Mathias Wenes’s research while affiliated with University of Geneva and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (31)


A novel mitochondrial pyruvate carrier inhibitor drives stem cell-like memory CAR T cell generation and enhances antitumor efficacy
  • Article

October 2024

·

30 Reads

·

1 Citation

Mathias Wenes

·

Anouk Lepez

·

·

[...]

·

Adoptive cell transfer with chimeric antigen receptor (CAR)-expressing T cells can induce remarkable complete responses in cancer patients. Therapeutic success has been correlated with central and stem cell-like memory T cell subsets in the infusion product, which are better able to drive efficient CAR T cell in vivo expansion and long-term persistence. We previously reported that inhibition of the mitochondrial pyruvate carrier (MPC) during mouse CAR T cell culture induces a memory phenotype and enhances antitumor efficacy against melanoma. Here, we use a novel MPC inhibitor, MITO-66, which robustly induces a stem cell-like memory phenotype in CD19-CAR T cells generated from healthy donors and patients with relapsed/refractory B cell malignancies. MITO-66-conditioned CAR T cells were superior in controlling human pre-B cell acute lymphoblastic leukemia in mice. Following adoptive cell transfer, MITO-66-conditioned CAR T cells maintained a memory phenotype and protected cured mice against tumor rechallenge. Furthermore, in an in vivo B cell leukemia stress model, CD19-CAR T cells generated in the presence of MITO-66 largely outperformed clinical-stage AKT and PI-3Kδ inhibitors. Thus, we provide compelling preclinical evidence that MPC inhibition with MITO-66 during CAR T cell manufacturing dramatically enhances their antitumor efficacy, thereby paving the way to clinical translation.


