Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.500
Filtrer
1.
Cancer Immunol Immunother ; 73(10): 189, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093404

RÉSUMÉ

BACKGROUND: The interplay between regulatory T cells (Tregs) and neighboring cells, which is pivotal for anti-tumor immunity and closely linked to patient prognosis, remains to be fully elucidated. METHODS: Tissue microarrays of 261 operable NSCLC patients were stained by multiplex immunofluorescence (mIF) assay, and the interaction between Tregs and neighboring cells in the tumor microenvironment (TME) was evaluated. Employing various machine learning algorithms, we developed a spatial immune signature to predict the prognosis of NSCLC patients. Additionally, we explored the interplay between programmed death-1/programmed death ligand-1 (PD-1/PD-L1) interactions and their relationship with Tregs. RESULTS: Survival analysis indicated that the interplay between Tregs and neighboring cells in the invasive margin (IM) and tumor center was associated with recurrence in NSCLC patients. We integrated the intersection of the three algorithms to identify four crucial spatial immune features [P(CD8+Treg to CK) in IM, P(CD8+Treg to CD4) in IM, N(CD4+Treg to CK) in IM, N(CD4+Tcon to CK) in IM] and employed these characteristics to establish SIS, an independent prognosticator of recurrence in NSCLC patients [HR = 2.34, 95% CI (1.53, 3.58), P < 0.001]. Furthermore, analysis of cell interactions demonstrated that a higher number of Tregs contributed to higher PD-L1+ cells surrounded by PD-1+ cells (P < 0.001) with shorter distances (P = 0.004). CONCLUSION: We dissected the cell interplay network within the TME, uncovering the spatial architecture and intricate interactions between Tregs and neighboring cells, along with their impact on the prognosis of NSCLC patients.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Récidive tumorale locale , Lymphocytes T régulateurs , Microenvironnement tumoral , Humains , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/mortalité , Lymphocytes T régulateurs/immunologie , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Microenvironnement tumoral/immunologie , Récidive tumorale locale/immunologie , Mâle , Femelle , Pronostic , Adulte d'âge moyen , Récepteur-1 de mort cellulaire programmée/métabolisme , Antigène CD274/métabolisme , Sujet âgé , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme
2.
BMC Cancer ; 24(1): 984, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39123197

RÉSUMÉ

PURPOSE: Chimeric antigen receptor T-cell (CART) therapy has shown clinical efficacy in refractory and relapsed large B-cell lymphomas, but is associated with serious acute and long-term toxicities. To understand the patient perspective, we measured a patient-reported outcome (PRO), specifically, health-related quality of life (HRQoL), at multiple time points over one year. METHODS: This was a prospective feasibility study of a cohort of patients who were eligible for standard of care CART therapy, tisagenlecleucel. Demographic data and disease characteristics were collected. HRQoL was measured using FACT-Lym at baseline, and months 1, 3, 6 and 12. FACT-Lym includes FACT-G (physical, social, emotional and functional well-being domains), plus a lymphoma subscale. RESULTS: Thirty-four of 35 patients approached, consented to participate. Two of them did not receive their infusion due to progressive disease. 50% were female and median age was 62 (23-77). Twenty-nine patients (91%) completed baseline FACT-Lym and 20 of 21 (95%) eligible patients completed 12-month FACT-Lym. 52% completed all 4 post-baseline FACT-Lym measures. Exploratory analyses for changes in FACT-Lym scores are reported. CONCLUSION: It is feasible to measure longitudinal PROs in patients who receive CART therapy. This study will inform future studies in evaluating the patient perspective on CART therapy.


Sujet(s)
Études de faisabilité , Immunothérapie adoptive , Lymphome B diffus à grandes cellules , Mesures des résultats rapportés par les patients , Qualité de vie , Humains , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Adulte , Lymphome B diffus à grandes cellules/thérapie , Lymphome B diffus à grandes cellules/immunologie , Lymphome B diffus à grandes cellules/traitement médicamenteux , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Études prospectives , Jeune adulte , Récepteurs chimériques pour l'antigène/usage thérapeutique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs aux antigènes des cellules T/usage thérapeutique , Récepteurs aux antigènes des cellules T/immunologie , Études longitudinales , Récidive tumorale locale/immunologie , Résultat thérapeutique
3.
Cells ; 13(15)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39120312

