. ISHAGE Committees -- the Whole is, Indeed, and in Deed, Greater Than the Sum of its Parts.
- Author
-
Collins, NH and Gee, AP
- Subjects
- *
ASSOCIATIONS, institutions, etc. , *SOCIETIES , *BLOOD , *TRANSPLANTATION of organs, tissues, etc. , *CELLS - Abstract
Highlights the contributions of the members of the Scientific and Standing Committees of the International Society for Hematotherapy and Graft Engineering (ISHAGE). Function in the society; Report of the Scientific Committee on assays that measure primitive hematopoietic progenitor cells; Mandate of the ISHAGE committee structure.
- Published
- 1999
18. Environmental oxygen affects ex vivo growth and proliferation of mesenchymal progenitors by modulating mitogen-activated protein kinase and mammalian target of rapamycin signaling.
- Author
-
da Graça Cabreira M, Wang X, Critsinelis A, Setegne M, Lotfi P, Wan YW, Barrios G, Mei Z, Gee AP, Buja LM, and Perin E
- Subjects
- Cells, Cultured, Sirolimus, Cell Proliferation, Cell Differentiation physiology, TOR Serine-Threonine Kinases, Oxygen metabolism, Mitogen-Activated Protein Kinases
- Abstract
Background Aims: Stem and progenitor cells of hematopoietic and mesenchymal lineages reside in the bone marrow under low oxygen (O
2 ) saturation. O2 levels used in ex vivo expansion of multipotent mesenchymal stromal cells (MSCs) affect proliferation, metabolism and differentiation., Methods: Using cell-based assays and transcriptome and proteome data, the authors compared MSC cultures simultaneously grown under a conventional 19.95% O2 atmosphere or at 5% O2 ., Results: In 5% O2 , MSCs showed better proliferation and higher self-renewal ability, most probably sustained by enhanced signaling activity of mitogen-activated protein kinase and mammalian target of rapamycin pathways. Non-oxidative glycolysis-based energy metabolism supported growth and proliferation in 5% O2 cultures, whereas MSCs grown under 19.95% O2 also utilized oxidative phosphorylation. Cytoprotection mechanisms used by cells under 5% O2 2 2 2 for MSC culture., (Copyright © 2022 International Society for Cell & Gene Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
19. Long-term follow-up for the development of subsequent malignancies in patients treated with genetically modified IECs.
- Author
-
Steffin DHM, Muhsen IN, Hill LC, Ramos CA, Ahmed N, Hegde M, Wang T, Wu M, Gottschalk S, Whittle SB, Lulla PD, Mamonkin M, Omer B, Rouce RH, Heczey A, Metelitsa LS, Grilley BJ, Robertson C, Torrano V, Lapteva N, Gee AP, Rooney CM, Brenner MK, and Heslop HE
- Subjects
- Adult, Child, Follow-Up Studies, Humans, Leukocytes, Mononuclear, Retrospective Studies, Hematologic Neoplasms genetics, Hematologic Neoplasms therapy, Neoplasms genetics, Neoplasms therapy
- Abstract
Subsequent malignancies are well-documented complications in long-term follow-up of cancer patients. Recently, genetically modified immune effector (IE) cells have shown benefit in hematologic malignancies and are being evaluated in clinical trials for solid tumors. Although the short-term complications of IE cells are well described, there is limited literature summarizing long-term follow-up, including subsequent malignancies. We retrospectively reviewed data from 340 patients treated across 27 investigator-initiated pediatric and adult clinical trials at our center. All patients received IE cells genetically modified with γ-retroviral vectors to treat relapsed and/or refractory hematologic or solid malignancies. In a cumulative 1027 years of long-term follow-up, 13 patients (3.8%) developed another cancer with a total of 16 events (4 hematologic malignancies and 12 solid tumors). The 5-year cumulative incidence of a first subsequent malignancy in the recipients of genetically modified IE cells was 3.6% (95% confidence interval, 1.8% to 6.4%). For 11 of the 16 subsequent tumors, biopsies were available, and no sample was transgene positive by polymerase chain reaction. Replication-competent retrovirus testing of peripheral blood mononuclear cells was negative in the 13 patients with subsequent malignancies tested. Rates of subsequent malignancy were low and comparable to standard chemotherapy. These results suggest that the administration of IE cells genetically modified with γ retroviral vectors does not increase the risk for subsequent malignancy., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
20. The National Heart, Lung, and Blood Institute-funded Production Assistance for Cellular Therapies (PACT) program: Eighteen years of cell therapy.
- Author
-
El Fiky A, Ibenana L, Anderson R, Hare JM, Khan A, Gee AP, Rooney C, McKenna DH, Gold J, Kelley L, Lundberg MS, Welniak LA, and Lindblad R
- Subjects
- Academies and Institutes, Government Regulation, United States, Cell- and Tissue-Based Therapy economics, Financing, Government, National Heart, Lung, and Blood Institute (U.S.)
- Abstract
The Production Assistance for Cellular Therapies (PACT) Program, is funded and supported by the US Department of Health and Human Services' National Institutes of Health (NIH) National Heart Lung and Blood Institute (NHLBI) to advance development of somatic cell and genetically modified cell therapeutics in the areas of heart, lung, and blood diseases. The program began in 2003, continued under two competitive renewals, and ended June 2021. PACT has supported cell therapy product manufacturing, investigational new drug enabling preclinical studies, and translational services, and has provided regulatory assistance for candidate cell therapy products that may aid in the repair and regeneration of damaged/diseased cells, tissues, and organs. PACT currently supports the development of novel cell therapies through five cell processing facilities. These facilities offer manufacturing processes, analytical development, technology transfer, process scale-up, and preclinical development expertise necessary to produce cell therapy products that are compliant with Good Laboratory Practices, current Good Manufacturing Practices, and current Good Tissue Practices regulations. The Emmes Company, LLC, serves as the Coordinating Center and assists with the management and coordination of PACT and its application submission and review process. This paper discusses the impact and accomplishments of the PACT program on the cell therapy field and its evolution over the duration of the program. It highlights the work that has been accomplished and provides a foundation to build future programs with similar goals to advance cellular therapeutics in a coordinated and centralized programmatic manner to support unmet medical needs within NHLBI purview., (© 2021 The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2021
- Full Text
- View/download PDF
21. Clinical effects of administering leukemia-specific donor T cells to patients with AML/MDS after allogeneic transplant.
- Author
-
Lulla PD, Naik S, Vasileiou S, Tzannou I, Watanabe A, Kuvalekar M, Lulla S, Carrum G, Ramos CA, Kamble R, Hill L, Randhawa J, Gottschalk S, Krance R, Wang T, Wu M, Robertson C, Gee AP, Chung B, Grilley B, Brenner MK, Heslop HE, Vera JF, and Leen AM
- Subjects
- Adolescent, Adult, Aged, Allografts, Antigens, Neoplasm immunology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Combined Modality Therapy, Female, Graft vs Host Disease etiology, Graft vs Host Disease prevention & control, Humans, Leukemia, Myeloid, Acute drug therapy, Male, Middle Aged, Myelodysplastic Syndromes drug therapy, Recurrence, T-Cell Antigen Receptor Specificity, T-Lymphocytes immunology, Tissue Donors, Young Adult, Graft vs Leukemia Effect, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute therapy, Lymphocyte Transfusion adverse effects, Myelodysplastic Syndromes therapy, Salvage Therapy, T-Lymphocytes transplantation
- Abstract
Relapse after allogeneic hematopoietic stem cell transplantation (HCT) is the leading cause of death in patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Infusion of unselected donor lymphocytes (DLIs) enhances the graft-versus-leukemia (GVL) effect. However, because the infused lymphocytes are not selected for leukemia specificity, the GVL effect is often accompanied by life-threatening graft-versus-host disease (GVHD), related to the concurrent transfer of alloreactive lymphocytes. Thus, to minimize GVHD and maximize GVL, we selectively activated and expanded stem cell donor-derived T cells reactive to multiple antigens expressed by AML/MDS cells (PRAME, WT1, Survivin, and NY-ESO-1). Products that demonstrated leukemia antigen specificity were generated from 29 HCT donors. In contrast to DLIs, leukemia-specific T cells (mLSTs) selectively recognized and killed leukemia antigen-pulsed cells, with no activity against recipient's normal cells in vitro. We administered escalating doses of mLSTs (0.5 to 10 × 107 cells per square meter) to 25 trial enrollees, 17 with high risk of relapse and 8 with relapsed disease. Infusions were well tolerated with no grade >2 acute or extensive chronic GVHD seen. We observed antileukemia effects in vivo that translated into not-yet-reached median leukemia-free and overall survival at 1.9 years of follow-up and objective responses in the active disease cohort (1 complete response and 1 partial response). In summary, mLSTs are safe and promising for the prevention and treatment of AML/MDS after HCT. This trial is registered at www.clinicaltrials.com as #NCT02494167., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
22. T-Cell Therapy for Lymphoma Using Nonengineered Multiantigen-Targeted T Cells Is Safe and Produces Durable Clinical Effects.
