Skip to main content
Erschienen in: Journal of Translational Medicine 1/2024

Open Access 01.12.2024 | Research

Deciphering the importance of culture pH on CD22 CAR T-cells characteristics

verfasst von: Michaela Prochazkova, Alexandra Dreyzin, Lipei Shao, Pam Garces, Yihua Cai, Rongye Shi, Alejandra Pelayo, Yong Soo Kim, Victoria Pham, Sue Ellen Frodigh, Shannon Fenton, Catherine Karangwa, Yan Su, Kathryn Martin, Nan Zhang, Steven L. Highfill, Robert P. Somerville, Nirali N. Shah, David F. Stroncek, Ping Jin

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2024

Abstract

Background

Chimeric antigen receptor (CAR) T-cells have demonstrated significant efficacy in targeting hematological malignancies, and their use continues to expand. Despite substantial efforts spent on the optimization of protocols for CAR T-cell manufacturing, critical parameters of cell culture such as pH or oxygenation are rarely actively monitored during cGMP CAR T-cell generation. A comprehensive understanding of the role that these factors play in manufacturing may help in optimizing patient-specific CAR T-cell therapy with maximum benefits and minimal toxicity.

Methods

This retrospective study examined cell culture supernatants from the manufacture of CAR T-cells for 20 patients with B-cell malignancies enrolled in a phase 1/2 clinical trial of anti-CD22 CAR T-cells. MetaFLEX was used to measure supernatant pH, oxygenation, and metabolites, and a Bio-Plex assay was used to assess protein levels. Correlations were assessed between the pH of cell culture media throughout manufacturing and cell proliferation as well as clinical outcomes. Next-generation sequencing was conducted to examine gene expression profiles of the final CAR T-cell products.

Results

A pH level at the lower range of normal at the beginning of the manufacturing process significantly correlated with measures of T-cell expansion and metabolism. Stable or rising pH during the manufacturing process was associated with clinical response, whereas a drop in pH was associated with non-response.

Conclusions

pH has potential to serve as an informative factor in predicting CAR T-cell quality and clinical outcomes. Thus, its active monitoring during manufacturing may ensure a more effective CAR T-cell product.
Begleitmaterial
Additional file 1: Figure S1. Manufacturing schema for CD22 CAR T-cells and simplified representative gating strategy of the final product. a) Simplified culture schematic with major processing steps during CD22 CAR T-cells manufacturing at Center for Cellular Engineering, Clinical Center, NIH. b) CD56, CD14, and CD15% in the apheresis bag and selected CD4/CD8 T cells. c) Representative flow cytometry staining including protein L gating strategy in final CD22 CAR T product. Viable cells (7-AAD negative population) were initially gated from the singlets and CD45+ cells. Viable CD3+ cells were then gated and analyzed for protein L expression. The untransduced cells were stained at the same time and were used to identify protein L+ population in transduced cells. Figure S2. CD3% and pO2 levels did not correlate with D2 pH values. a) pH did not impact % of CD3 (87% for low pH vs 89% for high pH) and showed no correlation with pH on D2 of the CD22 CAR T-cells manufacturing process. b) No obvious changes were observed in oxygen levels comparing cultures with low or high pH. pO2 did not correlate with pH on D2 of the CD22 CAR T-cells manufacturing process. Figure S3. The relationship between low pH and CR in patients with high baseline disease burden and prior HSCT. a) Of the 6 patients with M3 baseline disease burden, the 4 who started out with low pH achieved a complete response, whereas the two patients with high disease burden who started out with higher pH had no response. b) A similar pattern was noted in patients that have previously received a hematopoietic stem cell transplant, but not among those who had not been exposed to transplant.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-024-05197-5.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
GMP
Good Manufacturing Practice
CAR
Chimeric antigen receptor
ALL
Acute lymphoblastic leukemia
FDA
Food and Drug Administration
CCE
Center for Cellular Engineering
TNC
Total nucleated cell
GSEA
Gene Set Enrichment Analysis
HSC
Hematopoietic stem cell
HSCT
Hematopoietic stem cell transplant
CRS
Cytokine release syndrome
CR
Complete response
L
Lentivirus
TR
Transduction
CHM
Change of media

Background

Chimeric antigen receptor (CAR) T-cell therapy has been highly effective in patients with B-cell hematologic malignancies, including acute lymphoblastic leukemia [15] and non-Hodgins’ lymphoma [68], and ongoing research aims to expand applications to solid tumors [9, 10] and autoimmune diseases [1113]. Since the initial approval of the CD19-targeted CAR T-cell, tisagenlecleucel (Kymriah), in 2017, the US Food and Drug Administration (FDA) has approved five more CAR T-cell therapies for acute lymphoblastic leukemia, non-Hodgkin lymphoma, and multiple myeloma. The rapid growth of this therapy is illustrated by the more than 700 CART trials actively recruiting patients (clinicaltrials.gov, February 2024).
As new CAR T-cell therapies become commercialized and enter into clinical use, it is essential to produce CAR T-cells that are consistently of high quality and potency. With more clinical centers aiming to establish point-of-care processes of CAR T-cell manufacturing, maintaining consistent cell culture conditions will become increasingly challenging.
Cell culture is a dynamic process, so understanding the interactions between cells, culture media, nutrients, and growth factors is critical for producing high quality and potent cell therapy products. Cell counts, viability, and expansion are routinely monitored during CAR T-cell manufacturing, but little attention is paid to variables such as metabolites, oxygen and carbon dioxide tension, or pH. These factors, however, may be associated with CAR T-cell proliferation and fitness. The pH in particular is a simple indicator that may reflect carbon dioxide tension, lactate accumulation, and rate of T-cell proliferation. Thus, monitoring these variables could be valuable in predicting properties of the final cell therapy product.
Not only could culture media characteristics serve as indicators of T-cell expansion, but they could also be a potential point of intervention. The pH of the culture media has been shown to affect T-cell proliferation, differentiation, and metabolism. T-cells cultured in acidic media (pH 6.6 and lower) have reduced activation, proliferation, and slower differentiation to antigen-specific cytotoxic T-lymphocytes [14, 15]. Moreover, in a study of tumor-infiltrating T-lymphocytes, acidic media led to suppression of T-cell function, including reduced cytokine secretion and lower expression of T-cell receptors [16]. On the other hand, while prolonged (12 days) ex vivo expansion of T-cells in an acidic environment (pH 6.6) reduced rate of proliferation, it also promoted CD8+ T-cell stemness [17]. Additionally, this extended expansion of T-cells in acidic media triggered T-cell metabolic reprogramming with restricted glycolysis and increased long chain fatty acid metabolism [17]. Studies of how changes in culture media affect T-cells in the context of CAR T-cell production specifically are limited.
Accurate monitoring and control of pH and metabolites may be an important opportunity to ensure optimal CAR T-cell performance and function. However, these factors are rarely monitored during clinical Good Manufacturing Practice (GMP) CAR T-cell generation and their effect on CAR T-cell growth and differentiation has yet to be fully investigated. In this study, we examined the pH and metabolic parameters of anti-CD22 CAR T-cell cultures and explored the relationship between culture conditions and clinical outcomes.