RÉSUMÉ

Cellular senescence has been increasingly recognized as a hallmark of cancer, reflecting its association with aging and inflammation, its role as a response to deregulated proliferation and oncogenic stress, and its induction by cancer therapies. While therapy-induced senescence (TIS) has been linked to resistance, recurrence, metastasis, and normal tissue toxicity, TIS also has the potential to enhance therapy response and stimulate anti-tumor immunity. In this review, we examine the Jekyll and Hyde nature of senescent cells (SnCs), focusing on how their persistence while expressing the senescence-associated secretory phenotype (SASP) modulates the tumor microenvironment through autocrine and paracrine mechanisms. Through the SASP, SnCs can mediate both resistance and response to cancer therapies. To fulfill the unmet potential of cancer immunotherapy, we consider how SnCs may influence tumor inflammation and serve as an antigen source to potentiate anti-tumor immune response. This new perspective suggests treatment approaches based on TIS to enhance immune checkpoint blockade. Finally, we describe strategies for mitigating the detrimental effects of senescence, such as modulating the SASP or targeting SnC persistence, which may enhance the overall benefits of cancer treatment.


Sujet(s)
Vieillissement de la cellule , Résistance aux médicaments antinéoplasiques , Tumeurs , Humains , Tumeurs/anatomopathologie , Tumeurs/immunologie , Microenvironnement tumoral/immunologie , Phénotype sécrétoire associé à la sénescence , Animaux , Immunothérapie/méthodes , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie
5.
Cancer Rep (Hoboken) ; 7(6): e2099, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38837676

RÉSUMÉ

BACKGROUND: An elevated neutrophil-to-lymphocyte ratio (NLR) in peripheral blood is an independent prognostic indicator of various cancers. AIMS: In this study, we aimed to investigate the prognostic relevance of the intratumoral immune cell balance in gastric cancer. METHODS AND RESULTS: The study included 82 patients who underwent curative resection for gastric cancer. The intratumoral cluster of differentiation (CD) 15- and CD8-positive cells were evaluated using immunohistochemical staining. Additionally, clinicopathological factors and prognoses were analyzed. Patients with high intratumoral CD15/CD8 ratios had significantly lower overall survival (OS) and relapse-free survival (RFS) compared to those with low CD15/CD8 ratios (p = .0026 and p < .0001, respectively). Additionally, a high CD15/CD8 ratio was associated with lymph node metastasis (p = .019). Patients with high NLR had a significantly lower RFS than those with low NLR (p = .0050). Multivariate analysis revealed that the intratumoral CD15/CD8 ratio, NLR, and venous invasion were independent prognostic indicators of RFS (CD15/CD8 ratio: p < .001, hazard ratio (HR) = 14.7, 95% confidence interval (CI) = 3.8-56.8; NLR: p = .010, HR = 5.4, 95% CI = 1.5-19.6; venous invasion: p = .005, HR = 7.4, 95% CI = 1.8-29.7). CONCLUSION: In summary, we found that the intratumoral CD15/CD8 ratio is an independent prognostic factor following gastric cancer resection and its increase is associated with lymph node metastasis and microscopic lymph vessel invasion. Immunological evaluation with additional aspects of innate immunity may be useful in predicting cancer prognosis.


Sujet(s)
Lymphocytes T CD8+ , Récidive tumorale locale , Granulocytes neutrophiles , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/chirurgie , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/mortalité , Tumeurs de l'estomac/anatomopathologie , Mâle , Femelle , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/anatomopathologie , Lymphocytes T CD8+/immunologie , Adulte d'âge moyen , Sujet âgé , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie , Pronostic , Antigènes CD15/analyse , Antigènes CD15/métabolisme , Adulte , Sujet âgé de 80 ans ou plus , Gastrectomie , Métastase lymphatique/anatomopathologie , Lymphocytes TIL/immunologie , Études rétrospectives , Survie sans rechute
6.
Nat Commun ; 15(1): 4241, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38762500