- Author
-
Vasileiou S, Lulla PD, Tzannou I, Watanabe A, Kuvalekar M, Callejas WL, Bilgi M, Wang T, Wu MJ, Kamble R, Ramos CA, Rouce RH, Zeng Z, Gee AP, Grilley BJ, Vera JF, Bollard CM, Brenner MK, Heslop HE, Rooney CM, Leen AM, and Carrum G
- Subjects
- Adolescent, Adult, Aged, Female, Humans, Lymphoma immunology, Male, Middle Aged, Prognosis, Young Adult, Antigens, Neoplasm immunology, Cell- and Tissue-Based Therapy methods, Lymphoma therapy, Salvage Therapy, T-Lymphocytes transplantation
- Abstract
Purpose: Patients with relapsed lymphomas often fail salvage therapies including high-dose chemotherapy and mono-antigen-specific T-cell therapies, highlighting the need for nontoxic, novel treatments. To that end, we clinically tested an autologous T-cell product that targets multiple tumor-associated antigens (TAAs) expressed by lymphomas with the intent of treating disease and preventing immune escape., Patients and Methods: We expanded polyclonal T cells reactive to five TAAs: PRAME, SSX2, MAGEA4, SURVIVIN, and NY-ESO-1. Products were administered to 32 patients with Hodgkin lymphomas (n = 14) or non-Hodgkin lymphomas (n = 18) in a two-part phase I clinical trial, where the objective of the first phase was to establish the safety of targeting all five TAAs (fixed dose, 0.5 × 10
7 cells/m2 ) simultaneously and the second stage was to establish the maximum tolerated dose. Patients had received a median of three prior lines of therapy and either were at high risk for relapse (adjuvant arm, n = 17) or had chemorefractory disease (n = 15) at enrollment., Results: Infusions were safe with no dose-limiting toxicities observed in either the antigen- or dose-escalation phases. Although the maximum tolerated dose was not reached, the maximum tested dose at which efficacy was observed (two infusions, 2 × 107 cells/m2 ) was determined as the recommended phase II dose. Of the patients with chemorefractory lymphomas, two (of seven) with Hodgkin lymphomas and four (of eight) with non-Hodgkin lymphomas achieved durable complete remissions (> 3 years)., Conclusion: T cells targeting five TAAs and administered at doses of up to two infusions of 2 × 107 cells/m2 are well-tolerated by patients with lymphoma both as adjuvant and to treat chemorefractory lymphoma. Preliminary indicators of antilymphoma activity were seen in the chemorefractory cohort across both antigen- and dose-escalation phases.- Published
- 2021
- Full Text
- View/download PDF
23. A Phase II study of autologous mesenchymal stromal cells and c-kit positive cardiac cells, alone or in combination, in patients with ischaemic heart failure: the CCTRN CONCERT-HF trial.
- Author
-
Bolli R, Mitrani RD, Hare JM, Pepine CJ, Perin EC, Willerson JT, Traverse JH, Henry TD, Yang PC, Murphy MP, March KL, Schulman IH, Ikram S, Lee DP, O'Brien C, Lima JA, Ostovaneh MR, Ambale-Venkatesh B, Lewis G, Khan A, Bacallao K, Valasaki K, Longsomboon B, Gee AP, Richman S, Taylor DA, Lai D, Sayre SL, Bettencourt J, Vojvodic RW, Cohen ML, Simpson L, Aguilar D, Loghin C, Moyé L, Ebert RF, Davis BR, and Simari RD
- Subjects
- Humans, Minnesota, Quality of Life, Stroke Volume, Treatment Outcome, Ventricular Function, Left, Heart Failure, Mesenchymal Stem Cell Transplantation, Mesenchymal Stem Cells
- Abstract
Aims: CONCERT-HF is an NHLBI-sponsored, double-blind, placebo-controlled, Phase II trial designed to determine whether treatment with autologous bone marrow-derived mesenchymal stromal cells (MSCs) and c-kit positive cardiac cells (CPCs), given alone or in combination, is feasible, safe, and beneficial in patients with heart failure (HF) caused by ischaemic cardiomyopathy., Methods and Results: Patients were randomized (1:1:1:1) to transendocardial injection of MSCs combined with CPCs, MSCs alone, CPCs alone, or placebo, and followed for 12 months. Seven centres enrolled 125 participants with left ventricular ejection fraction of 28.6 ± 6.1% and scar size 19.4 ± 5.8%, in New York Heart Association class II or III. The proportion of major adverse cardiac events (MACE) was significantly decreased by CPCs alone (-22% vs. placebo, P = 0.043). Quality of life (Minnesota Living with Heart Failure Questionnaire score) was significantly improved by MSCs alone (P = 0.050) and MSCs + CPCs (P = 0.023) vs. placebo. Left ventricular ejection fraction, left ventricular volumes, scar size, 6-min walking distance, and peak oxygen consumption did not differ significantly among groups., Conclusions: This is the first multicentre trial assessing CPCs and a combination of two cell types from different tissues in HF patients. The results show that treatment is safe and feasible. Even with maximal guideline-directed therapy, both CPCs and MSCs were associated with improved clinical outcomes (MACE and quality of life, respectively) in ischaemic HF without affecting left ventricular function or structure, suggesting possible systemic or paracrine cellular mechanisms. Combining MSCs with CPCs was associated with improvement in both these outcomes. These results suggest potential important beneficial effects of CPCs and MSCs and support further investigation in HF patients., (© 2021 European Society of Cardiology.)
- Published
- 2021
- Full Text
- View/download PDF
24. Two Decades of Global Progress in Authorized Advanced Therapy Medicinal Products: An Emerging Revolution in Therapeutic Strategies.
- Author
-
Ramezankhani R, Torabi S, Minaei N, Madani H, Rezaeiani S, Hassani SN, Gee AP, Dominici M, Silva DN, Baharvand H, and Hajizadeh-Saffar E
- Abstract
The introduction of advanced therapy medicinal products (ATMPs) to the global pharma market has been revolutionizing the pharmaceutical industry and has opened new routes for treating various types of cancers and incurable diseases. In the past two decades, a noticeable part of clinical practices has been devoting progressively to these products. The first step to develop such an ATMP product is to be familiar with other approved products to obtain a general view about this industry trend. The present paper depicts an overall perspective of approved ATMPs in different countries, while reflecting the degree of their success in a clinical point of view and highlighting their main safety issues and also related market size as a whole. In this regard, published articles regarding safety, efficacy, and market size of approved ATMPs were reviewed using the search engines PubMed, Scopus, and Google Scholar. For some products which the related papers were not available, data on the relevant company website were referenced. In this descriptive study, we have introduced and classified approved cell, gene, and tissue engineering-based products by different regulatory agencies, along with their characteristics, manufacturer, indication, approval date, related regulatory agency, dosage, product description, price and published data about their safety and efficacy. In addition, to gain insights about the commercial situation of each product, we have gathered accessible sale reports and market size information that pertain to some of these products., 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 © 2020 Ramezankhani, Torabi, Minaei, Madani, Rezaeiani, Hassani, Gee, Dominici, Silva, Baharvand and Hajizadeh-Saffar.)
- Published
- 2020
- Full Text
- View/download PDF
25. Anti-CD30 CAR-T Cell Therapy in Relapsed and Refractory Hodgkin Lymphoma.
- Author
-
Ramos CA, Grover NS, Beaven AW, Lulla PD, Wu MF, Ivanova A, Wang T, Shea TC, Rooney CM, Dittus C, Park SI, Gee AP, Eldridge PW, McKay KL, Mehta B, Cheng CJ, Buchanan FB, Grilley BJ, Morrison K, Brenner MK, Serody JS, Dotti G, Heslop HE, and Savoldo B
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bendamustine Hydrochloride administration & dosage, Bendamustine Hydrochloride therapeutic use, Combined Modality Therapy, Cyclophosphamide administration & dosage, Epitopes, Female, Hodgkin Disease drug therapy, Hodgkin Disease immunology, Humans, Immunotherapy, Adoptive adverse effects, Ki-1 Antigen antagonists & inhibitors, Lymphocyte Depletion, Male, Middle Aged, Stem Cell Transplantation methods, Vidarabine administration & dosage, Vidarabine analogs & derivatives, Young Adult, Hodgkin Disease therapy, Immunotherapy, Adoptive methods, Ki-1 Antigen immunology
- Abstract
Purpose: Chimeric antigen receptor (CAR) T-cell therapy of B-cell malignancies has proved to be effective. We show how the same approach of CAR T cells specific for CD30 (CD30.CAR-Ts) can be used to treat Hodgkin lymphoma (HL)., Methods: We conducted 2 parallel phase I/II studies (ClinicalTrials.gov identifiers: NCT02690545 and NCT02917083) at 2 independent centers involving patients with relapsed or refractory HL and administered CD30.CAR-Ts after lymphodepletion with either bendamustine alone, bendamustine and fludarabine, or cyclophosphamide and fludarabine. The primary end point was safety., Results: Forty-one patients received CD30.CAR-Ts. Treated patients had a median of 7 prior lines of therapy (range, 2-23), including brentuximab vedotin, checkpoint inhibitors, and autologous or allogeneic stem cell transplantation. The most common toxicities were grade 3 or higher hematologic adverse events. Cytokine release syndrome was observed in 10 patients, all of which were grade 1. No neurologic toxicity was observed. The overall response rate in the 32 patients with active disease who received fludarabine-based lymphodepletion was 72%, including 19 patients (59%) with complete response. With a median follow-up of 533 days, the 1-year progression-free survival and overall survival for all evaluable patients were 36% (95% CI, 21% to 51%) and 94% (95% CI, 79% to 99%), respectively. CAR-T cell expansion in vivo was cell dose dependent., Conclusion: Heavily pretreated patients with relapsed or refractory HL who received fludarabine-based lymphodepletion followed by CD30.CAR-Ts had a high rate of durable responses with an excellent safety profile, highlighting the feasibility of extending CAR-T cell therapies beyond canonical B-cell malignancies.
