cellular therapies
Recently Published Documents


TOTAL DOCUMENTS

402
(FIVE YEARS 113)

H-INDEX

28
(FIVE YEARS 5)

2022 ◽  
Vol 20 (1) ◽  
Author(s):  
David F. Stroncek ◽  
Robert P. T. Somerville ◽  
Steven L. Highfill

AbstractThe use of cellular therapies to treat cancer, inherited immune deficiencies, hemoglobinopathies and viral infections is growing rapidly. The increased interest in cellular therapies has led to the development of reagents and closed-system automated instruments for the production of these therapies. For cellular therapy clinical trials involving multiple sites some people are advocating a decentralized model of manufacturing where patients are treated with cells produced using automated instruments at each participating center using a single, centrally held Investigational New Drug Application (IND). Many academic centers are purchasing these automated instruments for point-of-care manufacturing and participation in decentralized multiple center clinical trials. However, multiple site manufacturing requires harmonization of product testing and manufacturing in order to interpret the clinical trial results. Decentralized manufacturing is quite challenging since all centers should use the same manufacturing protocol, the same or comparable in-process and lot release assays and the quality programs from each center must work closely together. Consequently, manufacturing cellular therapies using a decentralized model is in many ways more difficult than manufacturing cells in a single centralized facility. Before an academic center decides to establish a point-of-care cell processing laboratory, they should consider all costs associated with such a program. For many academic cell processing centers, point-of-care manufacturing may not be a good investment.


Stroke ◽  
2022 ◽  
pp. 900-911.e5
Author(s):  
Nikunj Satani ◽  
Kaushik Parsha ◽  
Sean I. Savitz

2022 ◽  
pp. 29-45
Author(s):  
George F. Muschler ◽  
Hannah Simmons ◽  
Venkata Mantripragada ◽  
Nicolas S. Piuzzi

2021 ◽  
Vol 12 ◽  
Author(s):  
Matthieu Roulleaux Dugage ◽  
Elise F. Nassif ◽  
Antoine Italiano ◽  
Rastislav Bahleda

Anti-PD-(L)1 therapies yield a disappointing response rate of 15% across soft-tissue sarcomas, even if some subtypes benefit more than others. The proportions of TAMs and TILs in their tumor microenvironment are variable, and this heterogeneity correlates to histotype. Tumors with a richer CD8+ T cell, M1 macrophage, and CD20+ cells infiltrate have a better prognosis than those infiltrated by M0/M2 macrophages and a high immune checkpoint protein expression. PD-L1 and CD8+ infiltrate seem correlated to response to immune checkpoint inhibitors (ICI), but tertiary lymphoid structures have the best predictive value and have been validated prospectively. Trials for combination therapies are ongoing and focus on the association of ICI with chemotherapy, achieving encouraging results especially with pembrolizumab and doxorubicin at an early stage, or ICI with antiangiogenics. A synergy with oncolytic viruses is seen and intratumoral talimogene laherpavec yields an impressive 35% ORR when associated to pembrolizumab. Adoptive cellular therapies are also of great interest in tumors with a high expression of cancer-testis antigens (CTA), such as synovial sarcomas or myxoid round cell liposarcomas with an ORR ranging from 20 to 50%. It seems crucial to adapt the design of clinical trials to histology. Leiomyosarcomas are characterized by complex genomics but are poorly infiltrated by immune cells and do not benefit from ICI. They should be tested with PIK3CA/AKT inhibition, IDO blockade, or treatments aiming at increasing antigenicity (radiotherapy, PARP inhibitors). DDLPS are more infiltrated and have higher PD-L1 expression, but responses to ICI remain variable across clinical studies. Combinations with MDM2 antagonists or CDK4/6 inhibitors may improve responses for DDLPS. UPS harbor the highest copy number alterations (CNA) and mutation rates, with a rich immune infiltrate containing TLS. They have a promising 15-40% ORR to ICI. Trials for ICB should focus on immune-high UPS. Association of ICI with FGFR inhibitors warrants further exploration in the immune-low group of UPS. Finally translocation-related sarcomas are heterogeneous, and although synovial sarcomas a poorly infiltrated and have a poor response rate to ICI, ASPS largely benefit from ICB monotherapy or its association with antiangiogenics agents. Targeting specific neoantigens through vaccine or adoptive cellular therapies is probably the most promising approach in synovial sarcomas.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3891-3891
Author(s):  
Roni Tamari ◽  
Ioannis Politikos ◽  
David Knorr ◽  
Santosha Vardhana ◽  
Jennifer Young ◽  
...  