RÉSUMÉ

Glioblastoma multiforme (GBM) is a highly aggressive brain tumor characterized by invasive behavior and a compromised immune response, presenting treatment challenges. Surgical debulking of GBM fails to address its highly infiltrative nature, leaving neoplastic satellites in an environment characterized by impaired immune surveillance, ultimately paving the way for tumor recurrence. Tracking and eradicating residual GBM cells by boosting antitumor immunity is critical for preventing postoperative relapse, but effective immunotherapeutic strategies remain elusive. Here, we report a cavity-injectable bacterium-hydrogel superstructure that targets GBM satellites around the cavity, triggers GBM pyroptosis, and initiates innate and adaptive immune responses, which prevent postoperative GBM relapse in male mice. The immunostimulatory Salmonella delivery vehicles (SDVs) engineered from attenuated Salmonella typhimurium (VNP20009) seek and attack GBM cells. Salmonella lysis-inducing nanocapsules (SLINs), designed to trigger autolysis, are tethered to the SDVs, eliciting antitumor immune response through the intracellular release of bacterial components. Furthermore, SDVs and SLINs administration via intracavitary injection of the ATP-responsive hydrogel can recruit phagocytes and promote antigen presentation, initiating an adaptive immune response. Therefore, our work offers a local bacteriotherapy for stimulating anti-GBM immunity, with potential applicability for patients facing malignancies at a high risk of recurrence.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Récidive tumorale locale , Salmonella typhimurium , Glioblastome/thérapie , Glioblastome/immunologie , Animaux , Souris , Salmonella typhimurium/immunologie , Mâle , Récidive tumorale locale/prévention et contrôle , Récidive tumorale locale/immunologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Humains , Lignée cellulaire tumorale , Souris de lignée C57BL , Pyroptose , Immunité acquise , Immunité innée , Hydrogels/composition chimique , Immunothérapie/méthodes
7.
World J Gastroenterol ; 30(19): 2496-2501, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38817664

RÉSUMÉ

Immune checkpoint inhibitor therapy has dramatically improved patient prognosis, and thereby transformed the treatment in various cancer types including esophageal squamous cell carcinoma (ESCC) in the past decade. Monoclonal antibodies that selectively inhibit programmed cell death-1 (PD-1) activity has now become standard of care in the treatment of ESCC in metastatic settings, and has a high expectation to provide clinical benefit during perioperative period. Further, anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) monoclonal antibody has also been approved in the treatment of recurrent/metastatic ESCC in combination with anti-PD-1 antibody. Well understanding of the existing evidence of immune-based treatments for ESCC, as well as recent clinical trials on various combinations with chemotherapy for different clinical settings including neoadjuvant, adjuvant, and metastatic diseases, may provide future prospects of ESCC treatment for better patient outcomes.


Sujet(s)
Antigène CTLA-4 , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Traitement néoadjuvant , Humains , Tumeurs de l'oesophage/thérapie , Tumeurs de l'oesophage/immunologie , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/traitement médicamenteux , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Carcinome épidermoïde de l'oesophage/thérapie , Carcinome épidermoïde de l'oesophage/immunologie , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Traitement néoadjuvant/méthodes , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Immunothérapie/méthodes , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Résultat thérapeutique , Traitement médicamenteux adjuvant/méthodes , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Pronostic , Récidive tumorale locale/prévention et contrôle , Récidive tumorale locale/immunologie
8.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38731992

RÉSUMÉ

Non-muscle-invasive papillary urothelial carcinoma (NMIPUC) of the urinary bladder is the most common type of bladder cancer. Intravesical Bacille Calmette-Guerin (BCG) immunotherapy is applied in patients with a high risk of recurrence and progression of NMIPUC to muscle-invasive disease. However, the tumor relapses in about 30% of patients despite the treatment, raising the need for better risk stratification. We explored the potential of spatial distributions of immune cell subtypes (CD20, CD11c, CD163, ICOS, and CD8) within the tumor microenvironment to predict NMIPUC recurrence following BCG immunotherapy. Based on analyses of digital whole-slide images, we assessed the densities of the immune cells in the epithelial-stromal interface zone compartments and their distribution, represented by an epithelial-stromal interface density ratio (IDR). While the densities of any cell type did not predict recurrence, a higher IDR of CD11c (HR: 0.0012, p-value = 0.0002), CD8 (HR: 0.0379, p-value = 0.005), and ICOS (HR: 0.0768, p-value = 0.0388) was associated with longer recurrence-free survival (RFS) based on the univariate Cox regression. The history of positive repeated TUR (re-TUR) (HR: 4.93, p-value = 0.0001) and T1 tumor stage (HR: 2.04, p-value = 0.0159) were associated with shorter RFS, while G3 tumor grade according to the 1973 WHO classification showed borderline significance (HR: 1.83, p-value = 0.0522). In a multivariate analysis, the two models with a concordance index exceeding 0.7 included the CD11c IDR in combination with either a history of positive re-TUR or tumor stage. We conclude that the CD11c IDR is the most informative predictor of NMIPUC recurrence after BCG immunotherapy. Our findings highlight the importance of assessment of the spatial distribution of immune cells in the tumor microenvironment.