- Published
- 2020
- Full Text
- View/download PDF
26. Allogeneic Mesenchymal Cell Therapy in Anthracycline-Induced Cardiomyopathy Heart Failure Patients: The CCTRN SENECA Trial.
- Author
-
Bolli R, Perin EC, Willerson JT, Yang PC, Traverse JH, Henry TD, Pepine CJ, Mitrani RD, Hare JM, Murphy MP, March KL, Ikram S, Lee DP, O'Brien C, Durand JB, Miller K, Lima JA, Ostovaneh MR, Ambale-Venkatesh B, Gee AP, Richman S, Taylor DA, Sayre SL, Bettencourt J, Vojvodic RW, Cohen ML, Simpson LM, Lai D, Aguilar D, Loghin C, Moyé L, Ebert RF, Davis BR, and Simari RD
- Abstract
Background: Anthracycline-induced cardiomyopathy (AIC) may be irreversible with a poor prognosis, disproportionately affecting women and young adults. Administration of allogeneic bone marrow-derived mesenchymal stromal cells (allo-MSCs) is a promising approach to heart failure (HF) treatment., Objectives: SENECA (Stem Cell Injection in Cancer Survivors) was a phase 1 study of allo-MSCs in AIC., Methods: Cancer survivors with chronic AIC (mean age 56.6 years; 68% women; NT-proBNP 1,426 pg/ml; 6 enrolled in an open-label, lead-in phase and 31 subjects randomized 1:1) received 1 × 10
8 allo-MSCs or vehicle transendocardially. Primary objectives were safety and feasibility. Secondary efficacy measures included cardiac function and structure measured by cardiac magnetic resonance imaging (CMR), functional capacity, quality of life (Minnesota Living with Heart Failure Questionnaire), and biomarkers., Results: A total of 97% of subjects underwent successful study product injections; all allo-MSC-assigned subjects received the target dose of cells. Follow-up visits were well-attended (92%) with successful collection of endpoints in 94% at the 1-year visit. Although 58% of subjects had non-CMR compatible devices, CMR endpoints were successfully collected in 84% of subjects imaged at 1 year. No new tumors were reported. There were no significant differences between allo-MSC and vehicle groups with regard to clinical outcomes. Secondary measures included 6-min walk test (p = 0.056) and Minnesota Living with Heart Failure Questionnaire score (p = 0.048), which tended to favor the allo-MSC group., Conclusions: In this first-in-human study of cell therapy in patients with AIC, transendocardial administration of allo-MSCs appears safe and feasible, and CMR was successfully performed in the majority of the HF patients with devices. This study lays the groundwork for phase 2 trials aimed at assessing efficacy of cell therapy in patients with AIC.- Published
- 2020
- Full Text
- View/download PDF
27. The safety and clinical effects of administering a multiantigen-targeted T cell therapy to patients with multiple myeloma.
- Author
-
Lulla PD, Tzannou I, Vasileiou S, Carrum G, Ramos CA, Kamble R, Wang T, Wu M, Bilgi M, Gee AP, Mukhi S, Chung B, Wang L, Watanabe A, Kuvalekar M, Jeong M, Li Y, Ketkar S, French-Kim M, Grilley B, Brenner MK, Heslop HE, Vera JF, and Leen AM
- Subjects
- Antigens, Neoplasm, Cell- and Tissue-Based Therapy, Humans, Neoplasm Recurrence, Local, Receptors, Antigen, T-Cell, Multiple Myeloma therapy
- Abstract
Multiple myeloma (MM) is an almost always incurable malignancy of plasma cells. Despite the advent of new therapies, most patients eventually relapse or become treatment-refractory. Consequently, therapies with nonoverlapping mechanisms of action that are nontoxic and provide long-term benefit to patients with MM are greatly needed. To this end, we clinically tested an autologous multitumor-associated antigen (mTAA)-specific T cell product for the treatment of patients with high-risk, relapsed or refractory MM. In this study, we expanded polyclonal T cells from 23 patients with MM. T cells whose native T cell receptors were reactive toward five myeloma-expressed target TAAs (PRAME, SSX2, MAGEA4, Survivin, and NY-ESO-1) were enriched ex vivo. To date, we have administered escalating doses of these nonengineered mTAA-specific T cells (0.5 × 10
7 to 2 × 107 cells/m2 ) to 21 patients with MM, 9 of whom were at high risk of relapse after a median of 3 lines of prior therapy and 12 with active, relapsed or refractory disease after a median of 3.5 prior lines. The cells were well tolerated, with only two transient, grade III infusion-related adverse events. Furthermore, patients with active relapsed or refractory myeloma enjoyed a longer than expected progression-free survival and responders included three patients who achieved objective responses concomitant with detection of functional TAA-reactive T cell clonotypes derived from the infused mTAA product., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2020
- Full Text
- View/download PDF
28. T-Cell Receptor Stimulation Enhances the Expansion and Function of CD19 Chimeric Antigen Receptor-Expressing T Cells.
- Author
-
Lapteva N, Gilbert M, Diaconu I, Rollins LA, Al-Sabbagh M, Naik S, Krance RA, Tripic T, Hiregange M, Raghavan D, Dakhova O, Rouce RH, Liu H, Omer B, Savoldo B, Dotti G, Cruz CR, Sharpe K, Gates M, Orozco A, Durett A, Pacheco E, Gee AP, Ramos CA, Heslop HE, Brenner MK, and Rooney CM
- Subjects
- Adenoviridae physiology, Adolescent, Antigens, CD19 metabolism, Child, Child, Preschool, Genetic Vectors, Herpesvirus 4, Human physiology, Humans, Lymphoma, B-Cell immunology, Lymphoma, B-Cell virology, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma virology, Retroviridae physiology, T-Lymphocytes immunology, T-Lymphocytes virology, Young Adult, Antigens, CD19 immunology, Immunotherapy, Adoptive methods, Lymphoma, B-Cell therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Antigen, T-Cell immunology, T-Lymphocytes transplantation
- Abstract
Purpose: Current protocols for CD19 chimeric antigen receptor-expressing T cells (CD19.CAR-T cells) require recipients to tolerate preinfusion cytoreductive chemotherapy, and the presence of sufficient target antigen on normal or malignant B cells., Patients and Methods: We investigated whether additional stimulation of CD19.CAR-T cells through their native receptors can substitute for cytoreductive chemotherapy, inducing expansion and functional persistence of CD19.CAR-T even in patients in remission of B-cell acute lymphocytic leukemia. We infused a low dose of CD19.CAR-modified virus-specific T cells (CD19.CAR-VST) without prior cytoreductive chemotherapy into 8 patients after allogeneic stem cell transplant., Results: Absent virus reactivation, we saw no CD19.CAR-VST expansion. In contrast, in patients with viral reactivation, up to 30,000-fold expansion of CD19.CAR-VSTs was observed, with depletion of CD19
+ B cells. Five patients remain in remission at 42-60+ months., Conclusions: Dual T-cell receptor and CAR stimulation can thus potentiate effector cell expansion and CAR-target cell killing, even when infusing low numbers of effector cells without cytoreduction., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
29. "Mini" bank of only 8 donors supplies CMV-directed T cells to diverse recipients.
- Author
-
Tzannou I, Watanabe A, Naik S, Daum R, Kuvalekar M, Leung KS, Martinez C, Sasa G, Wu M, Gee AP, Krance RA, Gottschalk S, Heslop HE, and Omer B
- Subjects
- Cytomegalovirus Infections therapy, HLA Antigens immunology, Humans, Tissue Banks, Adoptive Transfer methods, Biological Specimen Banks supply & distribution, Cytomegalovirus immunology, T-Lymphocytes immunology, Tissue Donors supply & distribution, Transplant Recipients
- Abstract
Cytomegalovirus (CMV) infections remain a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT), and standard antiviral therapies are associated with significant side effects and development of drug-resistant mutants. Adoptively transferred donor-derived CMV-specific T cells (CMVSTs) can provide an alternative treatment modality with few side effects but are not widely available due to their patient-specific nature. Here we report the establishment and use of a bank of CMVSTs derived from just 8 CMV-seropositive donors, with HLA types representing the diverse US population, as an "off-the-shelf" therapy to treat drug-refractory infections. To date, we have screened 29 patients for study participation and identified a suitable line, with ≥2 of 8 shared HLA antigens, for 28 (96.6%) patients with a median of 4 shared HLA antigens. Of these, 10 patients with persistent/refractory CMV infections or disease were eligible for treatment; a single infusion of cells produced 3 partial responses and 7 complete responses, for a cumulative response rate of 100% (95% confidence interval, 69.2-100) with no graft-versus-host disease, graft failure, or cytokine release syndrome. Potential wider use of the tested CMVSTs across transplant centers is made more feasible by our ability to produce sufficient material to generate cells for >2000 infusions from a single donor collection. Our data indicate that a "mini" bank of CMVSTs prepared from just 8 well-chosen third-party donors can supply the majority of patients with an appropriately matched line that produces safe and effective anti-CMV activity post-HSCT., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
30. Safety and feasibility of virus-specific T cells derived from umbilical cord blood in cord blood transplant recipients.
