The improved detection of this patient's post-CAR T-cell therapy relapse, using peripheral blood minimal residual disease (MRD) and 18F-fluorodeoxyglucose positron emission tomography (PET) imaging, highlights a superior sensitivity to the standard bone marrow aspiration technique. Relapse patterns in relapsed B-ALL cases, often encompassing dispersed medullary and/or extramedullary disease manifestations, may be more effectively detected through peripheral blood minimal residual disease monitoring and/or whole-body imaging approaches, compared to the standard bone marrow biopsy approach for certain patient cohorts.
In this instance, both peripheral blood MRD and 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) imaging demonstrated heightened sensitivity in identifying post-CAR T-cell therapy relapse in this patient, in contrast to standard bone marrow biopsy. Clinical/biologic insights in multiply relapsed B-ALL, characterized by potentially patchy medullary and/or extramedullary disease, might reveal increased sensitivity in detecting relapse using peripheral blood minimal residual disease (MRD) and/or whole-body imaging compared to traditional bone marrow examination in select patient populations.
The presence of cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) is detrimental to the function of natural killer (NK) cells, a promising avenue for therapeutic intervention. The intricate relationship between cancer-associated fibroblasts (CAFs) and natural killer (NK) cells within the tumor microenvironment (TME) profoundly inhibits immune responses, thus highlighting the prospect of CAF-targeted therapies as a potential means to achieve more effective NK-mediated cancer cell killing.
To combat the CAF-induced suppression of NK cell function, we have chosen nintedanib, an antifibrotic drug, as part of a synergistic therapeutic combination. We generated an in vitro 3D spheroid model comprising Capan2 cells and patient-derived CAF cells, or an in vivo mixed Capan2/CAF tumor xenograft model, to quantify the synergistic therapeutic efficacy. Through in vitro studies, the molecular mechanism of the synergistic therapeutic combination of nintedanib and NK cells was elucidated. In vivo, the efficacy of the combined therapy was subsequently assessed. Using immunohistochemistry, the expression scores of target proteins were ascertained in patient-derived tumor tissue samples.
By inhibiting the platelet-derived growth factor receptor (PDGFR) signaling pathway, nintedanib suppressed CAF activation and growth, significantly decreasing the IL-6 released by CAFs. Co-treatment with nintedanib also improved the efficacy of mesothelin (MSLN) targeting chimeric antigen receptor (CAR)-NK cell-mediated tumor killing in CAF/tumor spheroids or xenograft models. A synergistic interaction, within the living system, triggered a substantial infiltration of natural killer cells. In contrast to the lack of effect from nintedanib alone, blocking IL-6 trans-signaling promoted the activity of NK cells. A notable outcome arises from the concurrence of MSLN expression and PDGFR activation.
Inferior clinical outcomes were statistically associated with a particular CAF population area, a potential prognostic and therapeutic indicator.
Our approach to managing PDGFR.
The presence of CAF in pancreatic cancer facilitates advancements in the therapeutic approach to pancreatic ductal adenocarcinoma.
By targeting PDGFR+-CAF-containing pancreatic cancer, our strategy fosters improvements in the treatment of pancreatic ductal adenocarcinoma.
Chimeric antigen receptor (CAR) T-cell therapy encounters significant obstacles in treating solid tumors, including the limited persistence of the introduced T cells, their restricted ability to enter and stay within the tumor, and the immunosuppressive nature of the tumor's microenvironment. Up to this point, the efforts to clear these hurdles have fallen short of expectations. Herein, we present a combined strategy.
Generating CAR-T cells with both central memory and tissue-resident memory characteristics, to address these limitations, necessitates the combination of ex vivo protein kinase B (AKT) inhibition and RUNX family transcription factor 3 overexpression.
Second-generation murine CAR-T cells, designed to express a CAR targeting human carbonic anhydrase 9, were engineered and produced.
Overexpression of these elements was amplified in the presence of AKTi-1/2, a reversible and selective inhibitor of AKT1/AKT2. We researched the consequences of AKT pathway blockade (AKTi).
Phenotypes of CAR-T cells, in response to overexpression and their combined treatment, were investigated via flow cytometry, transcriptome profiling, and mass cytometry. CAR-T cell persistence, tumor-infiltration capabilities, and antitumor effectiveness were examined within subcutaneous pancreatic ductal adenocarcinoma (PDAC) tumor models.
AKTi's method cultivated a population of CAR-T cells, expressing CD62L and central memory characteristics, with enhanced persistence and preserved cytotoxic potential.
3-overexpression and AKTi's joint efforts yielded CAR-T cells that displayed central memory and tissue-resident memory characteristics.