Sujet(s)
Vaccin BCG , Immunothérapie , Macrophages , Récidive tumorale locale , Microenvironnement tumoral , Tumeurs de la vessie urinaire , Humains , Microenvironnement tumoral/immunologie , Tumeurs de la vessie urinaire/immunologie , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/thérapie , Mâle , Vaccin BCG/usage thérapeutique , Récidive tumorale locale/immunologie , Femelle , Immunothérapie/méthodes , Sujet âgé , Adulte d'âge moyen , Macrophages/immunologie , Macrophages/métabolisme , Carcinome papillaire/anatomopathologie , Carcinome papillaire/immunologie , Carcinome papillaire/thérapie , Sous-populations de lymphocytes/immunologie , Sous-populations de lymphocytes/métabolisme , Pronostic , Sujet âgé de 80 ans ou plus
9.
Mol Immunol ; 170: 156-169, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38692097

RÉSUMÉ

Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer.


Sujet(s)
Tumeurs du sein , Protéines de liaison à l'ADN , Facteurs de transcription , Animaux , Femelle , Humains , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Régulation de l'expression des gènes tumoraux , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Facteur-1 de régulation d'interféron/immunologie , Facteur-3 de régulation d'interféron/métabolisme , Facteur-3 de régulation d'interféron/génétique , Interférons/métabolisme , Interférons/immunologie , Interférons/génétique , Récidive tumorale locale/immunologie , ARN double brin/immunologie , Transduction du signal/immunologie , Facteur de transcription RelA/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/immunologie
10.
Leuk Res ; 142: 107519, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38761562

RÉSUMÉ

Although follicular lymphoma (FL) is traditionally classified as an indolent subtype of B cell non-Hodgkin lymphoma, clinical trajectories are often diverse based on unique disease biology, and many patients will eventually experience relapse of their disease. Furthermore, progression of disease within 24 months is associated with increased mortality rates for FL. In the last five years, we have witnessed an upsurge in the commercial availability of targeted therapies for relapsed/refractory (R/R) FL, including chimeric antigen receptor-T (CAR-T) products, bispecific T cell engagers (BiTEs), epigenetic modifier therapies, and next-generation Bruton tyrosine kinase (BTK) inhibitors. Furthermore, clinical trial options have increased tremendously and now include combinatorial strategies that exert synergy against malignant germinal center B cells. Here, we provide a 2024 update of novel therapeutic agents whose development has been informed by recent advances in the genetics and immunobiology of R/R FL. Specifically, we emphasize high-value targeted therapeutics, including anti-CD3 x anti-CD20 BiTEs and adoptive T cell therapies. We discuss prospects on selection and sequencing of BiTEs and CAR-T therapies for patients with R/R FL. We underscore the principles of FL pathobiology that are paving way for future drug discovery and shed insight into therapeutic targeting within nodal basins based on our increasing understanding of the FL microenvironment. Finally, we summarize how a greater knowledge of FL immunobiology can inform risk stratification and therapy selection on a personalized basis for R/R FL in 2025.


Sujet(s)
Lymphome folliculaire , Humains , Lymphome folliculaire/thérapie , Lymphome folliculaire/génétique , Lymphome folliculaire/immunologie , Lymphome folliculaire/anatomopathologie , Récidive tumorale locale/génétique , Récidive tumorale locale/thérapie , Récidive tumorale locale/immunologie , Récidive tumorale locale/anatomopathologie , Immunothérapie adoptive/méthodes , Résistance aux médicaments antinéoplasiques/génétique , , Thérapie moléculaire ciblée/méthodes
11.
Int Immunopharmacol ; 134: 112174, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38703571

RÉSUMÉ

CD19-directed chimeric antigen receptor (CAR) T cell therapy has been shown to achieve a considerably durable response in patients with refractory or relapsed B cell non-Hodgkin lymphomas. Most of these CARs were generated by lentivirus. With the exception of Yescarta and Tecartus, few patients with relapsed-/refractory- lymphoma have been treated clinically with a CARs using retroviral vector (RV). Here, we reported a relapsed/refractory grade 2 follicular lymphoma patient with multiple chemotherapy failures, and was treated with a novel CD19 CAR-T cell manufactured from a RV. After tumor burden was reduced with Obinutuzumab and Duvelisib, the patient was infused novel CD19 CAR-T cells at a dose of 3 × 106 cells/ kg. Then he experienced a rapid response and achieved almost complete remission by day 26. Only grade 2 CRS, bilateral submaxillary lymph node enlargement and cytomegalovirus (CMV) infection occurred without neurotoxicity, and the patient's condition improved after a series of symptomatic treatments. In addition, CAR copy number peaked at 532,350 copies/µg on day 15 and continued to expand for 5 months. This may be the first case report of RV preparation of novel CD19 CAR-T cells for direct treatment of recurrent follicular lymphoma. We will observe its long-term efficacy and conduct trials in more patients in the future.