- Author
-
Abraham AA, John TD, Keller MD, Cruz CRN, Salem B, Roesch L, Liu H, Hoq F, Grilley BJ, Gee AP, Dave H, Jacobsohn DA, Krance RA, Shpall EJ, Martinez CA, Hanley PJ, and Bollard CM
- Abstract
Adoptive transfer of virus-specific T cells (VSTs) has been shown to be safe and effective in stem cell transplant recipients. However, the lack of virus-experienced T cells in donor cord blood (CB) has prevented the development of ex vivo expanded donor-derived VSTs for recipients of this stem cell source. Here we evaluated the feasibility and safety of ex vivo expansion of CB T cells from the 20% fraction of the CB unit in pediatric patients receiving a single CB transplant (CBT). In 2 clinical trials conducted at 2 separate sites, we manufactured CB-derived multivirus-specific T cells (CB-VSTs) targeting Epstein-Barr virus (EBV), adenovirus, and cytomegalovirus (CMV) for 18 (86%) of 21 patients demonstrating feasibility. Manufacturing for 2 CB-VSTs failed to meet lot release because of insufficient cell recovery, and there was 1 sterility breach during separation of the frozen 20% fraction. Delayed engraftment was not observed in patients who received the remaining 80% fraction for the primary CBT. There was no grade 3 to 4 acute graft-versus-host disease (GVHD) associated with the infusion of CB-VSTs. None of the 7 patients who received CB-VSTs as prophylaxis developed end-organ disease from CMV, EBV, or adenovirus. In 7 patients receiving CB-VSTs for viral reactivation or infection, only 1 patient developed end-organ viral disease, which was in an immune privileged site (CMV retinitis) and occurred after steroid therapy for GVHD. Finally, we demonstrated the long-term persistence of adoptively transferred CB-VSTs using T-cell receptor-Vβ clonotype tracking, suggesting that CB-VSTs are a feasible addition to antiviral pharmacotherapy., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
31. In Vivo Fate and Activity of Second- versus Third-Generation CD19-Specific CAR-T Cells in B Cell Non-Hodgkin's Lymphomas.
- Author
-
Ramos CA, Rouce R, Robertson CS, Reyna A, Narala N, Vyas G, Mehta B, Zhang H, Dakhova O, Carrum G, Kamble RT, Gee AP, Mei Z, Wu MF, Liu H, Grilley B, Rooney CM, Heslop HE, Brenner MK, Savoldo B, and Dotti G
- Subjects
- Aged, Combined Modality Therapy, Female, Hematopoietic Stem Cell Transplantation, Humans, Lymphoma, Non-Hodgkin diagnosis, Male, Middle Aged, Positron Emission Tomography Computed Tomography, Receptors, Antigen, T-Cell genetics, Receptors, Chimeric Antigen genetics, T-Lymphocytes metabolism, Transplantation, Autologous, Treatment Outcome, Antigens, CD19 immunology, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Lymphoma, Non-Hodgkin immunology, Lymphoma, Non-Hodgkin therapy, Receptors, Antigen, T-Cell metabolism, Receptors, Chimeric Antigen metabolism, T-Lymphocytes immunology
- Abstract
Second-generation (2G) chimeric antigen receptors (CARs) targeting CD19 are highly active against B cell malignancies, but it is unknown whether any of the costimulatory domains incorporated in the CAR have superior activity to others. Because CD28 and 4-1BB signaling activate different pathways, combining them in a single third-generation (3G) CAR may overcome the limitations of each individual costimulatory domain. We designed a clinical trial in which two autologous CD19-specific CAR-transduced T cell products (CD19.CARTs), 2G (with CD28 only) and 3G (CD28 and 4-1BB), were infused simultaneously in 16 patients with relapsed or refractory non-Hodgkin's lymphoma. 3G CD19.CARTs had superior expansion and longer persistence than 2G CD19.CARTs. This difference was most striking in the five patients with low disease burden and few circulating normal B cells, in whom 2G CD19.CARTs had limited expansion and persistence and correspondingly reduced area under the curve. Of the 11 patients with measurable disease, three achieved complete responses and three had partial responses. Cytokine release syndrome occurred in six patients but was mild, and no patient required anti-IL-6 therapy. Hence, 3G CD19.CARTs combining 4-1BB with CD28 produce superior CART expansion and may be of particular value when treating low disease burden in patients whose normal B cells are depleted by prior therapy., (Copyright © 2018 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
32. EBV/LMP-specific T cells maintain remissions of T- and B-cell EBV lymphomas after allogeneic bone marrow transplantation.
- Author
-
McLaughlin LP, Rouce R, Gottschalk S, Torrano V, Carrum G, Wu MF, Hoq F, Grilley B, Marcogliese AM, Hanley PJ, Gee AP, Brenner MK, Rooney CM, Heslop HE, and Bollard CM
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Epstein-Barr Virus Infections immunology, Female, Herpesvirus 4, Human isolation & purification, Humans, Lymphoma, B-Cell immunology, Lymphoma, B-Cell virology, Lymphoma, T-Cell immunology, Lymphoma, T-Cell virology, Male, Middle Aged, Neoplasm Recurrence, Local immunology, T-Lymphocytes immunology, Transplantation, Homologous methods, Treatment Outcome, Viral Matrix Proteins immunology, Young Adult, Epstein-Barr Virus Infections complications, Hematopoietic Stem Cell Transplantation methods, Herpesvirus 4, Human immunology, Lymphoma, B-Cell therapy, Lymphoma, T-Cell therapy, Neoplasm Recurrence, Local prevention & control, T-Lymphocytes transplantation
- Abstract
Autologous T cells targeting Epstein-Barr virus (EBV) latent membrane proteins (LMPs) have shown safety and efficacy in the treatment of patients with type 2 latency EBV-associated lymphomas for whom standard therapies have failed, including high-dose chemotherapy followed by autologous stem-cell rescue. However, the safety and efficacy of allogeneic donor-derived LMP-specific T cells (LMP-Ts) have not been established for patients who have undergone allogeneic hematopoietic stem-cell transplantation (HSCT). Therefore, we evaluated the safety and efficacy of donor-derived LMP-Ts in 26 patients who had undergone allogeneic HSCT for EBV-associated natural killer/T-cell or B-cell lymphomas. Seven patients received LMP-Ts as therapy for active disease, and 19 were treated with adjuvant therapy for high-risk disease. There were no immediate infusion-related toxicities, and only 1 dose-limiting toxicity potentially related to T-cell infusion was seen. The 2-year overall survival (OS) was 68%. Additionally, patients who received T-cell therapy while in complete remission after allogeneic HSCT had a 78% OS at 2 years. Patients treated for B-cell disease (n = 10) had a 2-year OS of 80%. Patients with T-cell disease had a 2-year OS of 60%, which suggests an improvement compared with published posttransplantation 2-year OS rates of 30% to 50%. Hence, this study shows that donor-derived LMP-Ts are a safe and effective therapy to prevent relapse after transplantation in patients with B cell- or T cell-derived EBV-associated lymphoma or lymphoproliferative disorder and supports the infusion of LMP-Ts as adjuvant therapy to improve outcomes in the posttransplantation setting. These trials were registered at www.clinicaltrials.gov as #NCT00062868 and #NCT01956084., (© 2018 by The American Society of Hematology.)
- Published
- 2018
- Full Text
- View/download PDF
33. Rationale and Design of the SENECA (StEm cell iNjECtion in cAncer survivors) Trial.
- Author
-
Bolli R, Hare JM, Henry TD, Lenneman CG, March KL, Miller K, Pepine CJ, Perin EC, Traverse JH, Willerson JT, Yang PC, Gee AP, Lima JA, Moyé L, Vojvodic RW, Sayre SL, Bettencourt J, Cohen M, Ebert RF, and Simari RD
- Subjects
- Adolescent, Adult, Aged, Anthracyclines therapeutic use, Double-Blind Method, Feasibility Studies, Female, Follow-Up Studies, Heart Failure chemically induced, Heart Failure physiopathology, Humans, Male, Middle Aged, Transplantation, Autologous, Treatment Outcome, Young Adult, Anthracyclines adverse effects, Cancer Survivors statistics & numerical data, Heart Failure surgery, Mesenchymal Stem Cell Transplantation methods, Neoplasms drug therapy, Quality of Life, Ventricular Function, Left physiology
- Abstract
Objectives: SENECA (StEm cell iNjECtion in cAncer survivors) is a phase I, randomized, double-blind, placebo-controlled study to evaluate the safety and feasibility of delivering allogeneic mesenchymal stromal cells (allo-MSCs) transendocardially in subjects with anthracycline-induced cardiomyopathy (AIC)., Background: AIC is an incurable and often fatal syndrome, with a prognosis worse than that of ischemic or nonischemic cardiomyopathy. Recently, cell therapy with MSCs has emerged as a promising new approach to repair damaged myocardium., Methods: The study population is 36 cancer survivors with a diagnosis of AIC, left ventricular (LV) ejection fraction ≤40%, and symptoms of heart failure (NYHA class II-III) on optimally-tolerated medical therapy. Subjects must be clinically free of cancer for at least two years with a ≤ 30% estimated five-year risk of recurrence. The first six subjects participated in an open-label, lead-in phase and received 100 million allo-MSCs; the remaining 30 will be randomized 1:1 to receive allo-MSCs or vehicle via 20 transendocardial injections. Efficacy measures (obtained at baseline, 6 months, and 12 months) include MRI evaluation of LV function, LV volumes, fibrosis, and scar burden; assessment of exercise tolerance (six-minute walk test) and quality of life (Minnesota Living with Heart Failure Questionnaire); clinical outcomes (MACE and cumulative days alive and out of hospital); and biomarkers of heart failure (NT-proBNP)., Conclusions: This is the first clinical trial using direct cardiac injection of cells for the treatment of AIC. If administration of allo-MSCs is found feasible and safe, SENECA will pave the way for larger phase II/III studies with therapeutic efficacy as the primary outcome., (Copyright © 2018. Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