Overexpression's contribution to the heightened capacity of CD4+CAR T cells, interacting with AKTi, restrained the terminal differentiation of CD8+CAR T cells, a consequence of consistent stimulation. CAR-T cell central memory phenotype enhancement, along with a prominent improvement in expansion ability, was achieved through AKTi.
Enhanced CAR-T cell overexpression resulted in a tissue-resident memory phenotype and a heightened degree of persistence, effector function, and tumor residence. ML324 price These are novelties, originating from AKTi generation.
In subcutaneous PDAC tumor models, overexpressed CAR-T cells showcased impressive antitumor activity, accompanied by a favorable response to programmed cell death 1 blockade.
Ex vivo AKTi, combined with overexpression strategies, yielded CAR-T cells with prominent tissue-resident and central memory traits, thus bolstering their persistence, cytotoxic properties, and tumor-infiltrating potential, consequently overcoming barriers in solid tumor therapy.
The combined effects of Runx3 overexpression and ex vivo AKTi on CAR-T cells resulted in cells with both tissue-resident and central memory qualities. This augmented their persistence, cytotoxic potential, and capacity to reside in tumors, offering an improved therapeutic approach for solid tumors.
Immune checkpoint blockade (ICB) shows restricted impact on hepatocellular carcinoma (HCC) outcomes. The current study examined the feasibility of utilizing tumor metabolic shifts to boost HCC's responsiveness to immunotherapeutic interventions.
Paired tissue samples (non-tumor and tumor) from hepatocellular carcinoma (HCC) were examined for levels of one-carbon (1C) metabolism and the expression of phosphoserine phosphatase (PSPH), an enzyme upstream in the 1C pathway. This investigation further assessed the role of PSPH in the regulation of monocyte/macrophage and CD8+ T-cell infiltration.
Employing in vitro and in vivo experimental setups, researchers examined T lymphocytes.
Hepatocellular carcinoma (HCC) tumor tissues demonstrated a marked increase in PSPH expression, a factor positively linked to disease progression. ML324 price PSPH knockdown resulted in tumor growth suppression in immunocompetent mice, but this suppression was absent in mice lacking either macrophages or T lymphocytes, indicating that PSPH's promotion of tumor growth is contingent upon both immune cell types. The mechanism by which PSPH functioned entailed the induction of C-C motif chemokine 2 (CCL2), thereby increasing the infiltration of monocytes/macrophages, however, this was accompanied by a decrease in the count of CD8 cells.
Tumor necrosis factor alpha (TNF-) conditioned cancer cells, by inhibiting the production of C-X-C Motif Chemokine 10 (CXCL10), contribute to the recruitment of T lymphocytes. The production levels of CCL2 and CXCL10 were partly influenced by glutathione and S-adenosyl-methionine, respectively. ML324 price A list of sentences forms the output of this JSON schema.
The in vivo application of (short hairpin RNA) to cancer cells boosted their sensitivity to anti-programmed cell death protein 1 (PD-1) treatment. Remarkably, metformin proved capable of inhibiting PSPH expression in cancer cells, mimicking the results seen with shRNA.
Tumor susceptibility to anti-PD-1 therapies is heightened in this procedure.
PSPH's ability to influence the immune response in a way that favors tumor growth could make it a valuable marker for selecting patients appropriate for immune checkpoint blockade therapies and a compelling target for treating human hepatocellular carcinoma.
The potential of PSPH to alter the immune environment, creating a more tumor-conducive one, suggests its application as a stratification factor for immunotherapy patients and a compelling therapeutic option for human hepatocellular carcinoma.
The presence of PD-L1 (CD274) amplification in a limited number of malignancies might potentially predict the success of anti-PD-1/PD-L1 immunotherapy. A hypothesis was formed suggesting that both copy number (CN) and the localization of cancer-associated PD-L1 amplifications affect protein expression, leading us to examine solid tumors comprehensively profiled at Foundation Medicine from March 2016 through February 2022. A comparative genomic hybridization-like method was used to detect PD-L1 CN alterations. IHC staining using the DAKO 22C3 antibody for PD-L1 protein showed a relationship between PD-L1 copy number (CN) changes and PD-L1 expression. From the analysis of 60,793 samples, the most frequently observed histologies were lung adenocarcinoma (20% of the total), colon adenocarcinoma (12%), and lung squamous carcinoma (8%). Tumor samples exhibiting a CD274 CN specimen ploidy of +4 (six copies) showcased PD-L1 amplification in 121% of cases, equivalent to 738 out of 60,793. The following focality category breakdown was observed: less than 0.1 mB (n=18, 24%); 0.1 mB to less than 4 mB (n=230, 311%); 4 mB to less than 20 mB (n=310, 42%); and 20 mB or greater (n=180, 244%). Lower PD-L1 amplification levels, specifically those below the specimen's ploidy plus four, manifested more frequently as non-focal amplifications compared to the higher level amplifications.