Methods

Patients’ cohort

The products included in this study were derived from a cohort of 20 children and young adults who were diagnosed with recurrent or refractory CD22-expressing B-cell malignancies and were enrolled on a Phase 1/2 clinical trial of anti-CD22 CAR T-cells (NCT 02315612). All patients were treated in the Pediatric Oncology Branch of the National Cancer Institute, National Institutes of Health at a dose level of 0.3 × 106 cells.

CD22 CAR-T manufacturing process

All anti-CD22 CAR T-cell products were manufactured at the Center for Cellular Engineering (CCE) between September 2019 and August 2022. The CCE is compliant with good manufacturing practice (GMP) and good clinical laboratory practice (GCLP) standards and has manufactured numerous CAR T-cell products for phase 1 and 2 clinical trials at the National Institutes of Health. All the products were manufactured using autologous mononuclear cells collected by apheresis. T-cells from fresh (n = 5) or cryopreserved (n = 15) autologous mononuclear cells were selected on CliniMACS (Miltenyi Biotec) using anti-CD4 and anti-CD8 magnetic beads and stimulated with anti-CD3/CD28 Dynabeads (ThermoFisher) with a ratio of 1 cell:3 beads. Cells were seeded into culture bags at a density of 1.5–2.0 × 106 viable CD3+ cells. Cell culture was maintained in AIM-V medium (Gibco) containing heat inactivated human AB serum (Valley Biomedical), Glutamax (Gibco) and IL2 (Aldesleukin) in a 37 °C, 5% CO2 incubator for 9 days. On day 2 of the culturing process, cells were transduced with GMP grade CD22 lentiviral vector (Lentigen) using bag spinoculation (1000 g for a period of 2 h). On day 3 (D3), transduction was stopped by media exchange. Debeading and cell density adjustment to 0.4 × 106 viable cells/mL was performed on day 4 (D4) of the culturing process. Cell density was further adjusted on Day 7 (D7) to 0.6–1 × 106 viable cells/mL. All cultures were harvested on Day 9 (D9) and were either freshly infused (n = 7) or cryopreserved (n = 9). A simplified 9-day manufacturing process is illustrated in Additional file 1: Supplementary Figure 1a.

Collection of culture media supernatants

Culture media supernatants were collected at four different time points during the CD22 CAR T-cell culture (on Day 2, 3, 4 and 9). 10 mL of culture supernatant was collected before transduction on D2, when transduction was stopped on D3, and before debeading on D4. Final supernatants were collected during the cell harvest on D9. All supernatants were aliquoted (1 mL per vial), immediately frozen and stored at − 80 °C until further analysis.

Metabolite analysis

A Vi-CELL MetaFLEX bioanalyzer (Beckman Coulter) was used to measure pH, glucose, lactate, pCO2 and pO2 levels during the manufacturing process. At the time of the analysis, supernatants collected at various time-points during the culturing process were thawed and warmed to 24 °C. 65 uL of temperature adjusted supernatants were loaded into the MetaFLEX device for testing.

Bio-Plex assay

Culture media cytokine, chemokine, and growth factor levels were measured at 2 different time points (D2 and D9) during the culture process using the Bio-Plex 200 system (Bio-Rad Laboratories). Bio-Plex assays were performed according to the manufacturers’ instructions using commercially available Bio-Plex Pro Human Cytokine Screening 48-plex Panel Assay. All assay values are reported in pg/mL.

Bulk mRNA sequencing

We have previously described the bulk RNA sequencing procedures in detail [18]. In brief, total mRNA from CAR T-cell products was isolated using the miRNeasy Mini Kit. The concentration and quality were assessed using Nanodrop 8000 and 2100 Bioanalyzer, respectively. DNA libraries were prepared using the TruSeq Stranded Total RNA kit and sequenced on the Illumina Nextseq550 platform.

Transcriptome data analysis

Raw fastq files underwent quality control using FastQC and were further processed with Trimmomatic to remove adapter sequences and low-quality reads. After filtering, the reads were aligned to the human reference genome (GENCODE hg38) using the STAR aligner. Gene expression levels were quantified using subread (featureCounts), and TPM values were utilized for downstream analysis. For differential expression analysis, we employed the limma package in RStudio with custom scripts. Threshold for definition of differentially expressed genes (DEGs) is p value < 0.01 & |FoldChange|≥ 2. We then performed gene set enrichment analysis (GSEA), a bioinformatic method to determine whether a predefined set of genes showed statistically significant, concordant differences between two biological states. We divided the gene expression from our RNA-seq data into two sets: up-regulated and down-regulated set. Subsequently, we examined the presence of genes associated with each pathway in either the up-regulated or down-regulated set. The degree of enrichment was quantified by a normalized enrichment score (NES). A significant positive NES value suggests that members of the selected pathway gene set tend to appear in the up-regulated set, whereas a significant negative NES value indicates the opposite scenario. The analysis was conducted by the built-in function of the cluster Profiler package [19] in RStudio.

Flow cytometry

Transduction efficiency, viability and percentage of cells expressing CD3, CD4, CD8, CD14, CD15, and CD56 were measured using flow cytometry following staining with fluorescently labelled antibodies (BD Biosciences). The CD22-CAR vector transduction efficiency was detected using protein L (ThermoFisher). Untransduced cells were used to determine the positive gate for transduction efficiency. The gating strategy of protein L expression on CD22 CAR T-cells is displayed in Additional file 1: Supplementary Figure 1c. Frequencies of the above-mentioned markers were acquired on the BD FACS CantoII (BD Biosciences). Thirty thousand events in total were collected for each sample. Data were analyzed using BD FACSDiva software.

Statistical analysis

All data are presented as mean ± SEM. Graphical and statistical analysis was performed using GraphPad Prism 7 software. Statistical differences between the groups were assessed either using an unpaired t test, Mann–Whitney, or nonparametric Spearman correlation. P value less than 0.05 was considered significant, except for the next-generation sequencing data analysis.

Results

Overview of CD22 CAR-T cell expansion during the manufacturing process

The clinical characteristics of patients whose CD22 CAR T-cell products were included in the study are detailed in Table 1. In total, twenty CD22 CAR T products were analyzed. All 20 products met the 0.3 × 106/kg target dose. Four products were not infused, as the patients experienced rapidly progressive disease during the manufacturing process. Seven patients received infusions of fresh CAR T-cells, and nine patients received cryopreserved CAR T-cells.
Table 1
Patients and manufacturing process characteristics
Number of patients
20
Disease
B cell malignancies (B-ALL in 19 patients, DLBCL in 1 patient)
Age (mean, years)
13
Weight (mean, kg)
45.9
Sex
8F/12M
Race
White
13
Asian
2
AA
2
UNK
2
Multiple
1
Ethnicity
NH
14
H
6
Prior HSCT
10/20
Prior CAR T-cell therapy
15/20
Expansion method
Bag culture
Dose
0.3 × 106/kg
Starting material
CliniMACS enriched CD4/CD8
Media and cytokines
AimV, IL2
Stimulation
CD3/CD28 Dynabeads
Length of the culture
9 days
Vector
Lentivirus
B-ALL B-cell acute lymphoblastic leukemia, DLBCL diffuse large B cell lymphoma, F Female, M Male, AA Afro-American, UNK Unknown, NH non-Hispanic, H Hispanic, HSCT hematopoietic stem cell transplant
During the manufacturing process, viable total nucleated cells (TNC) were counted on days 0, 2, 4, 7 and 9. The mean TNC at the start of the manufacturing process was 470 × 106 cells (range 288–565 × 106). On Day 2, a mean of 343 × 106 total nucleated cells underwent transduction with lentiviral vector. On Day 4, there was a drop in total nucleated cells with a mean of 52 × 106 cells (range 26–76 × 106). The total number of cells harvested on Day 9 was 1.1 × 109 (range 0.3–1.7 × 109, Fig. 1a). Fold expansion was determined between the following days of the cell culture: 0–2, 2–4, 4–7 and 7–9. The maximum fold expansion was achieved on Days 4–7 of the culturing process (mean 8.5, range 4.6–11.9) with a decrease in expansion between Days 7 and 9 (mean 2.9, range 2.0–3.9, Fig. 1b). As shown in Fig. 1c, the percentage of CD3+ cells increased from 47.9 in the apheresis bag to 91.2 post-CD4/CD8 selection to 99.7 at the time of the harvest of CAR T-cells. The apheresis bag and selected CD4/CD8 T-cells were analyzed for additional markers such as CD56, CD14, or CD15. Additional file 1: Supplementary Figure 1b shows the analysis of these markers. The transduction efficiency (percent of protein L+ cells) was assessed on Days 7 and Day 9 of the manufacturing process. On Day 7, after the fastest period of expansion, 31% transduction was observed (data not shown). By Day 9, a significantly larger proportion of cells expressed protein L, (42.8%, p = 0.0073, Fig. 1d), suggesting preferential expansion of transduced cells. The final CD4/CD8 ratio was 2.8 (Fig. 1e).

The pH of the cell culture at the beginning of the manufacturing process correlated with CAR T-cell growth, expansion, and metabolic activity