In vitro characterizations of IL-10 CAR-T cells prior to infusion
a-e, C57BL/6 mice were inoculated subcutaneously (s.c.) with MC38-HER2 colon cancer cells (3 × 10⁵) and received intravenously (i.v.) adoptive cell transfer of HER2 CAR-T cells (3 × 10⁶), untransduced T cells (Ctrl T, equivalent cell number infused), or phosphate buffered saline (PBS) on day 6. Mice were either monitored for tumor growth (n = 5 mice) or sacrificed on day 14 for the analysis of CAR-T cells (n = 10 mice). a, Experimental timeline. b–e, Shown are individual tumor growth curves (b), survival curves (c), frequencies of granzyme B⁺IFNγ⁺TNFα⁺ polyfunctional CAR-T cells in spleen and tumor (d), and MFI of PD-1 and TIM-3 (e). Indicated are numbers of tumor-free mice/total mice in each group (c). f, HER2 CAR or IL-10 HER2 CAR constructs were introduced via retroviral vectors. CAR expression levels were assessed by flow cytometry. Data are one representative of ten independent experiments. g, HER2 CAR-T or IL-10 HER2 CAR-T cells were co-cultured with MC38-HER2 tumor cells for 3 days (n = 3 biologically independent samples)). The culture supernatants were assessed for IL-10 concentration using an enzyme-linked immunosorbent assay (ELISA). h-q, CAR-T cells prior to infusion were examined for the expression levels of indicated markers by flow cytometry (n = 9 biologically independent samples for IL-7Rα, and n = 10 biologically independent samples for others). Shown are average MFI of CD44 (h), CD62L (i), Sca-1 (j), CD122 (k), IL-7Rα (l), KLRG-1 (m), PD-1 (n), TIM-3 (o), and CD69 (p) expression in HER2 CAR-T and IL-10 HER2 CAR-T cells. q, Average frequencies of viable CAR-T cells. All data represent the mean ± s.e.m. and are analyzed by two-tailed Student’s t-test (d, e, g-q), or one- or two-way ANOVA with Tukey’s multiple-comparisons test (b) or log-rank test (c). ns, not significant (P > 0.05). Data are one representative of two independent experiments.
Source data
IL-10 expression shows negligible effects on other immune cells or endogenous T cells in tumor
a-c, The experimental setting was the same as described in Fig. 1b (n = 5 mice). a, Counts of TCF-1⁺PD-1⁺TIM3⁻CD8⁺ HER2 CAR-T cells in tumors. b,c, Frequencies of IFNγ⁺Granzyme B⁺TNFα⁺ polyfunctional cells among PD-1⁺TCF-1⁺TIM3⁻ (b) and PD-1⁺TCF-1⁻TIM3⁺ (c) CD8⁺ CAR-T cells. d, The experimental setting was the same as described in Fig. 1b (n = 10 mice). Counts of indicated immune cell subsets in the MC38-HER2 tumors from each treatment group. e-g, C57BL/6 mice were inoculated s.c. with MC38-HER2 colon cancer cells (1 × 10⁶) and received i.v. adoptive transfer of IL-10 HER2 CAR-T cells (3 × 10⁶) or HER2 CAR-T cells (3 × 10⁶) on day 6 (n = 5 mice). e, Experimental timeline. f, Counts of endogenous T cells. g, Counts of Foxp3⁺CD25⁺CD4⁺ endogenous Tregs. h,i, The experimental setting is described in Fig. 1b. CAR-T cells in tumors were classified into several subpopulations based on the gating of CAR density (Fig. 1j). Shown are response curves of MFI of TNFα (h) and TIM-3 (i) as a function of CAR density (n = 4 mice). Indicated are P values. All data represent the mean ± s.e.m. and are analyzed by two-tailed Student’s t-test (a-c, h, i), or one-way ANOVA with Tukey’s multiple-comparisons test (d, f, g). Data are one representative of two independent experiments.
Source data
Preparation and in vitro characterizations of IL-10-secreting CD19 hCAR-T cells
a, Schematic depicting constructs of CD19-targeted second-generation CD28-based human CAR (CD19 hCAR) and human IL-10-secreting CD19 hCAR (IL-10 CD19 hCAR). b, The expression levels of CD19 hCAR were analyzed by flow cytometry. Ctrl T, untransduced control human T cells. c, The culture supernatants were examined for the concentration of hIL-10 on day 8 after hCAR-T cell preparation (n = 12 biologically independent samples). d, e, CD19 hCAR-T and IL-10 CD19 hCAR-T cells were cocultured with PANC1-CD19 cells or Raji cells at different E:T ratios for 48 h (n = 3 biologically independent samples). Shown are the percentage of lysis of PANC1-CD19 cell (d) or Raji cell (e). f,g, The proliferation of CD19 hCAR and IL-10 CD19 hCAR-T cells after multiple weekly stimulation (indicated by arrows; n = 3) with irradiated NIH/3T3-CD19 (f) or PANC1-CD19 (g) cells (n = 3 biologically independent samples). All data represent the mean ± s.e.m. and are analyzed by two-tailed Student’s t-test. Data are one representative of two independent experiments.
Source data
IL-10 expression improved mitochondrial fitness and increased the levels of pyruvate production intermediates and TCA cycle intermediates in CAR-T cells
The experimental setting is described in Fig. 2a. a, MFI of PD-1, TIM-3, and CD107a in MDR/MGhi and MDR/MGlo CAR-T cells in tumor (n = 10 mice). b, Quantification of roundness index of mitochondria in CAR-T cells as shown in Fig. 2i (n = 15 biologically independent samples). c, Representative contour plot showing CAR-T cells in tumor were classified into three subpopulations based on CAR density. d-f, The experimental setting is described in Fig. 3a-c (n = 3 biologically independent samples). d, Real-time analysis of ECAR. e, Average basal and maximal ECAR. f, Average basal and maximal OCR of HER2 CAR-T and IL-10 HER2 CAR-T cells in the presence or absence of IL-10 without antigen stimulation. g-j, C57BL/6 mice were inoculated s.c. with MC38-HER2 colon cancer cells (1 × 10⁶), lymphodepleted, and received IL-10 HER2 CAR-T cells or HER2 CAR-T cells (3 × 10⁶, i.v.) on day 6. On day 14, tumor-infiltrating CAR-T cells were sorted for metabolomics analysis (n = 3 biologically independent samples, each sample is pooled from n = 5 mice). g, Experimental timeline. h, Clustered heatmap of significantly altered metabolites (P < 0.05) in HER2 CAR-T and IL-10 HER2 CAR-T cells. The color blocks indicate relative metabolite levels, with high expression in red and low expression in green. i,j, Fold changes of phosphoenolpyruvate (i) and succinate (j) (relative to HER2 CAR-T cells). k,l, HER2 CAR-T or IL-10 HER2 CAR-T cells were cocultured with MC38-HER2 cells in the presence of indicated inhibitors (n = 3 biologically independent samples). Shown are relative CAR-T cell counts (k) and relative frequencies of granzyme B⁺IFNγ⁺TNFα⁺ polyfunctional CAR-T cells (l) (IL-10 HER2 CAR-T vs. HER2 CAR-T). Data are one representative of two independent experiments. All data represent the mean ± s.e.m. and are analyzed by two-tailed Student’s t-test (a, b, i, j) or by one- or two-way ANOVA with Tukey’s multiple-comparisons test (e, f, k, l).
Source data
IL-10-expressing HER2 CAR-T cells exhibit an expanded subpopulation of CD8⁺ terminally exhausted T cells with upregulated gene expression encoding cell cycling, cytotoxicity, and OXPHOS
a, The experimental setting and timeline is described in Fig. 4. b, Heatmap representing the expression levels of representative marker genes in each cluster. c, Projection of IL-10 HER2 CAR-T cells and HER2 CAR-T cells into a reference atlas of tumor-infiltrating lymphocytes. Reference atlas states are indicated as colored cells. CD8⁺ terminally-exhausted T cell cluster was characterized as high expression of granzymes, multiple inhibitory receptors (Pdcd1, Ctla4, Lag3, Tigit, Havcr2/TIM-3, etc.) and Tox¹⁶. Shown are cell subtype compositions for each group. Black contour lines represent the density of projected cells. d, Gene signature scores for each group of samples (HER2 CAR-T and IL-10 HER2 CAR-T cells) for the indicated gene sets.