Sujet(s)
Antigènes CD19 , Infections à cytomégalovirus , Immunothérapie adoptive , Lymphome folliculaire , Humains , Mâle , Adulte d'âge moyen , Antigènes CD19/immunologie , Infections à cytomégalovirus/immunologie , Infections à cytomégalovirus/thérapie , Immunothérapie adoptive/méthodes , Lymphome folliculaire/thérapie , Lymphome folliculaire/immunologie , Récidive tumorale locale/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Résultat thérapeutique
12.
Oncoimmunology ; 13(1): 2347441, 2024.
Article de Anglais | MEDLINE | ID: mdl-38694625

RÉSUMÉ

In clinical practice, the administration of adjuvant chemotherapy (ACT) following tumor surgical resection raises a critical dilemma for stage II colon cancer (CC) patients. The prognostic features used to identify high-risk CC patients rely on the pathological assessment of tumor cells. Currently, these factors are considered for stratifying patients who may benefit from ACT at early CC stages. However, the extent to which these factors predict clinical outcomes (i.e. recurrence, survival) remains highly controversial, also uncertainty persists regarding patients' response to treatment, necessitating further investigation. Therefore, an imperious need is to explore novel biomarkers that can reliably stratify patients at risk, to optimize adjuvant treatment decisions. Recently, we evaluated the prognostic and predictive value of Immunoscore (IS), an immune digital-pathology assay, in stage II CC patients. IS emerged as the sole significant parameter for predicting disease-free survival (DFS) in high-risk patients. Moreover, IS effectively stratified patients who would benefit most from ACT based on their risk of recurrence, thus predicting their outcomes. Notably, our findings revealed that digital IS outperformed the visual quantitative assessment of the immune response conducted by expert pathologists. The latest edition of the WHO classification for digestive tumor has introduced the evaluation of the immune response, as assessed by IS, as desirable and essential diagnostic criterion. This supports the revision of current cancer guidelines and strongly recommends the implementation of IS into clinical practice as a patient stratification tool, to guide CC treatment decisions. This approach may provide appropriate personalized therapeutic decisions that could critically impact early-stage CC patient care.


Sujet(s)
Tumeurs du côlon , Humains , Tumeurs du côlon/immunologie , Tumeurs du côlon/anatomopathologie , Appréciation des risques , Traitement médicamenteux adjuvant , Pronostic , Stadification tumorale , Marqueurs biologiques tumoraux/métabolisme , Récidive tumorale locale/immunologie , Survie sans rechute
13.
Clin Cancer Res ; 30(14): 2865-2871, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38713595

RÉSUMÉ

In February 2022, the FDA approved ciltacabtagene autoleucel, a chimeric antigen receptor (CAR) T-cell therapy targeting the B-cell maturation antigen, for adult patients with relapsed/refractory multiple myeloma after ≥4 lines of therapy, including an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 monoclonal antibody. Approval was based on overall response rate (ORR), complete response (CR) rate, and duration of response (DoR) in 97 adult patients in a single-arm, open-label, multicenter phase 2 trial (CARTITUDE-1 [NCT03548207]). Patients received a single infusion of ciltacabtagene autoleucel, preceded by lymphodepleting chemotherapy. Of the 97 patients evaluable, ORR was 97.9% [95% confidence interval (CI), 92.7-99.7] with a stringent CR rate of 78.4% (95% CI, 68.8-86.1). After median follow-up of 18 months, the median DoR was 21.8 months (95% CI, 21.8-not estimable [NE]) in responders (PR or better) and NE (95% CI, 21.8 months-NE) in patients who achieved stringent CR. Serious adverse reactions occurred in 55% of the 97 patients evaluated for safety. Grade 3 or higher cytokine release syndrome (CRS) and neurologic toxicities occurred in 5% and 11% of the patients, respectively, leading to a Risk Evaluation and Mitigation Strategy. Neurologic toxicities included immune effector cell-associated neurologic syndrome, typically seen with CAR-T products, parkinsonism, peripheral neuropathy, cranial nerve palsies, and Guillain-Barré syndrome. One fatal case of hemophagocytic lymphohistiocytosis/macrophage activation syndrome occurred. Prolonged and recurrent grade 3 or 4 cytopenias occurred; a single patient required hematopoietic stem-cell rescue.