Culture media supernatants were collected at 4 different time points throughout CD22 CAR T-cell manufacturing: Days 2, 3, 4, and 9. Our primary focus was on the findings on Day 2 and Day 9. Prior to transduction on Day 2, the pH of cell culture media ranged from 7.1–7.5 (Fig. 2a). pH level showed a statistically significant negative correlation with the D2 TNC count and fold expansion. On retrospective evaluation, we separated the products into 2 groups: 12 samples had relatively low pH on Day 2 (ranging from 7.093–7.289; mean 7.2), and 8 samples had high pH (ranging from 7.323–7.541; mean 7.4). The differences in pH levels between these 2 groups were statistically significant (p < 0.0001, Fig. 2a). The cell counts, fold expansion, and metabolic activity of T-cells were compared between the low pH and high pH groups. Cell products in the low pH group had significantly elevated total nucleated cell counts (414 vs 236 × 106, p < 0.0061, Fig. 2b), greater fold expansion (0.85 vs 0.53, p < 0.0015, Fig. 2c), higher pCO2 (23.2 vs 17.6 mmHg, p < 0.001, Fig. 2d) and higher lactate levels (0.73 vs 0.38 g/L, p < 0.0001, Fig. 2f). Those in the low pH group also had reduced glucose levels (1.9 vs 2.3 g/L, p < 0.0003, Fig. 2e) compared with the high pH group. These results demonstrate the relationship between lower extracellular pH and T-cell growth, expansion, and glycolysis as pH levels correlated with pCO2, glucose, and lactate levels. On the other hand, Day 2 pH levels did not correlate with CD3% (Additional file 1: Supplementary Figure 2a) or pO2 levels (Additional file 1: Supplementary Figure 2b).
Cytokine and growth factor levels were also compared between low and high pH groups. On Day 2 of the manufacturing process, six analytes demonstrated significantly greater levels among samples in the low pH group compared with the high pH group (Fig. 3). Increased levels of stem cell factor (SCF, p < 0.04), macrophage colony-stimulation factor (M-CSF, p < 0.004), leukemia inhibitory factor (LIF, p < 0.0052), interleukin 15 (IL-15, p < 0.0066), interleukin receptor 2 alpha (IL2Rα, p < 0.0002), and tumor necrosis factor beta (TNF-β, p < 0.0001) were observed in products with more acidic pH. In the context of increased proliferation associated with lower pH as shown above, these findings were consistent with our expectation that more active T-cells will secrete more cytokines and growth factors into the cell culture media.

The pH of culture media at the end of manufacturing reflects metabolic profile, but does not correlate with expansion of final CAR T-cell products

Next, we investigated whether the pH level measured at the end of manufacturing was associated with cell or metabolic parameters. Overall, we observed a slight decline in pH values during the manufacturing process with higher values on Day 2 compared to Day 9. The Day 9 pH ranged from 7.0 to 7.4 and based on its values, the final CAR T-cell products were also separated into 2 groups representing low (9 patients, pH ranged from 7.008–7.235 with the mean pH of 7.1) and high pH (11 patients, pH ranged from 7.252 to 7.432 with mean pH of 7.3). The difference in final pH levels between these two groups was statistically significant (p < 0.0001, Fig. 4a). However, unlike the Day 2 pH groups, these final pH groups did not differ in terms of cell counts (Fig. 4b), fold expansion (Fig. 4c), CD3% (Fig. 4d), pCO2 (Fig. 4e), or pO2 (Fig. 4f). Higher pH at harvest correlated with higher glucose (1.91 vs 1.55 g/L; p < 0.0003, Fig. 4g) and lower lactate production (0.64 vs 0.89 g/L; p < 0.0002, Fig. 4h), suggesting reduced glycolysis and lower metabolic activity.
The Day 9 levels of 7 cytokines differed between the high and low pH groups. All 7 were elevated in the low pH group compared to the high pH group [interleukin 6 (IL6, p < 0.008), interleukin 12 subunit p70 (IL-12p70, p < 0.003), tumor necrosis factor alpha (TNF-α, p < 0.008), leukemia inhibitory factor (LIF, p < 0.007), macrophage migration inhibitory factor (MIF, p < 0.002), macrophage colony-stimulation factor (M-CSF, p < 0.003) and granulocyte macrophage colony-stimulation factor (GM-CSF, p < 0.02)] (Fig. 5). The levels of LIF and M-CSF were also greater in the low pH group on day 2 (Fig. 3), although others did not differ significantly.

Sequencing profile of final CD22 CAR T-cells products demonstrates differences in T-cell activation pathways associated with pH

CD22 CAR T-cell final product characteristics were analyzed by RNA sequencing (Fig. 6). Sequencing analysis of 17 final products assigned 78 differentially expressed genes between pH groups at harvest: 15 downregulated and 63 upregulated under low pH. GSEA analysis revealed that genes involved in T-cell activation, lymphocyte activation, differentiation, and mediated immunity, tend to be downregulated in low pH conditions. This highlights the value of pH as an indicator during CD22 manufacturing as it can help identify more activated CAR T-cell products (Table 2 shows the list of genes involved in each pathway).
Table 2
List of genes involved in each pathway
Lymphocyte activation
Lymphocyte differentiation
Lymphocyte mediated immunity
T cell activation
FCER1G
FCER1G
C4BPB
FCER1G
IGFBP2
KIT
FCER1G
IGFBP2
KIT
TCF7
RSAD2
KIT
TCF7
POU2AF1
MASP2
TCF7
POU2AF1
RSAD2
XCL1
RSAD2
RSAD2
CD79A
KLRC1
RELB
CD79A
RELB
CD8A
AIF1
RELB
LEF1
MALT1
LEF1
AIF1
CCR7
ZBTB1
CCR7
CXCR5
NTRK1
CD27
XCL1
CD38
CD83
CRTAM
TMIGD2
LEF1
GLI2
PRKCZ
CD83
CCR7
KLRC1
TNFSF4
SIRPG
XCL1
WNT4
SUSD4
GLI2
NTRK1
PRDM1
IL18RAP
TREML2
IRS2
CD8A
IL4R
CD8B
TMIGD2
MYB
LTA
KLRC1
CD83
FOXP1
THOC1
HHLA2
SIRPG
IRF4
IL23A
WNT4
GLI2
MALT1
IRF7
PIK3CA
TREML2
ZBTB1
ULBP2
PRDM1
CD8B
IL15
BATF
SLC11A1
KLRC1
CD27
TRAF6
CD8A
CD180
CRTAM
FOXP3
MYB
HHLA2
SOCS5
CD55
FOXP1
WNT4
HDAC5
ICAM1
IRF4
PIK3CA
RHOH
KLRC2
MALT1
TNFAIP3
DUSP10
NBN
ZBTB1
PRDM1
PRKCZ
PIK3R6
EBI3
SLC11A1
IL2RA
C1S
IL15
CD8A
EP300
IL20RB
CD27
MYB
CHD7
SLC22A13
WDFY4
FOXP1
IRF1
LGALS9
CTPS1
IRF4
TNFSF4
RFTN1
CRTAM
MALT1
RABL3
CEBPG
PELI1
ZBTB1
RIPK2
SMAD7
SOCS5
EBI3
TGFBR2
BCL3
RHOH
IL15
IL4R
NCR3
DUSP10
CD27
EZH2
SLC15A4
PRKCZ
WDFY4
GPR89A
AP1G1
IL2RA
CTPS1
MDK
CD160
CHD7
CRTAM
IL23A
 