+11

IL-10-expressing CAR T cells resist dysfunction and mediate durable clearance of solid tumors and metastases
  • Article
  • Publisher preview available

January 2024

·

1,399 Reads

·

85 Citations

Nature Biotechnology

The success of chimeric antigen receptor (CAR) T cell therapy in treating several hematopoietic malignancies has been difficult to replicate in solid tumors, in part because of T cell exhaustion and eventually dysfunction. To counter T cell dysfunction in the tumor microenvironment, we metabolically armored CAR T cells by engineering them to secrete interleukin-10 (IL-10). We show that IL-10 CAR T cells preserve intact mitochondrial structure and function in the tumor microenvironment and increase oxidative phosphorylation in a mitochondrial pyruvate carrier-dependent manner. IL-10 secretion promoted proliferation and effector function of CAR T cells, leading to complete regression of established solid tumors and metastatic cancers across several cancer types in syngeneic and xenograft mouse models, including colon cancer, breast cancer, melanoma and pancreatic cancer. IL-10 CAR T cells also induced stem cell-like memory responses in lymphoid organs that imparted durable protection against tumor rechallenge. Our results establish a generalizable approach to counter CAR T cell dysfunction through metabolic armoring, leading to solid tumor eradication and long-lasting immune protection.

View access options

Cytokine-armed dendritic cell progenitors for antigen-agnostic cancer immunotherapy

November 2023

·

309 Reads

·

24 Citations

Nature Cancer

Dendritic cells (DCs) are antigen-presenting myeloid cells that regulate T cell activation, trafficking and function. Monocyte-derived DCs pulsed with tumor antigens have been tested extensively for therapeutic vaccination in cancer, with mixed clinical results. Here, we present a cell-therapy platform based on mouse or human DC progenitors (DCPs) engineered to produce two immunostimulatory cytokines, IL-12 and FLT3L. Cytokine-armed DCPs differentiated into conventional type-I DCs (cDC1) and suppressed tumor growth, including melanoma and autochthonous liver models, without the need for antigen loading or myeloablative host conditioning. Tumor response involved synergy between IL-12 and FLT3L and was associated with natural killer and T cell infiltration and activation, M1-like macrophage programming and ischemic tumor necrosis. Antitumor immunity was dependent on endogenous cDC1 expansion and interferon-γ signaling but did not require CD8 ⁺ T cell cytotoxicity. Cytokine-armed DCPs synergized effectively with anti-GD2 chimeric-antigen receptor (CAR) T cells in eradicating intracranial gliomas in mice, illustrating their potential in combination therapies.


Deletion of IDH2 inhibits RC in TE cells and promotes the differentiation of TM cells
a, Oxidative (green) and reductive (red) glutamine metabolism. Ac-CoA, acetyl-CoA; OAA, oxaloacetate. b, Citrate mass isotopologues in mouse CD8⁺ TE cells labelled with [U-¹³C]-glutamine (n = 3 biological replicates). c, Ratio of m + 5 to m + 4 citrate detected by [U-¹³C]-glutamine labelling in TE or TM cells (n = 4 biological replicates). d, Gene expression of IDH1 and IDH2 in CD8⁺ T cells from volunteers who were vaccinated against yellow fever. Data are reads per kilobase of transcript per million reads mapped (RPKM) in the indicated cell subsets (n = 3 (TE), n = 5 (TM) and n = 6 (TN) biological replicates). e, Percentage of m + 1 citrate labelling from [1-¹³C]-glutamine in control scramble (Scr) gRNA-transduced cells or IDH2-deficient cells (n = 3 biological replicates). f, Schematic representation of the experiment. D, day. g, Number of Thy1.1⁺ cells per microlitre of blood. h, Percentage of CD44⁺CD62L⁺ cells among Thy1.1⁺ cells in the blood 28 days after infection. i, Number of Thy1.1⁺ cells per milligram of spleen seven days after secondary challenge. j,k, Percentages of IL-2⁺ (j) and IFNγ⁺TNF⁺ (k) cells among Thy1.1⁺ cells after restimulation seven days after secondary challenge. l,m, LCMV-Armstrong viral titres per kidney (l) and liver (m). PFU, plaque-forming units. In g,h: n = 13 (Idh2 gRNA) and n = 11 (Scr gRNA) biological replicates; pooled data from three independent experiments. In i–m: n = 8 (Idh2 gRNA) and n = 9 (Scr gRNA) biological replicates; pooled data from two independent experiments. Data are mean ± s.e.m. and were analysed by one-way ANOVA using Tukey’s multiple comparison test (d) or unpaired, two-tailed Student’s t-test (c,e,h–m). The P value in c is 0.00005633.
Source data
Inhibition of IDH2 improves the function of CAR T cells
a, Tumour growth of B16-HER2 tumour-bearing mice treated with DMSO-conditioned or IDH2i-conditioned BFP-transduced T cells or HER2-CAR T cells. b–d, Number of HER2-CAR⁺ T cells (b) and their percentage of TCF1⁺ (c) and CD44⁺CD62L⁺ cells (d) in tumour-draining lymph nodes (dLNs) at dissection. e, Number of HER2-CAR⁺ TILs per milligram of tumour at dissection. f,g, Percentages of PD-1⁺TCF1⁺ cells (f) and PD-1⁺TCF1⁻ cells (g) among HER2-CAR⁺ TILs. h, Percentage of IFNγ⁺TNF⁺IL-2⁺ HER2-CAR⁺ TILs. In a–h: n = 12 (IDH2i) and n = 13 (DMSO) biological replicates; pooled data from two independent experiments. i, Mean fluorescence intensity (MFI) of CD62L out of CD62L⁺ CD19-CAR⁺ CD8⁺ T cells nine days after activation, in mice infused with non-transduced (NTD) or anti-CD19 CAR T cells (n = 3 biological replicates; pooled data from two independent experiments). j, Survival curve of NALM6 tumour-bearing mice infused with NTD or anti-CD19 CAR T cells. k, Number of CD19⁺ NALM6 cells per microlitre of blood at days 14, 21 and 25 after tumour engraftment. In j,k: n = 7 (DMSO or IDH2i CAR), n = 5 (DMSO or IDH2i NTD) and n = 4 (untreated) mice; pooled data from two independent experiments using three human donors. l, MFI of CD62L out of CD62L⁺ BCMA-CAR⁺ CD8⁺ T cells nine days after activation, in mice infused with NTD or BCMA CAR T cells (n = 6 biological replicates; pooled data from two independent experiments). m, Survival curve of NCI-H929 tumour-bearing mice treated by ACT. n, Representative bioluminescence images of NCI-H929 tumour burden. In m,n: n = 19 (DMSO or IDH2i CAR), n = 9 (DMSO or IDH2i NTD) and n = 8 (untreated) mice; pooled data from two independent experiments using two human donors. Data are mean ± s.e.m. and were analysed by unpaired (a–h) or paired (i,l) two-tailed Student’s t-test, log-rank test (j,m) or two-way ANOVA using the original false discovery rate (FDR) test of Benjamini and Hochberg (k).
Source data
IDH2 inhibition leaves an epigenetic imprint
a, OVA-specific CD8⁺ T cells were activated and cultured for seven days with IDH2i or DMSO, followed by RNA-seq analysis. Shown are pathways significantly enriched among upregulated genes after IDH2 inhibition, identified by Enrichr²³ in the Elsevier Pathway Collection (https://www.elsevier.com/solutions/pathway-studio-biological-research) (n = 3 biological replicates). LDL, low-density lipoprotein. b, GSEA of a memory-versus-effector signature from genes upregulated in IDH2i- compared to DMSO-conditioned T cells. NES, normalized enrichment score; running ES, running enrichment score. c, Representative ATAC-seq tracks in OVA-specific CD8⁺ T cells activated and cultured for seven days with IDH2i or DMSO (n = 3 biological replicates). d, GSEA of a memory-versus-effector signature on more accessible chromatin regions after IDH2 inhibition. e, Differentially accessible chromatin regions in DMSO- versus IDH2i-conditioned cells (FDR P < 0.05), which were correlated with gene-expression data (log2-transformed fold change > 1 and Padj < 0.05). f, Over-representation analysis of the indicated gene signatures from genes exhibiting higher chromatin accessibility and transcription after IDH2 inhibition. g, Top transcription factors identified in more open chromatin regions of IDH2i-conditioned cells using HOMER. h, Top five most significant mouse transcription factors identified by Enrichr in the TRRUST 2019 database (https://www.grnpedia.org/trrust/) in upregulated genes after IDH2 inhibition. Histograms depict negative log of P values (−log10 > 1.5 and P < 0.05). i, Significantly enriched transcription factors in more accessible chromatin regions that correlated with increased gene expression after IDH2 blockade. Transcription factors were found in the TRRUST 2019 database (https://www.grnpedia.org/trrust/) using Enrichr software. Histograms depict negative log of P values (−log10 > 1.5 and P < 0.05).
Source data
IDH2 inhibition alters the balance of metabolites, which dictates epigenetic memory
a, Quantification of metabolites in TE cells. The TCA cycle is depicted in blue, with purple circles representing quantified metabolites and grey circles representing metabolites that were not quantified. IDH2 inhibition is represented by a red cross. (n = 6 biological replicates; pooled data from two independent experiments). b, Acetyl-CoA abundance (n = 3 biological replicates). a.u., arbitrary units. c, Percentages of acetyl-CoA m + 2 labelled by [U-¹³C]-glutamine, [U-¹³C]-glucose or [U-¹³C]-palmitate (n = 3 biological replicates). d, OCR by Seahorse (n = 4 biological replicates; pooled data from two independent experiments). e, Reduction in basal OCR after addition of BPTES, UK5099 or etomoxir (n = 4 (UK and BPTES) and n = 5 (Eto) biological replicates; pooled data from two independent experiments). f, Glutamine consumption in TE cells (n = 3 biological replicates). g, Immunoblot quantification of H3K4me3 (n = 5 biological replicates; pooled data from five independent experiments). h, Immunoblot of H3K4me3 in T cells supplemented or not with dimethyl 2-oxoglutarate (an analogue of α-KG). Representative of four independent experiments with four biological replicates. i, CD62L expression in T cells supplemented with α-KG (n = 3 biological replicates; pooled data from three independent experiments). j, Immunoblot of histone marks after supplementation with cell-permeable metabolites. Representative blot from three independent experiments with three biological replicates. k, CD62L expression in T cells with the indicated treatments. Representative histograms from three independent experiments with three biological replicates. l, KDM5 activity after treatment with DMSO or IDH2i, with α-KG supplementation. RFU, relative fluorescence units (n = 3 biological replicates). m,n, Immunoblot of histone marks (m) and CD62L expression (n). KDM5i, KDM5 inhibitor. Representative of two (m) or three (n) independent experiments with two (m) or 3 (n) biological replicates). Data are mean ± s.e.m. and were analysed by unpaired two-tailed Student’s t-test (b,c,e–g), multiple unpaired two-tailed t-tests using the Benjamini and Hochberg method (a), one-way ANOVA using Tukey’s multiple comparison test (i) or two-way ANOVA using the original FDR test of Benjamini and Hochberg (l). For gel source data, see Supplementary Fig. 1.
Source data
Reductive carboxylation epigenetically instructs T cell differentiation

September 2023

·

439 Reads

·

50 Citations

Nature

Protective immunity against pathogens or cancer is mediated by the activation and clonal expansion of antigen-specific naive T cells into effector T cells. To sustain their rapid proliferation and effector functions, naive T cells switch their quiescent metabolism to an anabolic metabolism through increased levels of aerobic glycolysis, but also through mitochondrial metabolism and oxidative phosphorylation, generating energy and signalling molecules. However, how that metabolic rewiring drives and defines the differentiation of T cells remains unclear. Here we show that proliferating effector CD8+ T cells reductively carboxylate glutamine through the mitochondrial enzyme isocitrate dehydrogenase 2 (IDH2). Notably, deletion of the gene encoding IDH2 does not impair the proliferation of T cells nor their effector function, but promotes the differentiation of memory CD8+ T cells. Accordingly, inhibiting IDH2 during ex vivo manufacturing of chimeric antigen receptor (CAR) T cells induces features of memory T cells and enhances antitumour activity in melanoma, leukaemia and multiple myeloma. Mechanistically, inhibition of IDH2 activates compensating metabolic pathways that cause a disequilibrium in metabolites regulating histone-modifying enzymes, and this maintains chromatin accessibility at genes that are required for the differentiation of memory T cells. These findings show that reductive carboxylation in CD8+ T cells is dispensable for their effector response and proliferation, but that it mainly produces a pattern of metabolites that epigenetically locks CD8+ T cells into a terminal effector differentiation program. Blocking this metabolic route allows the increased formation of memory T cells, which could be exploited to optimize the therapeutic efficacy of CAR T cells.


IOA-244 is a non-ATP-competitive, highly selective, tolerable phosphoinositide 3-kinase delta inhibitor that targets solid tumors and breaks immune tolerance

March 2023

·

117 Reads

·

14 Citations

Cancer Research Communications

PI3K delta (PI3Kδ) inhibitors are used to treat lymphomas but safety concerns and limited target selectivity curbed their clinical usefulness. PI3Kδ inhibition in solid tumors has recently emerged as a potential novel anticancer therapy through the modulation of T-cell responses and direct antitumor activity. Here we report the exploration of IOA-244/MSC2360844, a first-in-class non–ATP-competitive PI3Kδ inhibitor, for the treatment of solid tumors. We confirm IOA-244’s selectivity as tested against a large set of kinases, enzymes, and receptors. IOA-244 inhibits the in vitro growth of lymphoma cells and its activity correlates with the expression levels of PIK3CD, suggesting cancer cell–intrinsic effects of IOA-244. Importantly, IOA-244 inhibits regulatory T cell proliferation while having limited antiproliferative effects on conventional CD4+ T cells and no effect on CD8+ T cells. Instead, treatment of CD8 T cells with IOA-244 during activation, favors the differentiation of memory-like, long-lived CD8, known to have increased antitumor capacity. These data highlight immune-modulatory properties that can be exploited in solid tumors. In CT26 colorectal and Lewis lung carcinoma lung cancer models, IOA-244 sensitized the tumors to anti-PD-1 (programmed cell death protein 1) treatment, with similar activity in the Pan-02 pancreatic and A20 lymphoma syngeneic mouse models. IOA-244 reshaped the balance of tumor-infiltrating cells, favoring infiltration of CD8 and natural killer cells, while decreasing suppressive immune cells. IOA-244 presented no detectable safety concerns in animal studies and is currently in clinical phase Ib/II investigation in solid and hematologic tumors. Significance IOA-244 is a first-in-class non–ATP-competitive, PI3Kδ inhibitor with direct antitumor in vitro activity correlated with PI3Kδ expression. The ability to modulate T cells, in vivo antitumor activity in various models with limited toxicity in animal studies provides the rationale for the ongoing trials in patients with solid tumors and hematologic cancers.


Efficacy and safety of universal (TCRKO) ARI-0001 CAR-T cells for the treatment of B-cell lymphoma

October 2022

·

259 Reads

·

12 Citations

Autologous T cells expressing the Chimeric Antigen Receptor (CAR) have been approved as advanced therapy medicinal products (ATMPs) against several hematological malignancies. However, the generation of patient-specific CAR-T products delays treatment and precludes standardization. Allogeneic off-the-shelf CAR-T cells are an alternative to simplify this complex and time-consuming process. Here we investigated safety and efficacy of knocking out the TCR molecule in ARI-0001 CAR-T cells, a second generation αCD19 CAR approved by the Spanish Agency of Medicines and Medical Devices (AEMPS) under the Hospital Exemption for treatment of patients older than 25 years with Relapsed/Refractory acute B cell lymphoblastic leukemia (B-ALL). We first analyzed the efficacy and safety issues that arise during disruption of the TCR gene using CRISPR/Cas9. We have shown that edition of TRAC locus in T cells using CRISPR as ribonuleorproteins allows a highly efficient TCR disruption (over 80%) without significant alterations on T cells phenotype and with an increased percentage of energetic mitochondria. However, we also found that efficient TCRKO can lead to on-target large and medium size deletions, indicating a potential safety risk of this procedure that needs monitoring. Importantly, TCR edition of ARI-0001 efficiently prevented allogeneic responses and did not detectably alter their phenotype, while maintaining a similar anti-tumor activity ex vivo and in vivo compared to unedited ARI-0001 CAR-T cells. In summary, we showed here that, although there are still some risks of genotoxicity due to genome editing, disruption of the TCR is a feasible strategy for the generation of functional allogeneic ARI-0001 CAR-T cells. We propose to further validate this protocol for the treatment of patients that do not fit the requirements for standard autologous CAR-T cells administration.