Sujet(s)
Immunothérapie adoptive , Myélome multiple , Humains , Myélome multiple/thérapie , Myélome multiple/traitement médicamenteux , Myélome multiple/immunologie , Mâle , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , États-Unis , Adulte d'âge moyen , Sujet âgé , Femelle , Adulte , Food and Drug Administration (USA) , Agrément de médicaments , Récidive tumorale locale/immunologie , Récidive tumorale locale/traitement médicamenteux , Antigène de maturation des cellules B/antagonistes et inhibiteurs , Antigène de maturation des cellules B/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Résultat thérapeutique
14.
Gynecol Oncol ; 187: 85-91, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38735144

RÉSUMÉ

BACKGROUND: The combination of immune checkpoint inhibitors (ICIs) and platinum-based chemotherapy has emerged as a highly promising primary option for advanced or recurrent endometrial cancer (EC). The study aimed to evaluate treatment efficacy of ICIs with cytotoxic chemotherapy in EC. METHODS: We conducted a comprehensive review of randomized controlled trials up to November 11, 2023, focusing on immunotherapy combined with chemotherapy versus chemotherapy alone for EC. The primary endpoint was the pooled hazard ratio (HR), which was further analyzed across subgroups based on mismatch repair (MMR) status, race, histology, and programmed death-ligand 1 (PD-L1) status. The protocol was registered in PROSPERO (CRD42023475669). FINDINGS: Four trials with 2335 patients were analyzed. ICIs with chemotherapy significantly prolonged progression-free survival (PFS) (HR, 0.70; 95% CI, 0.62-0.79) and overall survival (OS) (HR, 0.75; 95% CI, 0.63-0.89) compared to chemotherapy alone. Stratification by MMR status showed substantial benefits for dMMR (PFS; HR, 0.33; 95% CI, 0.26-0.43; OS; HR, 0.37; 95% CI, 0.22-0.91) over pMMR cohorts in both PFS and OS. In the subgroup analysis, there was significant PFS advantage in Caucasian (HR, 0.63; 95% CI, 0.54-0.72) over non-Caucasian, in endometrioid histology (HR, 0.66; 95% CI, 0.56-0.78) over non-endometrioid, and in PD-L1 positive (HR, 0.39; 95% CI, 0.19-0.81) over PD-L1 negative population. INTERPRETATION: ICIs combined with platinum-based chemotherapy significantly prolonged PFS and OS in patients with advanced or recurrent EC. Patients with dMMR status, Caucasians, endometrioid histology, and positive PD-L1 status showed significant PFS benefits, emphasizing the need for personalized treatment approaches to improve outcomes.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs de l'endomètre , Inhibiteurs de points de contrôle immunitaires , Récidive tumorale locale , Humains , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/immunologie , Essais contrôlés randomisés comme sujet , Survie sans progression
15.
Cell Rep Med ; 5(6): 101571, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38776914

RÉSUMÉ

Iberdomide is a next-generation cereblon (CRBN)-modulating agent in the clinical development in multiple myeloma (MM). The analysis of biomarker samples from relapsed/refractory patients enrolled in CC-220-MM-001 (ClinicalTrials.gov: NCT02773030), a phase 1/2 study, shows that iberdomide treatment induces significant target substrate degradation in tumors, including in immunomodulatory agent (IMiD)-refractory patients or those with low CRBN levels. Additionally, some patients with CRBN genetic dysregulation who responded to iberdomide have a similar median progression-free survival (PFS) (10.9 months) and duration of response (DOR) (9.5 months) to those without CRBN dysregulation (11.2 month PFS, 9.4 month DOR). Iberdomide treatment promotes a cyclical pattern of immune stimulation without causing exhaustion, inducing a functional shift in T cells toward an activated/effector memory phenotype, including in triple-class refractory patients and those receiving IMiDs as a last line of therapy. This analysis demonstrates that iberdomide's clinical mechanisms of action are driven by both its cell-autonomous effects overcoming CRBN dysregulation in MM cells, and potent immune stimulation that augments anti-tumor immunity.