IRF1
PELI1
KAT7
 
TNFSF4
SOCS5
KLF6
 
PAWR
HDAC5
CTLA4
 
IL6ST
RHOH
BATF
 
RABL3
DUSP10
SP3
 
RIPK2
PRKCZ
ITPKB
 
CD7
IL2RA
PPP2R3C
 
TGFBR2
LST1
RUNX1
 
IL4R
SLC39A10
VCAM1
 
GPR89A
EP300
ATG5
 
MDK
CHD7
FOXP3
 
IL23A
IRF1
PATZ1
 
CTLA4
TNFSF4
HDAC4
 
BATF
VAV3
DOCK10
 
NCK2
PAWR
PIK3R6
 
SP3
IL6ST
KAT2A
 
ITPKB
RABL3
TPD52
 
RUNX1
RIPK2
RUNX3
 
VCAM1
CD7
TP53
 
TRAF6
TGFBR2
IL6R
 
ATG5
IL4R
STAT3
 
AZI2
EZH2
ZMIZ1
 
FOXP3
GPR89A
LGALS9
 
PATZ1
MDK
PIK3CD
 
CD55
THOC1
PTPN2
 
MAPK8IP1
IL23A
NFKBIZ
 
ICAM1
KAT7
CEBPG
 
SLC7A1
KLF6
IRF2BP2
 
CBLB
ULBP2
NFATC2
 
CASP8
CTLA4
SMAD7
 
PIK3R6
BATF
IKZF3
 
KAT2A
NCK2
NOTCH2
 
IL20RB
SP3
ZC3H8
 
RUNX3
ITPKB
ZFP36L2
 
CD44
PPP2R3C
  
TP53
RUNX1
  
IL6R
VCAM1
  
STAT3
TRAF6
  
ZMIZ1
ATG5
  
LGALS9
AZI2
  
FYN
FOXP3
  
PIK3CD
PATZ1
  
PTPN2
ZNF335
  
DLG5
CD55
  
SRC
MAPK8IP1
  
NFKBIZ
ICAM1
  
DPP4
HDAC4
  
NFATC2
SLC7A1
  
SMAD7
CHRNB2
  
SDC4
KLRC2
  
ZC3H8
DOCK10
  
ZFP36L2
CBLB
  
PAG1
NBN
  
PAK1
CASP8
  
SEMA4A
PIK3R6
  
BCL3
KAT2A
   
TPD52
   
IL20RB
   
RUNX3
   
CD44
   
TP53
   
IL6R
   
STAT3
   
ZMIZ1
   
TNIP2
   
LGALS9
   
FYN
   
PIK3CD
   
PTPN2
   
DLG5
   
SRC
   
NFKBIZ
   
DPP4
   
CEBPG
   
IRF2BP2
   
NFATC2
   
SMAD7
   
IKZF3
   
NOTCH2
   
SDC4
   
AHR
   
ZC3H8
   
ZFP36L2
   
PAG1
   
BANK1
   
PAK1
   
SEMA4A
   
BCL3
   
NCR3
   
ST3GAL1
   
IL21R
   
SLC15A4
   
AP1G1
   
JAK3
   

Low pH at D2 is associated with clinical outcome

To determine if product pH on Day 2 correlated with clinical outcomes, we compared disease response rates between the low and high pH groups (Fig. 7). Of the 12 patients whose samples were in the low pH group on D2, 10 were infused and 2 were not due to progressive disease. Among the patients receiving the cells, 8 (80%) achieved a complete response (CR), which is defined as complete bone marrow response at 28 days. The high pH group contained 8 patients. Six patients received CAR T-cell infusions and 2 did not due to rapidly progressive disease. In contrast to the low pH group, among those with high pH, only 2 patients (33%) achieved a clinical response, while 4 (67%) did not. The relationship between low starting pH and CR was most consistent in the subsets of patients with high baseline disease burden (M3 marrow, > 25% blasts) (Additional file 1: Supplemetary Figure 3a) and prior hematopoietic stem cell transplant (HSCT) (Additional file 1: Supplemetary Figure 3b).
Moreover, we observed a difference in toxicity patterns between the low and high pH groups. In the low pH group, 8 patients (all those with CR) developed cytokine release syndrome (CRS), 5 (62.5%) of them with Grade 1 and 3 with (37.5%) Grade 2. Non-responders in this group did not have any toxicities. Only 25% of the patients with clinical response in the low pH group experienced neurotoxicity and 50% had IEC-HS (immune-effector cell HLH-like syndrome) (Table 3). In the high pH group, both patients with clinical response developed Grade 2 CRS, neurotoxicity, and IEC-HS. Furthermore, in the high pH group, all non-responders experienced toxicities as well: all had CRS, 2 of them had neurotoxicity, and one had IEC-HS.
Table 3
pH correlation to clinical outcome and adverse events
Patient
pH
Clinical response
CRS gradea
IEC-HS
Neurotoxicity
5
Low
18
Low
11
Low
No
0
No
No
13
Low
No
0
No
No
3
Low
Yes
2
Yes
No
4
Low
Yes
1
No
No
7
Low
Yes
1
Yes
No
8
Low
Yes
1
No
No
12
Low
Yes
1
Yes
No
16
Low
Yes
2
No
Yes
19
Low
Yes
2
Yes
No
20
Low
Yes
1
No
Yes
10
High
17
High
6
High
No
1
No
No
9
High
No
2
Yes
Yes
14
High
No
1
No
Yes
15
High
No
1
No
No
1
High
Yes
2
Yes
Yes
2
High
Yes
2
Yes
Yes
CRS Cytokine Release Syndrome, IEC-HS Immune-Effector Cell HLH-like syndrome
aCRS graded based on American Society for Transplantation and Cellular Therapy criteria: Lee [28]
Analysis of pH changes over the course of manufacturing demonstrated that patients who achieved CR were more likely to have cells with stable or slightly increased pH values during the manufacturing process, whereas those who did not have CR had a drop in pH (Fig. 8a). Similar to the pattern observed with starting pH, the association between a drop in pH and lack of clinical response was most pronounced among patients with high baseline disease burden or history of prior HSCT (Fig. 8b and c).

Discussion

The aim of this study was to assess the relationship between culture conditions, specifically culture pH, and the growth kinetics and properties of CD22 CAR T-cells used for the treatment of pediatric B-cell acute lymphoblastic leukemia. Our data demonstrate that pH fluctuations during the CAR T-cell manufacturing process correlate with properties of the cellular product and may be indicative of clinical outcome. Based on our retrospective analysis, high pH at the start of manufacturing is associated with reduced number of viable nucleated cells, slowed cell expansion, and reprogrammed T-cell metabolism. Patients whose cells had a high pH at the start of the manufacturing tended to have a lower probability of clinical response. Since the data suggests that pH levels at 7.2 to 7.3 are indicative of optimal manufacturing conditions, the measure may be useful as an predictor of appropriate T-cell growth, differentiation, and clinical efficacy. We therefore propose that careful monitoring of pH during the manufacturing of cellular products is critical, and accurate pH monitoring during cGMP manufacturing should become a standard.
In our samples, lower pH generally correlated with a picture of increased T-cell proliferation. Lower pH was connected with a drop in glucose and an increase in lactate levels [20, 21]. This shows that T-cells grown in lower pH are more metabolically active compared to high pH. As the most rapid cell expansion occurs during the early days of CAR T-cell manufacturing, the Day 2 pH remains a better indicator of this process than the Day 9 pH, which is reflecting the media typically after several days of slower growth. It will be important in future studies to explore whether cell expansion patterns in vitro are recapitulated in vivo after cells are infused.
Lower pH may not only be a consequence of greater T-cell expansion but may actually be supporting further growth. Despite T-cell dysfunction in truly acidic environments, pH in the lower range of normal has been reported to be beneficial in the context of CAR T-cell expansion. In in vitro studies, reduction in extracellular pH to 6.6 significantly affected not only T cell proliferation but also reduced IL-2 and IFN-γ secretion, downregulated CD25 and CD71 expression, and upregulated CTLA4 expression leading to apoptosis [15]. Similarly, pH in the range of 6.0 to 6.5 has been shown to induce anergy in CD8+ T lymphocytes, with impaired proliferation, reduced cytokine production, decreased CD25 expression, and inhibition of STAT5 and p-ERK activation [16]. Although the pH fluctuations in our study might appear minor, the results are consistent with other studies showing that only small variations in culture pH can have a substantial effect on T-cells. Others have reported improved T-cell stimulation, expansion, and proliferation in lower pH, 7.0–7.3, compared to neutral pH [22, 23], which may be consistent with our findings. Similarly, only small variations in pH were also shown to be critical in hematopoietic cultures, mainly for progenitor cell differentiation and cloning efficiency [24].
In addition to predicting T-cell proliferation, the pH of culture medium may also help predict cytokine production. We demonstrated that cells starting out in lower pH had increased production of several cytokines including IL-6, IL12p70, TNF-a and GM-CSF, which play an important role not only in CAR T-cells’ targeted cytotoxicity, but also in development of off-target effects, such as CRS and neurotoxicity [2527]. Further analysis of cytokine production during CAR T-cell manufacturing may yield important insight into which patients are at risk for developing CAR T-cell associated toxicities.
There are important limitations to this exploratory study, and further investigation will be needed to confirm the trends reported here. The sample size is limited, and since our study included only anti-CD22 CAR T-cells, the findings might not be applicable to other CAR T-cell types. Overall, the relationship between pH and T-cell characteristics likely varies with the specific manufacturing protocol, including culture media, manufacturing platform, and timing. We are, however, continuing to collect more samples as new patients are treated, including those on the ongoing CD22 trial and trials of other novel CAR T-cell products. Importantly, the data reported here were collected over the course of our standard CAR T-cell manufacturing process, and no intentional adjustments to pH were made. Therefore, we can report only correlations with other parameters rather than any causal relationships. Future work will focus on pH modification during the manufacturing process to determine whether this will have any effect on the characteristics and efficacy of the final product.
Currently there are no guidelines or standards advising monitoring of pH during cell culture for CAR T-cell manufacturing. The emerging data, however, points to pH as an important and easily accessible tool for monitoring and standardizing CAR T-cell production. Furthermore, it has potential to serve as an early indicator of clinical outcome. Since adjusting the pH of cell culture media can be a relatively straightforward and inexpensive intervention, further prospective studies are needed to determine whether actively controlling pH during manufacturing is beneficial.

Conclusions

Measurements of pH throughout CAR T-cell manufacturing correlate with cell proliferation as well as other metabolic properties of CAR T-cells. Accurate pH tracking during CAR T-cell manufacturing may serve as an important metric of the quality of the cellular product and may become a predictive marker of CAR T-cell clinical efficacy. pH should be monitored throughout CAR T-cell manufacturing, and combined with other factors, it could significantly improve the quality, compatibility, and safety of CAR T-cells.