34. GMP CAR-T cell production.
- Author
-
Gee AP
- Subjects
- Antigens, CD19 biosynthesis, Humans, Receptors, Chimeric Antigen metabolism, T-Lymphocytes metabolism, Antigens, CD19 immunology, Immunotherapy, Adoptive, Receptors, Chimeric Antigen immunology, T-Lymphocytes immunology
- Abstract
The clinical success achieved using CD19-directed CAR-T cells has stimulated many academic institutions to explore the feasibility of manufacturing these, and other CAR-T cells, in-house. This article reviews the issues that must be addressed in order to achieve this goal. It includes the manufacturing infrastructure, the regulatory environment, practical aspects of production, and the costs involved., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
35. Tumor-Specific T-Cells Engineered to Overcome Tumor Immune Evasion Induce Clinical Responses in Patients With Relapsed Hodgkin Lymphoma.
- Author
-
Bollard CM, Tripic T, Cruz CR, Dotti G, Gottschalk S, Torrano V, Dakhova O, Carrum G, Ramos CA, Liu H, Wu MF, Marcogliese AN, Barese C, Zu Y, Lee DY, O'Connor O, Gee AP, Brenner MK, Heslop HE, and Rooney CM
- Subjects
- Adult, Cell Proliferation, Cells, Cultured, Female, Genetic Therapy adverse effects, Herpesvirus 4, Human immunology, Herpesvirus 4, Human metabolism, Hodgkin Disease immunology, Hodgkin Disease metabolism, Hodgkin Disease virology, Humans, Immunotherapy, Adoptive adverse effects, Lymphocyte Activation, Male, Middle Aged, Phenotype, Receptor, Transforming Growth Factor-beta Type II immunology, Receptor, Transforming Growth Factor-beta Type II metabolism, Recurrence, T-Lymphocytes immunology, T-Lymphocytes metabolism, Time Factors, Treatment Outcome, Tumor Microenvironment, Viral Matrix Proteins immunology, Viral Matrix Proteins metabolism, Genetic Therapy methods, Hodgkin Disease therapy, Immunotherapy, Adoptive instrumentation, Receptor, Transforming Growth Factor-beta Type II genetics, T-Lymphocytes transplantation, Tumor Escape
- Abstract
Purpose Transforming growth factor-β (TGF-β) production in the tumor microenvironment is a potent and ubiquitous tumor immune evasion mechanism that inhibits the expansion and function of tumor-directed responses; therefore, we conducted a clinical study to discover the effects of the forced expression of a dominant-negative TGF-β receptor type 2 (DNRII) on the safety, survival, and activity of infused tumor-directed T cells. Materials and Methods In a dose escalation study, eight patients with Epstein Barr virus-positive Hodgkin lymphoma received two to 12 doses of between 2 × 10
7 and 1.5 × 108 cells/m2 of DNRII-expressing T cells with specificity for the Epstein Barr virus-derived tumor antigens, latent membrane protein (LMP)-1 and LMP-2 (DNRII-LSTs). Lymphodepleting chemotherapy was not used before infusion. Results DNRII-LSTs were resistant to otherwise inhibitory concentrations of TGF-β in vitro and retained their tumor antigen-specific activity. After infusion, the signal from transgenic T cells in peripheral blood increased up to 100-fold as measured by quantitative polymerase chain reaction for the transgene, with a corresponding increase in the frequency of functional LMP-specific T cells. Expansion was not associated with any acute or long-term toxicity. DNRII-LSTs persisted for up to ≥ 4 years. Four of the seven evaluable patients with active disease achieved clinical responses that were complete and ongoing in two patients at > 4 years, including in one patient who achieved only a partial response to unmodified tumor-directed T cells. Conclusion TGF-β-resistant tumor-specific T cells safely expand and persist in patients with Hodgkin lymphoma without lymphodepleting chemotherapy before infusion. DNRII-LSTs can induce complete responses even in patients with resistant disease. Expression of DNRII may be useful for the many other tumors that exploit this potent immune evasion mechanism.- Published
- 2018
- Full Text
- View/download PDF
36. TIME Trial: Effect of Timing of Stem Cell Delivery Following ST-Elevation Myocardial Infarction on the Recovery of Global and Regional Left Ventricular Function: Final 2-Year Analysis.
- Author
-
Traverse JH, Henry TD, Pepine CJ, Willerson JT, Chugh A, Yang PC, Zhao DXM, Ellis SG, Forder JR, Perin EC, Penn MS, Hatzopoulos AK, Chambers JC, Baran KW, Raveendran G, Gee AP, Taylor DA, Moyé L, Ebert RF, and Simari RD
- Subjects
- Adult, Aged, Double-Blind Method, Female, Follow-Up Studies, Heart Ventricles pathology, Humans, Magnetic Resonance Imaging, Male, Microcirculation, Middle Aged, Organ Size, ST Elevation Myocardial Infarction complications, ST Elevation Myocardial Infarction pathology, Stroke Volume, Time Factors, Ventricular Dysfunction, Left etiology, Bone Marrow Transplantation methods, ST Elevation Myocardial Infarction therapy, Ventricular Dysfunction, Left therapy
- Abstract
Rationale: The TIME trial (Timing in Myocardial Infarction Evaluation) was the first cell therapy trial sufficiently powered to determine if timing of cell delivery after ST-segment-elevation myocardial infarction affects recovery of left ventricular (LV) function., Objective: To report the 2-year clinical and cardiac magnetic resonance imaging results and their modification by microvascular obstruction., Methods and Results: TIME was a randomized, double-blind, placebo-controlled trial comparing 150 million bone marrow mononuclear cells versus placebo in 120 patients with anterior ST-segment-elevation myocardial infarctions resulting in LV dysfunction. Primary end points included changes in global (LV ejection fraction) and regional (infarct and border zone) function. Secondary end points included changes in LV volumes, infarct size, and major adverse cardiac events. Here, we analyzed the continued trajectory of these measures out to 2 years and the influence of microvascular obstruction present at baseline on these long-term outcomes. At 2 years (n=85), LV ejection fraction was similar in the bone marrow mononuclear cells (48.7%) and placebo groups (51.6%) with no difference in regional LV function. Infarct size and LV mass decreased ≥30% in each group at 6 months and declined gradually to 2 years. LV volumes increased ≈10% at 6 months and remained stable to 2 years. Microvascular obstruction was present in 48 patients at baseline and was associated with significantly larger infarct size (56.5 versus 36.2 g), greater adverse LV remodeling, and marked reduction in LV ejection fraction recovery (0.2% versus 6.2%)., Conclusions: In one of the longest serial cardiac magnetic resonance imaging analyses of patients with large anterior ST-segment-elevation myocardial infarctions, bone marrow mononuclear cells administration did not improve recovery of LV function over 2 years. Microvascular obstruction was associated with reduced recovery of LV function, greater adverse LV remodeling, and more device implantations. The use of cardiac magnetic resonance imaging leads to greater dropout of patients over time because of device implantation in patients with more severe LV dysfunction resulting in overestimation of clinical stability of the cohort., Clinical Trial Registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00684021., (© 2017 American Heart Association, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
37. Absence of Replication-Competent Retrovirus in Vectors, T Cell Products, and Patient Follow-Up Samples.
- Author
-
Lyon D, Lapteva N, and Gee AP
- Subjects
- Follow-Up Studies, Humans, Retroviridae genetics, Virus Replication, Genetic Vectors, T-Lymphocytes
- Published
- 2018
- Full Text
- View/download PDF
38. Regulation of Regenerative Medicine Products.
- Author
-
Gee AP
- Subjects
- Animals, Cell- and Tissue-Based Therapy methods, Humans, Investigational New Drug Application, Public Policy trends, Regenerative Medicine methods, Regenerative Medicine standards, Risk Assessment, Tissue Engineering methods, Tissue Engineering standards, United States, United States Food and Drug Administration, Biological Products standards, Cell- and Tissue-Based Therapy standards, Extracellular Matrix, Regenerative Medicine legislation & jurisprudence, Tissue Engineering legislation & jurisprudence
- Abstract
Cellular therapies have moved to the forefront based upon promising results from clinical trials using both chimeric antigen receptor T lymphocytes to treat leukemia and other cell types to restore structure and function to tissues that have been damaged by disease or physical injury. The pace at which these treatments have evolved has posed a regulatory challenge to agencies, such as the Food and Drug Administration (FDA). This chapter describes how a specific regulatory strategy was developed and how it has evolved in response to the demand for these new therapies.