The mitochondrial pyruvate carrier regulates memory T cell differentiation and antitumor function

April 2022

·

175 Reads

·

119 Citations

Cell Metabolism

Glycolysis, including both lactate fermentation and pyruvate oxidation, orchestrates CD8⁺ T cell differentiation. However, how mitochondrial pyruvate metabolism and uptake controlled by the mitochondrial pyruvate carrier (MPC) impact T cell function and fate remains elusive. We found that genetic deletion of MPC drives CD8⁺ T cell differentiation toward a memory phenotype. Metabolic flexibility induced by MPC inhibition facilitated acetyl-coenzyme-A production by glutamine and fatty acid oxidation that results in enhanced histone acetylation and chromatin accessibility on pro-memory genes. However, in the tumor microenvironment, MPC is essential for sustaining lactate oxidation to support CD8⁺ T cell antitumor function. We further revealed that chimeric antigen receptor (CAR) T cell manufacturing with an MPC inhibitor imprinted a memory phenotype and demonstrated that infusing MPC inhibitor-conditioned CAR T cells resulted in superior and long-lasting antitumor activity. Altogether, we uncover that mitochondrial pyruvate uptake instructs metabolic flexibility for guiding T cell differentiation and antitumor responses.




Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity

June 2021

·

1,264 Reads

·

264 Citations

Nature Immunology

T cell exhaustion presents one of the major hurdles to cancer immunotherapy. Among exhausted CD8+ tumor-infiltrating lymphocytes, the terminally exhausted subset contributes directly to tumor cell killing owing to its cytotoxic effector function. However, this subset does not respond to immune checkpoint blockades and is difficult to be reinvigorated with restored proliferative capacity. Here, we show that a half-life-extended interleukin-10–Fc fusion protein directly and potently enhanced expansion and effector function of terminally exhausted CD8+ tumor-infiltrating lymphocytes by promoting oxidative phosphorylation, a process that was independent of the progenitor exhausted T cells. Interleukin-10–Fc was a safe and highly efficient metabolic intervention that synergized with adoptive T cell transfer immunotherapy, leading to eradication of established solid tumors and durable cures in the majority of treated mice. These findings show that metabolic reprogramming by upregulating mitochondrial pyruvate carrier-dependent oxidative phosphorylation can revitalize terminally exhausted T cells and enhance the response to cancer immunotherapy. Tang and colleagues show that a half-life-extended IL-10–Fc fusion protein acts directly on terminally exhausted PD1+TIM-3+CD8+ T cells to enhance their proliferation and effector function by reprogramming the cellular metabolism to oxidative phosphorylation in a mitochondrial pyruvate carrier–dependent manner. Treatment of tumor-bearing mice with IL-10–Fc and adoptive T cell therapy led to eradication of their established solid tumors and durable cures.


Citations (22)


... These findings underscore entacapone's potential off-target effects and suggest its utility as a scaffold for developing new MPC inhibitors. Table 3) as a novel MPC inhibitor designed to enhance the efficacy of CD19-CAR T cells in antitumor treatment [99]. MITO-66 inhibits MPC with an IC 50 of 119 nM, though its molecular structure has not been reported. ...

Reference:

Advances in the Development of Mitochondrial Pyruvate Carrier Inhibitors for Therapeutic Applications
A novel mitochondrial pyruvate carrier inhibitor drives stem cell-like memory CAR T cell generation and enhances antitumor efficacy
  • Citing Article
  • October 2024

... Of note, we also observed IL-4 and IL10 upregulation in activated MET-CAR T cells (Fig. 5). Both IL-4 and IL-10 are type 2 cytokines contributing to an immunosuppressive TME by activating tumorassociated M2 macrophage [46]; however, recent studies [50]. Thus, further investigations are needed to understand their functions during MET-CAR T cell therapy. ...

IL-10-expressing CAR T cells resist dysfunction and mediate durable clearance of solid tumors and metastases

Nature Biotechnology

... 20 22 Strategies to empower cDC1 immunostimulatory potential are beginning Open access to show promising results in lung cancer models and clinical trials. [23][24][25][26] Here, we will discuss key general concepts and focus specifically on cDC1 in lung cancer, in humans and relevant experimental models. ...

Cytokine-armed dendritic cell progenitors for antigen-agnostic cancer immunotherapy

Nature Cancer

... Isocitrate dehydrogenase 2 (IDH2) restricts the pentose phosphate pathway and cytoplasmic citrate availability, thereby decreasing acetyl-CoA levels. Treatment with IDH2 inhibitors decreases the expression of multiple genes in the glycolytic pathway by increasing histone acetylation, and leads to maintenance of chromatin accessibility for genes essential for memory T cell differentiation 117 . Down-regulation of CD38-mediated signaling pathways, inosine supplementation, and inhibition of mitochondrial pyruvate carriers have also been shown to inhibit glycolysis, increase mitochondrial activity, and induce the transcription of stem cell-associated genes via epigenetic regulation [118][119][120] . ...

Reductive carboxylation epigenetically instructs T cell differentiation

Nature

... Class I is further divided into IA and IB, with the catalytic subunit p110 of class IA comprising four isoforms: p110α, p110β, p110γ, and p110CD, encoded by the genes PI3KCA, PI3KCB, PI3KCG, and PI3KCD, respectively [9]. While PI3KCA is largely implicated in solid tumors such as GC, PI3KCD, traditionally associated with hematological cancers, has recently garnered interest for its role in solid tumors, including GC, due to its immunomodulatory effects and impact on cell-mediated tolerance to cancer [10]. The other types of PI3Ks include PI3K alpha and gamma, PI3Kα is a class I PI3K primarily involved in cell growth, and PI3Kγ plays a vital role in chemokine-dependent migration of neutrophils and macrophages [11]. ...

IOA-244 is a non-ATP-competitive, highly selective, tolerable phosphoinositide 3-kinase delta inhibitor that targets solid tumors and breaks immune tolerance

Cancer Research Communications

... HD-derived CAR-T cells have been found to contribute to more rapid tumor control and higher survival rates in lymphoma mouse models compared to CAR-T cells derived from patients 130 . TRAC-KO CAR-T cells obtained with CRISPR/Cas9 are essentially the same as wild-type CAR-T cells in terms of phenotype, antitumor ability, and mitochondrial activity, and have been found to improve the survival rate without GVHD incidence in a mouse model of Burkitt's lymphoma 131 . Another source of immunogenicity is allogeneic MHC. ...

Efficacy and safety of universal (TCRKO) ARI-0001 CAR-T cells for the treatment of B-cell lymphoma

... Emerging evidence emphasizes the role of mitochondrial metabolism in shaping T cell behavior, influencing epigenetic features and functionality. [8][9][10][11][12] While T eff cells rely on glycolysis and one-carbon metabolism, 11 T m cells adopt a distinct metabolic profile characterized by increased fatty acid oxidation (FAO) and mitochondrial spare respiratory capacity (SRC), supporting long-term survival. This metabolic shift is vital for memory and stem-like properties. ...

The mitochondrial pyruvate carrier regulates memory T cell differentiation and antitumor function

Cell Metabolism

... The latter has been described so far only for a half-life extended IL-10-Fc fusion cytokine. 46 IL-10-Fc was shown to directly enhance CD8 + Tex expansion and effector function by promoting their oxidative phosphorylation and metabolic reprogramming. 46 Although an enhanced oxidative phosphorylation gene signature in CD8 + Tex cells on mSOT201 treatment was detected (data not shown), it remains to be further investigated if mSOT201 induces a similar metabolic reprogramming. ...

Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity

Nature Immunology

... 116 Another study conducted by Jaccard et al indicates that pharmacological suppression of the metabolic enzyme isocitrate dehydrogenase 2 (IDH2) during CD8 + T cell priming resulted in greater memory formation and tumor growth inhibition upon adoptive cellular therapy (ACT) into melanoma tumor-bearing mice. 117 In addition, manipulation of cellular fatty acid metabolism may potentially be of therapeutic relevance, since changes in basic cellular lipid metabolism can have a major impact on T cell destiny and function. 118 Fatty acid synthesis (FAS) promotes the proliferation and differentiation of Teff cells in response to stimulation, whereas FAO is required for the formation of CD8 + T cell memory cells. ...

515 Metabolic reprogramming of antitumor CD8+ T cell immunity

... LAA treatment enhanced PGC-1α expression in SplCs in the present study. PGC-1α is a mitochondrial biogenesis marker [61], the increased expression of which promotes memory T cell formation [62]. Memory T cells are mainly metabolized via OXPHOS and fatty acid oxidation [63], and ketone bodies epigenetically regulate memory T cell formation and maintenance [64]. ...

Enforced PGC-1α expression promotes CD8 T cell fitness, memory formation and antitumor immunity

Cellular & Molecular Immunology