Acknowledgements

The authors would like to thank Hannah Song, Ph.D. and Sarah Underwood for their help and guidance with the cell manufacturing process.

Declarations

The study was approved by the NIH Institutional Review Board (IRB).
Not applicable.

Competing interests

The authors have no competing interest to declare.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Additional file 1: Figure S1. Manufacturing schema for CD22 CAR T-cells and simplified representative gating strategy of the final product. a) Simplified culture schematic with major processing steps during CD22 CAR T-cells manufacturing at Center for Cellular Engineering, Clinical Center, NIH. b) CD56, CD14, and CD15% in the apheresis bag and selected CD4/CD8 T cells. c) Representative flow cytometry staining including protein L gating strategy in final CD22 CAR T product. Viable cells (7-AAD negative population) were initially gated from the singlets and CD45+ cells. Viable CD3+ cells were then gated and analyzed for protein L expression. The untransduced cells were stained at the same time and were used to identify protein L+ population in transduced cells. Figure S2. CD3% and pO2 levels did not correlate with D2 pH values. a) pH did not impact % of CD3 (87% for low pH vs 89% for high pH) and showed no correlation with pH on D2 of the CD22 CAR T-cells manufacturing process. b) No obvious changes were observed in oxygen levels comparing cultures with low or high pH. pO2 did not correlate with pH on D2 of the CD22 CAR T-cells manufacturing process. Figure S3. The relationship between low pH and CR in patients with high baseline disease burden and prior HSCT. a) Of the 6 patients with M3 baseline disease burden, the 4 who started out with low pH achieved a complete response, whereas the two patients with high disease burden who started out with higher pH had no response. b) A similar pattern was noted in patients that have previously received a hematopoietic stem cell transplant, but not among those who had not been exposed to transplant.
Literatur
1.
Zurück zum Zitat Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–48.CrossRefPubMedPubMedCentral Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–48.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–17.CrossRefPubMedPubMedCentral Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–17.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224–5.CrossRef Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224–5.CrossRef
4.
Zurück zum Zitat Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–28.CrossRefPubMed Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–28.CrossRefPubMed
5.
Zurück zum Zitat Wang S, Wang X, Ye C, Cheng H, Shi M, Chen W, et al. Humanized CD19-targeted chimeric antigen receptor T (CAR-T) cells for relapsed/refractory pediatric acute lymphoblastic leukemia. Am J Hematol. 2021;96(5):E162–5.CrossRefPubMed Wang S, Wang X, Ye C, Cheng H, Shi M, Chen W, et al. Humanized CD19-targeted chimeric antigen receptor T (CAR-T) cells for relapsed/refractory pediatric acute lymphoblastic leukemia. Am J Hematol. 2021;96(5):E162–5.CrossRefPubMed
6.
Zurück zum Zitat Makita S, Yoshimura K, Tobinai K. Clinical development of anti-CD19 chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma. Cancer Sci. 2017;108(6):1109–18.CrossRefPubMedPubMedCentral Makita S, Yoshimura K, Tobinai K. Clinical development of anti-CD19 chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma. Cancer Sci. 2017;108(6):1109–18.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Jackson Z, Roe A, Sharma AA, Lopes F, Talla A, Kleinsorge-Block S, et al. Automated manufacture of autologous CD19 CAR-T cells for treatment of non-Hodgkin lymphoma. Front Immunol. 2020;11:1941.CrossRefPubMedPubMedCentral Jackson Z, Roe A, Sharma AA, Lopes F, Talla A, Kleinsorge-Block S, et al. Automated manufacture of autologous CD19 CAR-T cells for treatment of non-Hodgkin lymphoma. Front Immunol. 2020;11:1941.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Albelda SM. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat Rev Clin Oncol. 2024;21(1):47–66.CrossRefPubMed Albelda SM. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat Rev Clin Oncol. 2024;21(1):47–66.CrossRefPubMed
10.
Zurück zum Zitat Guzman G, Reed MR, Bielamowicz K, Koss B, Rodriguez A. CAR-T therapies in solid tumors: opportunities and challenges. Curr Oncol Rep. 2023;25(5):479–89.CrossRefPubMedPubMedCentral Guzman G, Reed MR, Bielamowicz K, Koss B, Rodriguez A. CAR-T therapies in solid tumors: opportunities and challenges. Curr Oncol Rep. 2023;25(5):479–89.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Ledford H. “It’s all gone”: CAR-T therapy forces autoimmune diseases into remission. Nature. 2023;624(7992):483–4.CrossRefPubMed Ledford H. “It’s all gone”: CAR-T therapy forces autoimmune diseases into remission. Nature. 2023;624(7992):483–4.CrossRefPubMed
14.
Zurück zum Zitat Nakagawa Y, Negishi Y, Shimizu M, Takahashi M, Ichikawa M, Takahashi H. Effects of extracellular pH and hypoxia on the function and development of antigen-specific cytotoxic T lymphocytes. Immunol Lett. 2015;167(2):72–86.CrossRefPubMed Nakagawa Y, Negishi Y, Shimizu M, Takahashi M, Ichikawa M, Takahashi H. Effects of extracellular pH and hypoxia on the function and development of antigen-specific cytotoxic T lymphocytes. Immunol Lett. 2015;167(2):72–86.CrossRefPubMed
15.