- Published
- 2018
- Full Text
- View/download PDF
39. Enantioselective transformation of fluoxetine in water and its ecotoxicological relevance.
- Author
-
Andrés-Costa MJ, Proctor K, Sabatini MT, Gee AP, Lewis SE, Pico Y, and Kasprzyk-Hordern B
- Subjects
- Biodegradation, Environmental, Fluoxetine analogs & derivatives, Kinetics, Metabolome, Rivers chemistry, Sewage chemistry, Stereoisomerism, Tetrahymena metabolism, Ecotoxicology, Fluoxetine chemistry, Fluoxetine metabolism, Water Pollutants, Chemical chemistry, Water Pollutants, Chemical metabolism
- Abstract
European legislation focusing on water quality is expected to broaden to encompass several pharmaceuticals as priority hazardous substances. This manuscript aims to challenge current regulatory approaches that do not recognize stereochemistry of chiral pharmaceuticals by testing the hypothesis that environmental transformation and effects of chiral pharmaceuticals are stereoselective. Our experiments revealed that, while degradation of chiral fluoxetine (FL) in river water occurs via non-enantioselective photochemical and mildly-enantioselective microbial processes favoring the (R)-enantiomer, a pronounced enantioselectivity favoring (S)-FL (leading to the formation of (S)-NFL (norfluoxetine)) is observed during activated sludge treatment. Toxicity tests proved strong enantiomer-specific toxicity in the case of Tetrahymena thermophila, protozoa that are utilized during activated sludge treatment ((R)-FL is 30× more toxic than (S)-FL; (S)-NFL is 10× more toxic than (S)-FL). This is of paramount importance as preferential degradation of (S)-FL in activated sludge microcosms leads to the enrichment of FL with 30× more toxic (R)-FL and formation of 10× more toxic (S)-NFL. It is commonly assumed that a decreased concentration of FL leads to decreased biological impact. Our study proves that despite the overall decrease in FL concentration, accumulation of toxic (R)-FL and formation of toxic (S)-NFL leads to much higher than presumed toxicological effects.
- Published
- 2017
- Full Text
- View/download PDF
40. Off-the-Shelf Virus-Specific T Cells to Treat BK Virus, Human Herpesvirus 6, Cytomegalovirus, Epstein-Barr Virus, and Adenovirus Infections After Allogeneic Hematopoietic Stem-Cell Transplantation.
- Author
-
Tzannou I, Papadopoulou A, Naik S, Leung K, Martinez CA, Ramos CA, Carrum G, Sasa G, Lulla P, Watanabe A, Kuvalekar M, Gee AP, Wu MF, Liu H, Grilley BJ, Krance RA, Gottschalk S, Brenner MK, Rooney CM, Heslop HE, Leen AM, and Omer B
- Subjects
- Adenoviruses, Human immunology, Adult, BK Virus immunology, DNA Virus Infections etiology, DNA Virus Infections virology, Female, Herpesvirus 4, Human immunology, Herpesvirus 6, Human immunology, Humans, Male, Transplantation, Homologous, Treatment Outcome, DNA Virus Infections therapy, DNA Viruses immunology, Hematopoietic Stem Cell Transplantation adverse effects, Hematopoietic Stem Cell Transplantation methods, Immunotherapy, Adoptive methods, T-Lymphocytes immunology, T-Lymphocytes transplantation
- Abstract
Purpose Improvement of cure rates for patients treated with allogeneic hematopoietic stem-cell transplantation (HSCT) will require efforts to decrease treatment-related mortality from severe viral infections. Adoptively transferred virus-specific T cells (VSTs) generated from eligible, third-party donors could provide broad antiviral protection to recipients of HSCT as an immediately available off-the-shelf product. Patient and Methods We generated a bank of VSTs that recognized five common viral pathogens: Epstein-Barr virus (EBV), adenovirus (AdV), cytomegalovirus (CMV), BK virus (BKV), and human herpesvirus 6 (HHV-6). The VSTs were administered to 38 patients with 45 infections in a phase II clinical trial. Results A single infusion produced a cumulative complete or partial response rate of 92% (95% CI, 78.1% to 98.3%) overall and the following rates by virus: 100% for BKV (n = 16), 94% for CMV (n = 17), 71% for AdV (n = 7), 100% for EBV (n = 2), and 67% for HHV-6 (n = 3). Clinical benefit was achieved in 31 patients treated for one infection and in seven patients treated for multiple coincident infections. Thirteen of 14 patients treated for BKV-associated hemorrhagic cystitis experienced complete resolution of gross hematuria by week 6. Infusions were safe, and only two occurrences of de novo graft-versus host disease (grade 1) were observed. VST tracking by epitope profiling revealed persistence of functional VSTs of third-party origin for up to 12 weeks. Conclusion The use of banked VSTs is a feasible, safe, and effective approach to treat severe and drug-refractory infections after HSCT, including infections from two viruses (BKV and HHV-6) that had never been targeted previously with an off-the-shelf product. Furthermore, the multispecificity of the VSTs ensures extensive antiviral coverage, which facilitates the treatment of patients with multiple infections.
- Published
- 2017
- Full Text
- View/download PDF
41. Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes.
- Author
-
Ramos CA, Ballard B, Zhang H, Dakhova O, Gee AP, Mei Z, Bilgi M, Wu MF, Liu H, Grilley B, Bollard CM, Chang BH, Rooney CM, Brenner MK, Heslop HE, Dotti G, and Savoldo B
- Subjects
- Adult, Antineoplastic Agents chemistry, Brentuximab Vedotin, CD28 Antigens chemistry, Disease Progression, Dose-Response Relationship, Drug, Female, Hodgkin Disease immunology, Humans, Immunoconjugates administration & dosage, Immunoconjugates therapeutic use, Immunophenotyping, Lymphoma, Large-Cell, Anaplastic immunology, Male, Middle Aged, Molecular Targeted Therapy, Neoplasm Recurrence, Local, Transplantation Conditioning, Treatment Outcome, Young Adult, Hodgkin Disease therapy, Ki-1 Antigen metabolism, Lymphoma, Large-Cell, Anaplastic therapy, Receptors, Antigen, T-Cell chemistry, T-Lymphocytes cytology
- Abstract
Background: Targeting CD30 with monoclonal antibodies in Hodgkin lymphoma (HL) and anaplastic large cell lymphoma (ALCL) has had profound clinical success. However, adverse events, mainly mediated by the toxin component of the conjugated antibodies, cause treatment discontinuation in many patients. Targeting CD30 with T cells expressing a CD30-specific chimeric antigen receptor (CAR) may reduce the side effects and augment antitumor activity., Methods: We conducted a phase I dose escalation study in which 9 patients with relapsed/refractory HL or ALCL were infused with autologous T cells that were gene-modified with a retroviral vector to express the CD30-specific CAR (CD30.CAR-Ts) encoding the CD28 costimulatory endodomain. Three dose levels, from 0.2 × 108 to 2 × 108 CD30.CAR-Ts/m2, were infused without a conditioning regimen. All other therapy for malignancy was discontinued at least 4 weeks before CD30.CAR-T infusion. Seven patients had previously experienced disease progression while being treated with brentuximab., Results: No toxicities attributable to CD30.CAR-Ts were observed. Of 7 patients with relapsed HL, 1 entered complete response (CR) lasting more than 2.5 years after the second infusion of CD30.CAR-Ts, 1 remained in continued CR for almost 2 years, and 3 had transient stable disease. Of 2 patients with ALCL, 1 had a CR that persisted 9 months after the fourth infusion of CD30.CAR-Ts. CD30.CAR-T expansion in peripheral blood peaked 1 week after infusion, and CD30.CAR-Ts remained detectable for over 6 weeks. Although CD30 may also be expressed by normal activated T cells, no patients developed impaired virus-specific immunity., Conclusion: CD30.CAR-Ts are safe and can lead to clinical responses in patients with HL and ALCL, indicating that further assessment of this therapy is warranted., Trial Registration: ClinicalTrials.gov NCT01316146., Funding: National Cancer Institute (3P50CA126752, R01CA131027 and P30CA125123), National Heart, Lung, and Blood Institute (R01HL114564), and Leukemia and Lymphoma Society (LLSTR 6227-08).
- Published
- 2017
- Full Text
- View/download PDF
42. Manufacturing Differences Affect Human Bone Marrow Stromal Cell Characteristics and Function: Comparison of Production Methods and Products from Multiple Centers.
- Author
-
Liu S, de Castro LF, Jin P, Civini S, Ren J, Reems JA, Cancelas J, Nayak R, Shaw G, O'Brien T, McKenna DH, Armant M, Silberstein L, Gee AP, Hei DJ, Hematti P, Kuznetsov SA, Robey PG, and Stroncek DF
- Subjects
- Adiposity, Animals, Cryopreservation methods, Hematopoiesis, Humans, Osteogenesis, Bone Marrow metabolism, Bone Marrow Cells metabolism, Gene Expression Profiling methods, Mesenchymal Stem Cells metabolism
- Abstract
Human bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) are manufactured using many different methods, but little is known about the spectrum of manufacturing methods used and their effects on BMSC characteristics and function. Seven centers using, and one developing, Good Manufacturing Practices (GMP) processes were surveyed as to their production methods. Among the seven centers, all used marrow aspirates as the starting material, but no two centers used the same manufacturing methods. Two to four BMSC lots from each center were compared using global gene expression. Among the twenty-four BMSC lots from the eight centers intra-center transcriptome variability was low and similar among centers. Principal component analysis and unsupervised hierarchical clustering analysis separated all the lots from five centers into five distinct clusters. BMSCs from six of the eight centers were tested for their ability to form bone and support hematopoiesis by in vivo transplantation (defining features of BMSCs). Those from all six centers tested formed bone, but the quantity formed was highly variable and BMSCs from only three centers supported hematopoiesis. These results show that differences in manufacturing resulted in variable BMSC characteristics including their ability to form bone and support hematopoiesis.
- Published
- 2017
- Full Text
- View/download PDF
43. Evaluation of Cell Therapy on Exercise Performance and Limb Perfusion in Peripheral Artery Disease: The CCTRN PACE Trial (Patients With Intermittent Claudication Injected With ALDH Bright Cells).
- Author
-
Perin EC, Murphy MP, March KL, Bolli R, Loughran J, Yang PC, Leeper NJ, Dalman RL, Alexander J, Henry TD, Traverse JH, Pepine CJ, Anderson RD, Berceli S, Willerson JT, Muthupillai R, Gahremanpour A, Raveendran G, Velasquez O, Hare JM, Hernandez Schulman I, Kasi VS, Hiatt WR, Ambale-Venkatesh B, Lima JA, Taylor DA, Resende M, Gee AP, Durett AG, Bloom J, Richman S, G'Sell P, Williams S, Khan F, Gyang Ross E, Santoso MR, Goldman J, Leach D, Handberg E, Cheong B, Piece N, DiFede D, Bruhn-Ding B, Caldwell E, Bettencourt J, Lai D, Piller L, Simpson L, Cohen M, Sayre SL, Vojvodic RW, Moyé L, Ebert RF, Simari RD, and Hirsch AT
- Subjects
- Aged, Aldehyde Dehydrogenase metabolism, Bone Marrow Cells metabolism, Bone Marrow Transplantation, Comorbidity, Exercise, Extremities blood supply, Female, Follow-Up Studies, Humans, Intermittent Claudication therapy, Male, Middle Aged, Perfusion, Peripheral Arterial Disease diagnosis, Peripheral Arterial Disease metabolism, Quality of Life, Risk Factors, Treatment Outcome, Cell- and Tissue-Based Therapy adverse effects, Cell- and Tissue-Based Therapy methods, Peripheral Arterial Disease therapy
- Abstract
Background: Atherosclerotic peripheral artery disease affects 8% to 12% of Americans >65 years of age and is associated with a major decline in functional status, increased myocardial infarction and stroke rates, and increased risk of ischemic amputation. Current treatment strategies for claudication have limitations. PACE (Patients With Intermittent Claudication Injected With ALDH Bright Cells) is a National Heart, Lung, and Blood Institute-sponsored, randomized, double-blind, placebo-controlled, phase 2 exploratory clinical trial designed to assess the safety and efficacy of autologous bone marrow-derived aldehyde dehydrogenase bright (ALDHbr) cells in patients with peripheral artery disease and to explore associated claudication physiological mechanisms., Methods: All participants, randomized 1:1 to receive ALDHbr cells or placebo, underwent bone marrow aspiration and isolation of ALDHbr cells, followed by 10 injections into the thigh and calf of the index leg. The coprimary end points were change from baseline to 6 months in peak walking time (PWT), collateral count, peak hyperemic popliteal flow, and capillary perfusion measured by magnetic resonance imaging, as well as safety., Results: A total of 82 patients with claudication and infrainguinal peripheral artery disease were randomized at 9 sites, of whom 78 had analyzable data (57 male, 21 female patients; mean age, 66±9 years). The mean±SEM differences in the change over 6 months between study groups for PWT (0.9±0.8 minutes; 95% confidence interval [CI] -0.6 to 2.5; P =0.238), collateral count (0.9±0.6 arteries; 95% CI, -0.2 to 2.1; P=0.116), peak hyperemic popliteal flow (0.0±0.4 mL/s; 95% CI, -0.8 to 0.8; P =0.978), and capillary perfusion (-0.2±0.6%; 95% CI, -1.3 to 0.9; P=0.752) were not significant. In addition, there were no significant differences for the secondary end points, including quality-of-life measures. There were no adverse safety outcomes. Correlative relationships between magnetic resonance imaging measures and PWT were not significant. A post hoc exploratory analysis suggested that ALDHbr cell administration might be associated with an increase in the number of collateral arteries (1.5±0.7; 95% CI, 0.1-2.9; P =0.047) in participants with completely occluded femoral arteries., Conclusions: ALDHbr cell administration did not improve PWT or magnetic resonance outcomes, and the changes in PWT were not associated with the anatomic or physiological magnetic resonance imaging end points. Future peripheral artery disease cell therapy investigational trial design may be informed by new anatomic and perfusion insights., Clinical Trial Registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01774097., (© 2017 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
44. Langmuir monolayers composed of single and double tail sulfobetaine lipids.
- Author
-
Hazell G, Gee AP, Arnold T, Edler KJ, and Lewis SE
- Subjects
- Betaine chemistry, Molecular Structure, Pressure, Surface Properties, Betaine analogs & derivatives, Lipids chemistry
- Abstract
Hypothesis: Owing to structural similarities between sulfobetaine lipids and phospholipids it should be possible to form stable Langmuir monolayers from long tail sulfobetaines. By modification of the density of lipid tail group (number of carbon chains) it should also be possible to modulate the two-dimensional phase behaviour of these lipids and thereby compare with that of equivalent phospholipids. Potentially this could enable the use of such lipids for the wide array of applications that currently use phospholipids. The benefit of using sulfobetaine lipids is that they can be synthesised by a one-step reaction from cheap and readily available starting materials and will degrade via different pathways than natural lipids. The molecular architecture of the lipid can be easily modified allowing the design of lipids for specific purposes. In addition the reversal of the charge within the sulfobetaine head group relative to the charge orientation in phospholipids may modify behaviour and thereby allow for novel uses of these surfactants., Experiments: Stable Langmuir monolayers were formed composed of single and double tailed sulfobetaine lipids. Surface pressure-area isotherm, Brewster Angle Microscopy and X-ray and neutron reflectometry measurements were conducted to measure the two-dimensional phase behaviour and out-of-plane structure of the monolayers as a function of molecular area., Findings: Sulfobetaine lipids are able to form stable Langmuir monolayers with two dimensional phase behaviour analogous to that seen for the well-studied phospholipids. Changing the number of carbon tail groups on the lipid from one to two promotes the existence of a liquid condensed phase due to increased Van der Waals interactions between the tail groups. Thus the structure of the monolayers appears to be defined by the relative sizes of the head and tail groups in a predictable way. However, the presence of sub-phase ions has little effect on the monolayer structure, behaviour that is surprisingly different to that seen for phospholipids., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
45. Clinical responses with T lymphocytes targeting malignancy-associated κ light chains.
- Author
-
Ramos CA, Savoldo B, Torrano V, Ballard B, Zhang H, Dakhova O, Liu E, Carrum G, Kamble RT, Gee AP, Mei Z, Wu MF, Liu H, Grilley B, Rooney CM, Brenner MK, Heslop HE, and Dotti G
- Subjects
- Adoptive Transfer, Adult, Aged, Antigens, CD19 immunology, Enzyme-Linked Immunosorbent Assay, Feasibility Studies, Female, Humans, Immunophenotyping, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Lymphoma, Non-Hodgkin immunology, Male, Middle Aged, Remission Induction, Retroviridae metabolism, Treatment Outcome, Immunoglobulin kappa-Chains immunology, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Lymphoma, Non-Hodgkin therapy, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology
- Abstract
Background: Treatment of B cell malignancies with adoptive transfer of T cells with a CD19-specific chimeric antigen receptor (CAR) shows remarkable clinical efficacy. However, long-term persistence of T cells targeting CD19, a pan-B cell marker, also depletes normal B cells and causes severe hypogammaglobulinemia. Here, we developed a strategy to target B cell malignancies more selectively by taking advantage of B cell light Ig chain restriction. We generated a CAR that is specific for the κ light chain (κ.CAR) and therefore recognizes κ-restricted cells and spares the normal B cells expressing the nontargeted λ light chain, thus potentially minimizing humoral immunity impairment., Methods: We conducted a phase 1 clinical trial and treated 16 patients with relapsed or refractory κ+ non-Hodgkin lymphoma/chronic lymphocytic leukemia (NHL/CLL) or multiple myeloma (MM) with autologous T cells genetically modified to express κ.CAR (κ.CARTs). Other treatments were discontinued in 11 of the 16 patients at least 4 weeks prior to T cell infusion. Six patients without lymphopenia received 12.5 mg/kg cyclophosphamide 4 days before κ.CART infusion (0.2 × 108 to 2 × 108 κ.CARTs/m2). No other lymphodepletion was used., Results: κ.CART expansion peaked 1-2 weeks after infusion, and cells remained detectable for more than 6 weeks. Of 9 patients with relapsed NHL or CLL, 2 entered complete remission after 2 and 3 infusions of κ.CARTs, and 1 had a partial response. Of 7 patients with MM, 4 had stable disease lasting 2-17 months. No toxicities attributable to κ.CARTs were observed., Conclusion: κ.CART infusion is feasible and safe and can lead to complete clinical responses., Trial Registration: ClinicalTrials.gov NCT00881920., Funding: National Cancer Institute (NCI) grants 3P50CA126752 and 5P30CA125123 and Leukemia and Lymphoma Society (LLS) Specialized Centers of Research (SCOR) grant 7018.