Sujet(s)
Myélome multiple , Thalidomide , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Myélome multiple/génétique , Thalidomide/usage thérapeutique , Thalidomide/pharmacologie , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Femelle , Mâle , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/immunologie , Récidive tumorale locale/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Récidive , Adulte d'âge moyen , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Sujet âgé
16.
Cell Rep Med ; 5(6): 101584, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38776911

RÉSUMÉ

Iberdomide is a potent cereblon E3 ligase modulator (CELMoD agent) with promising efficacy and safety as a monotherapy or in combination with other therapies in patients with relapsed/refractory multiple myeloma (RRMM). Using a custom mass cytometry panel designed for large-scale immunophenotyping of the bone marrow tumor microenvironment (TME), we demonstrate significant increases of effector T and natural killer (NK) cells in a cohort of 93 patients with multiple myeloma (MM) treated with iberdomide, correlating findings to disease characteristics, prior therapy, and a peripheral blood immune phenotype. Notably, changes are dose dependent, associated with objective response, and independent of prior refractoriness to MM therapies. This suggests that iberdomide broadly induces innate and adaptive immune activation in the TME, contributing to its antitumor efficacy. Our approach establishes a strategy to study treatment-induced changes in the TME of patients with MM and, more broadly, patients with cancer and establishes rational combination strategies for iberdomide with immune-enhancing therapies to treat MM.


Sujet(s)
Moelle osseuse , Immunité innée , Myélome multiple , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Immunité innée/effets des médicaments et des substances chimiques , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/anatomopathologie , Moelle osseuse/immunologie , Immunité acquise/effets des médicaments et des substances chimiques , Femelle , Mâle , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Sujet âgé , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/immunologie , Récidive tumorale locale/traitement médicamenteux
17.
Nat Commun ; 15(1): 4194, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38760364

RÉSUMÉ

The role of tumor-resident intracellular microbiota (TRIM) in carcinogenesis has sparked enormous interest. Nevertheless, the impact of TRIM-targeted antibacteria on tumor inhibition and immune regulation in the tumor microenvironment (TME) remains unexplored. Herein, we report long-term relapse-free survival by coordinating antibacteria with antitumor treatment, addressing the aggravated immunosuppression and tumor overgrowth induced by TRIM using breast and prostate cancer models. Combining Ag+ release with a Fenton-like reaction and photothermal conversion, simultaneous bacteria killing and multimodal antitumor therapy are enabled by a single agent. Free of immune-stimulating drugs, the agent restores antitumor immune surveillance and activates immunological responses. Secondary inoculation and distal tumor analysis confirm lasting immunological memory and systemic immune responses. A relapse-free survival of >700 days is achieved. This work unravels the crucial role of TRIM-targeted antibacteria in tumor inhibition and unlocks an unconventional route for immune regulation in TME and a complete cure for cancer.


Sujet(s)
Microenvironnement tumoral , Femelle , Mâle , Humains , Animaux , Souris , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tumeurs du sein/immunologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/thérapie , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/thérapie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Microbiote/effets des médicaments et des substances chimiques , Argent/composition chimique , Survie sans rechute , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Récidive tumorale locale/immunologie
18.
Horm Metab Res ; 56(6): 424-428, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38621693

RÉSUMÉ

Papillary thyroid carcinoma (PTC) is characterized by T cell infiltration and frequently by the presence of anti-thyroglobulin antibodies (TgAbs). The role of cellular immunity and of TbAbs in this context is a matter of debate. The aim of our study was to correlate the presence of TgAbs, tumor epitope-specific T cells and the clinical outcome of PTC patients. We studied n=183 consecutive patients with a diagnosis of PTC which were treated with total thyroidectomy plus 131I ablation. During a follow-up of in mean 97 months, most of the PTC patients had no signs of tumor relapse (n=157 patients). In contrast, one patient had serum Tg levels above the detection limit and<1 ng/ml, two patients Tg serum levels≥1 ng/ml and<2 ng/ml and n=23 patients had Tg serum levels≥2 ng/ml. Morphological signs of tumor recurrence were seen in 14 patients; all of these patients had serum Tg levels≥2 ng/ml. Importantly, with the exception of one patient, all TgAb positive PTC patients (n=27) had no signs of tumor recurrence as the serum Tg levels were below the assays functional sensitivities. Tetramer analyses revealed a higher number of tumor epitope-specific CD8+T cells in TgAb positive patients compared to TgAb negative PTC patients. In summary, we show that the occurrence of TgAbs may have an impact on the clinical outcome in PTC patients. This might be due to a tumor epitope-specific cellular immunity in PTC patients.