Zurück zum Zitat Bosticardo M, Ariotti S, Losana G, Bernabei P, Forni G, Novelli F. Biased activation of human T lymphocytes due to low extracellular pH is antagonized by B7/CD28 costimulation. Eur J Immunol. 2001;31(9):2829–38.CrossRefPubMed Bosticardo M, Ariotti S, Losana G, Bernabei P, Forni G, Novelli F. Biased activation of human T lymphocytes due to low extracellular pH is antagonized by B7/CD28 costimulation. Eur J Immunol. 2001;31(9):2829–38.CrossRefPubMed
16.
Zurück zum Zitat Calcinotto A, Filipazzi P, Grioni M, Iero M, De Milito A, Ricupito A, et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 2012;72(11):2746–56.CrossRefPubMed Calcinotto A, Filipazzi P, Grioni M, Iero M, De Milito A, Ricupito A, et al. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res. 2012;72(11):2746–56.CrossRefPubMed
17.
Zurück zum Zitat Cheng H, Qiu Y, Xu Y, Chen L, Ma K, Tao M, et al. Extracellular acidosis restricts one-carbon metabolism and preserves T cell stemness. Nat Metab. 2023;5(2):314–30.CrossRefPubMedPubMedCentral Cheng H, Qiu Y, Xu Y, Chen L, Ma K, Tao M, et al. Extracellular acidosis restricts one-carbon metabolism and preserves T cell stemness. Nat Metab. 2023;5(2):314–30.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Shao L, Shi R, Zhao Y, Liu H, Lu A, Ma J, et al. Genome-wide profiling of retroviral DNA integration and its effect on clinical pre-infusion CAR T-cell products. J Transl Med. 2022;20(1):514.CrossRefPubMedPubMedCentral Shao L, Shi R, Zhao Y, Liu H, Lu A, Ma J, et al. Genome-wide profiling of retroviral DNA integration and its effect on clinical pre-infusion CAR T-cell products. J Transl Med. 2022;20(1):514.CrossRefPubMedPubMedCentral
19.
20.
Zurück zum Zitat Grist JT, Jarvis LB, Georgieva Z, Thompson S, Kaur Sandhu H, Burling K, et al. Extracellular lactate: a novel measure of T cell proliferation. J Immunol. 2018;200(3):1220–6.CrossRefPubMed Grist JT, Jarvis LB, Georgieva Z, Thompson S, Kaur Sandhu H, Burling K, et al. Extracellular lactate: a novel measure of T cell proliferation. J Immunol. 2018;200(3):1220–6.CrossRefPubMed
21.
Zurück zum Zitat Palmer CS, Ostrowski M, Balderson B, Christian N, Crowe SM. Glucose metabolism regulates T cell activation, differentiation, and functions. Front Immunol. 2015;6:1.CrossRefPubMedPubMedCentral Palmer CS, Ostrowski M, Balderson B, Christian N, Crowe SM. Glucose metabolism regulates T cell activation, differentiation, and functions. Front Immunol. 2015;6:1.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Carswell KS, Papoutsakis ET. Extracellular pH affects the proliferation of cultured human T cells and their expression of the interleukin-2 receptor. J Immunother. 2000;23(6):669–74.CrossRefPubMed Carswell KS, Papoutsakis ET. Extracellular pH affects the proliferation of cultured human T cells and their expression of the interleukin-2 receptor. J Immunother. 2000;23(6):669–74.CrossRefPubMed
23.
Zurück zum Zitat Bohnenkamp H, Hilbert U, Noll T. Bioprocess development for the cultivation of human T-lymphocytes in a clinical scale. Cytotechnology. 2002;38(1–3):135–45.CrossRefPubMedPubMedCentral Bohnenkamp H, Hilbert U, Noll T. Bioprocess development for the cultivation of human T-lymphocytes in a clinical scale. Cytotechnology. 2002;38(1–3):135–45.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat McAdams TA, Miller WM, Papoutsakis ET. Variations in culture pH affect the cloning efficiency and differentiation of progenitor cells in ex vivo haemopoiesis. Br J Haematol. 1997;97(4):889–95.CrossRefPubMed McAdams TA, Miller WM, Papoutsakis ET. Variations in culture pH affect the cloning efficiency and differentiation of progenitor cells in ex vivo haemopoiesis. Br J Haematol. 1997;97(4):889–95.CrossRefPubMed
26.
Zurück zum Zitat Xu XJ, Tang YM. Cytokine release syndrome in cancer immunotherapy with chimeric antigen receptor engineered T cells. Cancer Lett. 2014;343(2):172–8.CrossRefPubMed Xu XJ, Tang YM. Cytokine release syndrome in cancer immunotherapy with chimeric antigen receptor engineered T cells. Cancer Lett. 2014;343(2):172–8.CrossRefPubMed
27.
Zurück zum Zitat Shalabi H, Qin H, Su A, Yates B, Wolters PL, Steinberg SM, et al. CD19/22 CAR T cells in children and young adults with B-ALL: phase 1 results and development of a novel bicistronic CAR. Blood. 2022;140(5):451–63.CrossRefPubMedPubMedCentral Shalabi H, Qin H, Su A, Yates B, Wolters PL, Steinberg SM, et al. CD19/22 CAR T cells in children and young adults with B-ALL: phase 1 results and development of a novel bicistronic CAR. Blood. 2022;140(5):451–63.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Lee DW, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625–38.CrossRefPubMed Lee DW, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625–38.CrossRefPubMed
Metadaten
Titel
Deciphering the importance of culture pH on CD22 CAR T-cells characteristics
verfasst von
Michaela Prochazkova
Alexandra Dreyzin
Lipei Shao
Pam Garces
Yihua Cai
Rongye Shi
Alejandra Pelayo
Yong Soo Kim
Victoria Pham
Sue Ellen Frodigh
Shannon Fenton
Catherine Karangwa
Yan Su
Kathryn Martin
Nan Zhang
Steven L. Highfill
Robert P. Somerville
Nirali N. Shah
David F. Stroncek
Ping Jin
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2024
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-024-05197-5

Weitere Artikel der Ausgabe 1/2024

Journal of Translational Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.