- Published
- 2016
- Full Text
- View/download PDF
46. International Society for Cellular Therapy perspective on immune functional assays for mesenchymal stromal cells as potency release criterion for advanced phase clinical trials.
- Author
-
Galipeau J, Krampera M, Barrett J, Dazzi F, Deans RJ, DeBruijn J, Dominici M, Fibbe WE, Gee AP, Gimble JM, Hematti P, Koh MB, LeBlanc K, Martin I, McNiece IK, Mendicino M, Oh S, Ortiz L, Phinney DG, Planat V, Shi Y, Stroncek DF, Viswanathan S, Weiss DJ, and Sensebe L
- Subjects
- Biomarkers metabolism, Flow Cytometry methods, Humans, Biological Assay methods, Cell- and Tissue-Based Therapy methods, Mesenchymal Stem Cell Transplantation methods, Mesenchymal Stem Cells cytology
- Abstract
Mesenchymal stromal cells (MSCs) as a pharmaceutical for ailments characterized by pathogenic autoimmune, alloimmune and inflammatory processes now cover the spectrum of early- to late-phase clinical trials in both industry and academic sponsored studies. There is a broad consensus that despite different tissue sourcing and varied culture expansion protocols, human MSC-like cell products likely share fundamental mechanisms of action mediating their anti-inflammatory and tissue repair functionalities. Identification of functional markers of potency and reduction to practice of standardized, easily deployable methods of measurements of such would benefit the field. This would satisfy both mechanistic research as well as development of release potency assays to meet Regulatory Authority requirements for conduct of advanced clinical studies and their eventual registration. In response to this unmet need, the International Society for Cellular Therapy (ISCT) addressed the issue at an international workshop in May 2015 as part of the 21st ISCT annual meeting in Las Vegas. The scope of the workshop was focused on discussing potency assays germane to immunomodulation by MSC-like products in clinical indications targeting immune disorders. We here provide consensus perspective arising from this forum. We propose that focused analysis of selected MSC markers robustly deployed by in vitro licensing and metricized with a matrix of assays should be responsive to requirements from Regulatory Authorities. Workshop participants identified three preferred analytic methods that could inform a matrix assay approach: quantitative RNA analysis of selected gene products; flow cytometry analysis of functionally relevant surface markers and protein-based assay of secretome. We also advocate that potency assays acceptable to the Regulatory Authorities be rendered publicly accessible in an "open-access" manner, such as through publication or database collection., (Copyright © 2015 International Society for Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. Large-Scale Culture and Genetic Modification of Human Natural Killer Cells for Cellular Therapy.
- Author
-
Lapteva N, Parihar R, Rollins LA, Gee AP, and Rooney CM
- Subjects
- Blood Component Removal, Cell Proliferation, Cell Transplantation, Feeder Cells cytology, Feeder Cells immunology, Feeder Cells metabolism, Humans, K562 Cells, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Lymphocyte Activation, Cell Culture Techniques methods, Killer Cells, Natural cytology, Transduction, Genetic
- Abstract
Recent advances in methods for the ex vivo expansion of human natural killer (NK) cells have facilitated the use of these powerful immune cells in clinical protocols. Further, the ability to genetically modify primary human NK cells following rapid expansion allows targeting and enhancement of their immune function. We have successfully adapted an expansion method for primary NK cells from peripheral blood mononuclear cells or from apheresis products in gas permeable rapid expansion devices (G-Rexes). Here, we describe an optimized protocol for rapid and robust NK cell expansion as well as a method for highly efficient retroviral transduction of these ex vivo expanded cells. These methodologies are good manufacturing practice (GMP) compliant and could be used for clinical-grade product manufacturing.
- Published
- 2016
- Full Text
- View/download PDF
48. Quantitative activation suppression assay to evaluate human bone marrow-derived mesenchymal stromal cell potency.
- Author
-
Salem B, Miner S, Hensel NF, Battiwalla M, Keyvanfar K, Stroncek DF, Gee AP, Hanley PJ, Bollard CM, Ito S, and Barrett AJ
- Subjects
- Antibodies immunology, Antibodies pharmacology, Biological Assay, CD28 Antigens immunology, CD3 Complex immunology, CD4-Positive T-Lymphocytes immunology, CD40 Ligand metabolism, Cell Line, Cell Proliferation, Cell Survival immunology, Coculture Techniques, Humans, Bone Marrow Cells cytology, CD4-Positive T-Lymphocytes cytology, Immunosuppression Therapy methods, Interferon-gamma pharmacology, Lymphocyte Activation immunology, Mesenchymal Stem Cells cytology
- Abstract
Background Aims: With the increasing use of cell therapies involving immune modulatory cells, there is a need for a simple standardized method to evaluate and compare the suppressive potency of different cell products. We used the Karpas 299 (K299) cell line as the reference suppressor cell to develop a standardized suppression assay to quantify the immune-modulatory capacity of bone marrow-derived mesenchymal stromal cells (BM-MSCs)., Methods: Healthy donor CD4 T cells were co-cultured with the K299 cell line or with third-party BM-MSCs. After stimulation with anti-CD3/CD28 beads, CD154 activation and proliferation of CD4 T cells were measured to calculate suppression., Results: The K299 cell line reproducibly suppressed both the activation and proliferation of healthy donor CD4 T cells in a dose-dependent manner. A rapid (16-h) assay that was based on activation-suppression was selected for development. In replicate testing, there was an inherent variability of suppression of 11% coefficient of variation between different responder T cells. Suppression by BM-MSCs on different responders correlated with suppression by K299. We therefore used K299 suppression as the reference to define suppression potency of BM-MSCs in K299 Suppression Units. We found that inter-donor variability, passage number, method of manufacture and exposure of BM-MSCs to steroids or interferon-γ all affected BM-MSC potency of suppression., Conclusions: This method provides a platform for standardizing suppressor function to facilitate comparisons between laboratories and for use as a cell product release assay., (Published by Elsevier Inc.)
- Published
- 2015
- Full Text
- View/download PDF
49. Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation.
- Author
-
Zhou X, Dotti G, Krance RA, Martinez CA, Naik S, Kamble RT, Durett AG, Dakhova O, Savoldo B, Di Stasi A, Spencer DM, Lin YF, Liu H, Grilley BJ, Gee AP, Rooney CM, Heslop HE, and Brenner MK
- Subjects
- Adolescent, Child, Child, Preschool, Female, Flow Cytometry, Genes, Transgenic, Suicide, Haplotypes, Hematopoietic Stem Cell Transplantation methods, Humans, Lymphoproliferative Disorders surgery, Male, Middle Aged, Organic Chemicals therapeutic use, Real-Time Polymerase Chain Reaction, Young Adult, Caspase 9 genetics, Graft vs Host Disease prevention & control, Hematopoietic Stem Cell Transplantation adverse effects, T-Lymphocytes transplantation
- Abstract
To test the feasibility of a single T-cell manipulation to eliminate alloreactivity while sparing antiviral and antitumor T cells, we infused 12 haploidentical hematopoietic stem cell transplant patients with increasing numbers of alloreplete haploidentical T cells expressing the inducible caspase 9 suicide gene (iC9-T cells). We determined whether the iC9-T cells produced immune reconstitution and if any resultant graft-versus-host disease (GVHD) could be controlled by administration of a chemical inducer of dimerization (CID; AP1903/Rimiducid). All patients receiving >10(4) alloreplete iC9-T lymphocytes per kilogram achieved rapid reconstitution of immune responses toward 5 major pathogenic viruses and concomitant control of active infections. Four patients received a single AP1903 dose. CID infusion eliminated 85% to 95% of circulating CD3(+)CD19(+) T cells within 30 minutes, with no recurrence of GVHD within 90 days. In one patient, symptoms and signs of GVHD-associated cytokine release syndrome (CRS-hyperpyrexia, high levels of proinflammatory cytokines, and rash) resolved within 2 hours of AP1903 infusion. One patient with varicella zoster virus meningitis and acute GVHD had iC9-T cells present in the cerebrospinal fluid, which were reduced by >90% after CID. Notably, virus-specific T cells recovered even after AP1903 administration and continued to protect against infection. Hence, alloreplete iC9-T cells can reconstitute immunity posttransplant and administration of CID can eliminate them from both peripheral blood and the central nervous system (CNS), leading to rapid resolution of GVHD and CRS. The approach may therefore be useful for the rapid and effective treatment of toxicities associated with infusion of engineered T lymphocytes. This trial was registered at www.clinicaltrials.gov as #NCT01494103., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
50. Manufacturing genetically modified T cells for clinical trials.
- Author
-
Gee AP
- Subjects
- Cell Lineage genetics, Cell Lineage immunology, Clinical Trials as Topic, Genetic Vectors, Humans, Immunotherapy, Adoptive, Neoplasms immunology, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell therapeutic use, T-Lymphocytes transplantation, United States, United States Food and Drug Administration, Cell Engineering, Genetic Engineering, Neoplasms therapy, T-Lymphocytes immunology
- Abstract
Compliance with Food and Drug Administration regulations relating to initiating early phase clinical trials of new cellular therapy products often presents a hurdle to new investigators. One of the biggest obstacles is the requirement to manufacture the therapeutic products under current Good Manufacturing Practices-a system that is usually poorly understood by both basic researchers and clinicians. This article reviews the major points that must be addressed when manufacturing genetically modified T cells for therapeutic use.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.