Abstract BACKGROUND Cellular therapies (allogeneic hematopoietic cell transplantation, allo-HCT, autologous hematopoietic cell transplantation, auto-HCT, and chimeric antigen receptor T cell therapy, CAR T) render patients severely immunocompromised for extended periods post-therapy. Emerging data suggest reduced immune responses to COVID-19 vaccines among patients with hematologic malignancies, but data for cellular therapy recipients are sparse. We therefore assessed immune responses to mRNA COVID-19 vaccines among patients who underwent cellular therapies at our center to identify predictors of response. PATIENT AND METHODS In this observational prospective study, anti-SARS-CoV-2 spike IgG antibody titers and circulating neutralizing antibodies were measured at 1 and 3 months after the 1 st dose of vaccination. CD4, CD19, mitogen, and IgG levels from patient samples collected prior to initiation of vaccination in a subset of patients were used to assess immune recovery and association with response. A concurrent healthy donor (HD) cohort provided control response rates. RESULTS Allo-HCT (N=149), auto HCT (N=61), and CAR T (N=7) patients vaccinated between 12/22/2020- 2/28/2021 with mRNA vaccines and 69 HD participated in this study. At 3 months, 188 pts (87%) had a positive anti-SARS-CoV-2 spike IgG levels (median 5,379 AU/mL, IQR 451-15,750), and 139 (77%) had a positive neutralization Ab assay (median 93%, IQR 36-96%). All HD (100%) had a positive anti-SARS-CoV-2 spike IgG and a positive neutralization Ab assay with median levels of 8,011 AU/mL (IQR 4573-11,159) and 96% (IQR 78- 96%), respectively. Time from vaccination to cellular therapy was associated with response; 67% of patients vaccinated in the first 12 months post-cellular therapy (N=42) mounted a serologic response, compared with patients vaccinated between 12-24 (89%) (N=45), 24-36 (91%) (N=32) and >36 (93%) (N=98) months post-treatment, p= 0.001 (figure 1). Patients with immune parameters below the recommended threshold for vaccinations post-cellular therapies were also less likely to mount a response (figure 2): CD4+ T-cell count < 200 vs >200 cells/μL, 66% vs 87% (p=0.012); CD19+ B-cell count <50 vs >50 cells/μL; 33% vs 95% (p<0.001), phytohemagglutinin mitogen response <40% vs >40%, 42% vs 89% (p<0.001), and IgG <500 vs >500 mg/dl, 71% vs 91% (p=0.003). Patient age, gender, prior COVID-19 infection, treatment with IVIG, and type of mRNA COVID-19 vaccine were not associated with the likelihood of serologic response. CONCLUSION This largest cohort to date, demonstrates that COVID-19 vaccine responses of cellular therapy recipients are reduced compared to healthy control and response varies based on time interval from cellular therapy and immune function at the time of vaccination, underscoring the importance of monitoring immune status parameters, as well as qualitative measures (neutralizing Ab) of vaccine response, in informing clinical decisions, including the indication for booster vaccines. Figure 1 Figure 1. Disclosures Politikos: Merck: Research Funding; ExcellThera, Inc: Other: Member of DSMB - Uncompensated. Vardhana: Immunai: Membership on an entity's Board of Directors or advisory committees. Perales: Equilium: Honoraria; Cidara: Honoraria; Sellas Life Sciences: Honoraria; Miltenyi Biotec: Honoraria, Other; Celgene: Honoraria; MorphoSys: Honoraria; Takeda: Honoraria; Incyte: Honoraria, Other; Karyopharm: Honoraria; Kite/Gilead: Honoraria, Other; Merck: Honoraria; NexImmune: Honoraria; Novartis: Honoraria, Other; Medigene: Honoraria; Omeros: Honoraria; Servier: Honoraria; Bristol-Myers Squibb: Honoraria; Nektar Therapeutics: Honoraria, Other. Shah: Amgen: Research Funding; Janssen Pharmaceutica: Research Funding.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4805-4805
Author(s):  
Jose V. Forero ◽  
Eider F. Moreno Cortes ◽  
Juan Esteban Garcia Robledo ◽  
Natalie Booth ◽  
Januario E Castro

Abstract Introduction: CAR T-cell therapy has revolutionized the treatment of patients with relapsed/refractory (R/R) acute leukemia, NHL, and multiple myeloma. However, there are still areas of improvement in their clinical activity, source of the effector cells, prevention, and management of adverse events that require particular attention. Because of those reasons, NK cells appear as a viable effector cell alternative that can help address these challenges. NK cells offer a profile of activation, expansion, persistence, and cytotoxicity that is different from T cells and, when modified to bear CAR constructs, may provide significant advantages. However, the preclinical development of NK-CARs is challenging mainly because of the difficulty of generating large quantities of cells for testing and well-established pathways for CAR optimization before in vivo evaluation. Therefore, we developed a CAR optimization platform using the NK-92 cell line. NK-92 cells conserve their cytotoxic ability and can be easily expanded in vitro and used for functional and phenotypical evaluations of novel CAR-NK constructs. Here we present a rechallenge cytotoxic assay that mimics repetitive in vivo effector interactions with the target cells and its use for optimization, comparison, and development of NK-based cellular therapies. Methods: We generated lentivirus transduced CD19 CARs (FMC63-41BB-z) using T cells from healthy donors and NK-92 cells for comparison.T cells were expanded for 12 days, and a 41.9% CAR+ expression was achieved (CART19). Transduced NK-92 cells were sorted by FACS to obtain a population of 98.3 % CAR+ cells (CARNK19) and subsequently expanded for 12 days. JeKo-1 cells were used as CD19+ targets and BxPC3 cells as CD19 neg control (both cell types were GFP-Luc-PuroR). We developed a Luciferase-based rechallenge cytotoxicity assay. For this, we diluted the effector to target (E/T) ratio to obtain a logarithmic trendline of the cells' cytotoxicity. E/T ratio to get viability of 50% (IC50) measured at 4h (for CARNK19) and 24h (for CART19) was used as a proxy of the product's potency. Both CAR Immune Effector Cells (IECs) were co-cultured with their targets at an E/T ratio to obtain 70% cytotoxicity. After 24 hours with the target, we estimated the remaining IEC amount in the culture using GFP exclusion in flow analysis (IEC cells/mL = total cells/mL x GFP neg%). We repeated the plating of E/T ratio dilutions to perform daily IC50 curves using this rechallenge strategy for a total of 5 days. CAR and PD1 expression were measured on Day 0 and Day 5 by flow cytometry. Results: CART19 showed a higher IC50 than CARNK19 at baseline, 1.7 vs. 0.19 (Figure 1A). The IC50 trend of both IECs over time showed an uptrend that suggests progressive functional exhaustion (Figure 1B). At 5 days of rechallenge, it was 29 times higher in T cells than in NK-92 (12.07 vs. 0.42) and with a slope 265 times higher (10.6 vs. 0.04). Furthermore, we observed that when comparing the levels of CAR expression on Day 0 vs. Day 5, CART19 showed a decrease in CAR expression that was not present in CARNK19 (41.9 to 10.9% vs. 98.3 to 95.5%) (Figure 1C). In addition, there was a higher increase in PD1 expression in CART19 cells than CARNK19 cells from Day 0 to Day 5 of the in vitro rechallenge (9.9 to 46.8% vs. 0.88 to 8.88%) (Figure 1D). Conclusion: Our data shows the use of NK-92 cells as a tool for optimization and preclinical development of NK cell-based cellular therapies. We demonstrated that it is feasible to set up repetitive cytotoxic challenges that mimic closer in vivo E/T engagement. Moreover, using the cytotoxic IC50 calculated with this platform, we show increased cytotoxicity, less functional exhaustion, and less expression of PD1 in CARNK19 than in its T cell counterpart. Overall, the NK-92 rechallenge cytotoxicity assay platform constitutes a helpful tool for research, development, and optimization of cellular therapies based on NK cell effector function. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4796-4796
Author(s):  
Hareth Nahi ◽  
Michael Chrobook ◽  
Stephan Meinke ◽  
Charlotte Gran ◽  
Nicole Marquardt ◽  
...  

Abstract Introduction: Immunotherapies are gaining more and more importance in the treatment of multiple myeloma (MM). Antibodies directed against MM antigens like CD38, SLAMF7 or BCMA are used either in their natural form, conjugated to drugs, or in the form of bispecific T-cell engagers. Cellular therapies make use of cytotoxic lymphocytes, i.e. T cells or NK cells that can also be modified to express chimeric antigen receptors to target MM cells. Combinations of antibody and cellular therapies could further improve the outcome as, for example, NK cells can mediate antibody dependent cellular cytotoxicity (ADCC). However, NK cells also express CD38 and SLAMF7 and would be targeted by the therapeutic antibodies against these antigens. We have recently reported our clinical study infusing multiple doses of ex vivo activated and expanded autologous NK cells in six patients with MM post autologous stem-cell transplantation (EudraCT 2010-022330-83). Here, we report results of a phenotypic analysis of the ex vivo expanded NK cells and peripheral blood NK cells before and after infusion with implications for possible combination therapies. Methods: Ex vivo activated and expanded NK cells and NK cells in peripheral blood of the patients were analyzed by multiparameter flow cytometry. Peripheral blood cells were taken from the non-NK cell infusion arm before and at three different timepoints after infusion. NK-cell sub-populations within these samples were analyzed using t-SNE clustering. Results: Upon ex vivo activation and expansion, we observed that the NK cells gained a unique activated phenotype including populations of CD56 brightCD16 +Ki67 +HLA-DR + NK cells. Interestingly, these NK cells showed a reduced expression of CD38 compared to peripheral blood NK cells. Clustering analyses of data from peripheral blood samples revealed the gradual appearance of a new NK cell population with a similar phenotype in a dose-dependent fashion over four hours following infusion of the NK cell product. Infused NK cells could be detected in circulation up to four weeks after the last infusion. Like the NK cell infusion product, these cells expressed little to none CD38, high levels of NKG2D, 2B4, TIM-3, and TIGIT and similar levels of SLAMF7 compared to peripheral blood NK cells. Conclusions: The persistent high expression of CD16 and the low expression of CD38 in infused NK cells offers the choice to combine ex vivo activated and expanded NK cells with anti-CD38 antibody therapy without concern for antibody-mediated NK-cell death. Based on these findings, we have started a clinical trial testing this combined therapy (NCT04558931). Disclosures Nahi: XNK Therapeutics AB: Consultancy. Chrobook: XNK Therapeutics AB: Consultancy. Meinke: XNK Therapeutics AB: Consultancy, Current holder of stock options in a privately-held company. Gilljam: XNK Therapeutics AB: Current holder of individual stocks in a privately-held company. Stellan: XNK Therapeutics AB: Current holder of individual stocks in a privately-held company. Walther-Jallow: XNK Therapeutics: Other: Shareholder in the company. Liwing: XNK Therapeutics AB: Current Employment. Gahrton: XNK Therapeutics AB: Current holder of individual stocks in a privately-held company; Fujimoto Pharmaceutical Corporation Japan: Membership on an entity's Board of Directors or advisory committees. Ljungman: Takeda: Consultancy, Other: Endpoint committee, speaker; OctaPharma: Other: DSMB; Enanta: Other: DSMB; Merck: Other: Investigator, speaker; AiCuris: Consultancy; Janssen: Other: Investigator. Ljunggren: XNK Therapeutics AB: Current holder of individual stocks in a privately-held company, Membership on an entity's Board of Directors or advisory committees. Alici: XNK Therapeutics AB: Current holder of individual stocks in a privately-held company.


2021 ◽  
Vol 8 (Supplement_1) ◽  
pp. S131-S132
Author(s):  
Georgios Angelidakis ◽  
Roy F Chemaly ◽  
Partow Kebriaei ◽  
Nadim J Ajami ◽  
Micah M Bhatti ◽  
...  

Abstract Background Infectious complications in cancer patients (pts) who have received T-cell therapies are similar to those in autologous hematopoietic stem cell transplant (HCT) recipients, who - because they lose prior acquired immunity after undergoing conditioning regimens and transplantation- may be at an increased risk for vaccine-preventable infections. We sought to determine seroprotection rates against pneumococcus and tetanus-diphtheria before and after cellular therapies. Methods In this ongoing prospective observational cohort study, we enrolled pts with any type of cancer who received cellular therapy with chimeric antigen receptor modified T cell (CAR-T), natural killer CAR-T, or T-cell receptor- directed immunotherapies at MD Anderson Cancer Center from January 2020 through May 2021. We performed antibody assays for diphtheria, tetanus, and pneumococcus before, at 1 month, and between 3-6 months after T-cell therapy for each pt regardless of vaccination history. Results Of 38 pts enrolled, 27 (71%) were men and 25 (66%) had non-Hodgkin lymphoma (Table 1); 38 (100%) and 17 (45%) had a history of previous diphtheria-tetanus-acellular pertussis (Tdap) and pneumococcal vaccination, respectively (Table 2). Tetanus serologies were positive for all pts tested before, at 1 month and 3-6 months after T cell therapy (37/37 [100%], 22/22 [100%], and 13/13 [100%], respectively). Diphtheria serologies were positive for most pts tested before, at 1 month and 3-6 months after therapy (35/37 [95%], 20/22 [91%], and 11/13 [85%], respectively]. Pneumococcal serologies were positive for 8 out of 37 [22%] pts before therapy, among these 8 pts, 4 had positive serologies 1 month after therapy, and 2 of 3 tested 3-6 months after therapy had positive serologies. One pt received a pneumococcal vaccine 10 months after therapy but had negative serologies post-vaccination. Conclusion Most pts who received T-cell therapy retained their immunity for diphtheria and tetanus, but most also lost their immunity for pneumococcus. This suggests that the standard of care for pts receiving T-cell therapy should include more robust strategy for pneumococcal vaccination, but its timing, need for booster dosing, and antibody response needs to be determined in future trials. Disclosures Roy F. Chemaly, MD, MPH, FACP, FIDSA, AiCuris (Grant/Research Support)Ansun Biopharma (Consultant, Grant/Research Support)Chimerix (Consultant, Grant/Research Support)Clinigen (Consultant)Genentech (Consultant, Grant/Research Support)Janssen (Consultant, Grant/Research Support)Karius (Grant/Research Support)Merck (Consultant, Grant/Research Support)Molecular Partners (Consultant, Advisor or Review Panel member)Novartis (Grant/Research Support)Oxford Immunotec (Consultant, Grant/Research Support)Partner Therapeutics (Consultant)Pulmotec (Consultant, Grant/Research Support)Shire/Takeda (Consultant, Grant/Research Support)Viracor (Grant/Research Support)Xenex (Grant/Research Support) Fareed Khawaja, MBBS, Eurofins Viracor (Research Grant or Support) Ella Ariza Heredia, MD, Merck (Grant/Research Support)


Sign in / Sign up

Export Citation Format

Share Document