Sujet(s)
Autoanticorps , Immunité cellulaire , Thyroglobuline , Cancer papillaire de la thyroïde , Tumeurs de la thyroïde , Humains , Mâle , Femelle , Adulte d'âge moyen , Tumeurs de la thyroïde/immunologie , Tumeurs de la thyroïde/sang , Tumeurs de la thyroïde/anatomopathologie , Cancer papillaire de la thyroïde/immunologie , Cancer papillaire de la thyroïde/sang , Cancer papillaire de la thyroïde/anatomopathologie , Thyroglobuline/immunologie , Thyroglobuline/sang , Adulte , Sujet âgé , Autoanticorps/sang , Autoanticorps/immunologie , Épitopes/immunologie , Carcinome papillaire/immunologie , Carcinome papillaire/anatomopathologie , Carcinome papillaire/sang , Jeune adulte , Adolescent , Récidive tumorale locale/immunologie , Récidive tumorale locale/sang
19.
Cancer Genomics Proteomics ; 21(3): 316-326, 2024.
Article de Anglais | MEDLINE | ID: mdl-38670590

RÉSUMÉ

BACKGROUND/AIM: Patients with triple-negative breast cancer (TNBC) have a high rate of recurrence within 3 years of diagnosis and a high rate of death within 5 years compared to other subtypes. The number of clinical trials investigating various new agents and combination therapies has recently increased; however, current strategies benefit only a minority of patients. This study aimed to identify specific genes that predict patients at high risk of recurrence and the immune status of the tumor microenvironment at an early stage, thereby providing insight into potential therapeutic targets to improve clinical outcomes in TNBC patients. MATERIALS AND METHODS: We evaluated the prognostic significance of microarray mRNA expression of 20,603 genes in 233 TNBC patients from the METABRIC dataset and further validated the results using RNA-seq mRNA expression data in 143 TNBC patients from the GSE96058 dataset. RESULTS: Eighteen differentially expressed genes (AKNA, ARHGAP30, CA9, CD3D, CD3G, CD6, CXCR6, CYSLTR1, DOCK10, ENO1, FLT3LG, IFNG, IL2RB, LPXN, PRKCB, PVRIG, RASSF5, and STAT4) identified in both datasets were found to be reliable biomarkers for predicting TNBC recurrence and progression. Notably, the genes whose low expression was associated with increased risk of recurrence and death were immune-related genes, with significant differences in levels of immune cell infiltration in the tumor microenvironment between high- and low- expression groups. CONCLUSION: Genes reported herein may be effective biomarkers to identify TNBC patients who will and will not benefit from immunotherapy and may be particularly important genes for developing future treatment strategies, including immunotherapy.


Sujet(s)
Récidive tumorale locale , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/anatomopathologie , Femelle , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/immunologie , Pronostic , Marqueurs biologiques tumoraux/génétique , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Transcriptome , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Adulte d'âge moyen
20.
Eur J Cancer ; 203: 114063, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38615592

RÉSUMÉ

BACKGROUND AND AIM: Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer with highly variable clinical behavior, but risk stratification is still challenging. We sought to identify immune-related gene expression signatures of pure DCIS associated with different risks of breast cancer recurrence. METHODS: A retrospective nested case-control study of 143 pure DCIS was performed including 70 women with subsequent ipsilateral breast event (IBE, in situ or invasive; cases) and 73 DCIS women with no IBE and matched for age, tumor size, treatment, hormone receptors/HER2 status, and follow-up time (controls). RNA was extracted from DCIS samples and subjected to next-generation sequencing gene expression analysis of 395 immune-related genes. Correlations between DCIS immune-related gene expression and IBE were analyzed using weighted Cox regression for nested case-control data. RESULTS: Eight immune-related genes were differentially expressed between cases and controls. MAGEA10 expression (present vs. absent) and high expression levels of IFNA17 and CBLB (Q4 vs. Q1) were observed more frequently in DCIS of women with subsequent IBE, mainly invasive (p-valueFDR < 0.05). Conversely, expression of IL3RA1, TAGAP, TNFAIP8, and high expression levels of CCL2 and LRP1 were associated with a lower risk of IBE (p-valueFDR < 0.05). CONCLUSION: This exploratory analysis of pure DCIS showed significant differences in immune-related gene expression profiles between women with and with no subsequent IBE, particularly as invasive IBE. These results, after additional validation, could improve risk stratification and management of DCIS patients.


Sujet(s)
Tumeurs du sein , Carcinome intracanalaire non infiltrant , Récidive tumorale locale , Humains , Femelle , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Carcinome intracanalaire non infiltrant/génétique , Carcinome intracanalaire non infiltrant/immunologie , Carcinome intracanalaire non infiltrant/anatomopathologie , Adulte d'âge moyen , Récidive tumorale locale/génétique , Récidive tumorale locale/immunologie , Études cas-témoins , Études rétrospectives , Sujet âgé , Adulte , Marqueurs biologiques tumoraux/génétique , Régulation de l'expression des gènes tumoraux , Analyse de profil d'expression de gènes , Transcriptome
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE