Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 297
Filter
1.
Sci Rep ; 14(1): 22324, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333597

ABSTRACT

Multimodal therapy for peritoneal metastasis (PM) including cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) provides long-term survival in highly selected colorectal cancer patients. Mechanisms behind HIPEC are unknown and may include induction of adaptive immunity. We therefore analyzed human PM samples and explored the impact of HIPEC in experimental models. Human samples from colorectal primary tumors (n = 19) and PM lesions (n = 37) were examined for the presence of CD8 + T-cells and their association with disease free (DFS) and overall survival (OS). CD8 + T cell response after HIPEC was assessed using an in-vivo PM mouse model, tumor cell lines and patient-derived tumor organoids. Patients with high intraepithelial CD8 + T cell counts showed longer DFS and OS. In the mouse model, HIPEC controlled growth of PM and increased numbers of functional granzyme positive CD8 + T cells within tumors. Cell lines and human organoids that were treated with heated chemotherapies showed immunogenic changes, reflected by significantly higher levels of MHC-class I molecules and expression of Cancer Testis Antigens Cyclin A1 and SSX-4. Using in-vitro co-culture assays, we noticed that cancer cells treated with heated chemotherapy primed dendritic cells, which subsequently enhanced effector functions of CD8 + T cells. The presence of CD8 + T-cells within PM lesions is associated with prolonged survival of patients with PM. Data from PM mouse model and in-vitro assay show that heated chemotherapies induce immunogenic changes on cancer cells leading to induction of CD8 + T-cells mediated immunity, which seems to control growth of PM lesions in mice after HIPEC.


Subject(s)
CD8-Positive T-Lymphocytes , Colorectal Neoplasms , Hyperthermic Intraperitoneal Chemotherapy , Peritoneal Neoplasms , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/immunology , Humans , Animals , Hyperthermic Intraperitoneal Chemotherapy/methods , Mice , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Cell Line, Tumor , Female , Male
2.
Front Immunol ; 15: 1403008, 2024.
Article in English | MEDLINE | ID: mdl-39192986

ABSTRACT

Background: As one of the most common cancer, colorectal cancer (CRC) is with high morbidity and mortality. Peritoneal metastasis (PM) is a fatal state of CRC, and few patients may benefit from traditional therapies. There is a complex interaction between PM and immune cell infiltration. Therefore, we aimed to determine biomarkers associated with colorectal cancer peritoneal metastasis (CRCPM) and their relationship with immune cell infiltration. Methods: By informatic analysis, differently expressed genes (DEGs) were selected and hub genes were screened out. RAB13, one of the hub genes, was identificated from public databases and validated in CRC tissues. The ESTIMATE, CEBERSORT and TIMER algorithms were applied to analyze the correlation between RAB13 and immune infiltration in CRC. RAB13's expression in different cells were analyzed at the single-cell level in scRNA-Seq. The Gene Set Enrichment Analysis (GSEA) was performed for RAB13 enrichment and further confirmed. Using oncoPredict algorithm, RAB13's impact on drug sensitivity was evaluated. Results: High RAB13 expression was identified in public databases and led to a poor prognosis. RAB13 was found to be positively correlated with the macrophages and other immune cells infiltration and from scRNA-Seq, RAB13 was found to be located in CRC cells and macrophages. GSEA revealed that high RAB13 expression enriched in a various of biological signaling, and oncoPredict algorithm showed that RAB13 expression was correlated with paclitaxel sensitivity. Conclusion: Our study indicated clinical role of RAB13 in CRC-PM, suggesting its potential as a therapeutic target in the future.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms , Peritoneal Neoplasms , rab GTP-Binding Proteins , Humans , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Prognosis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Female , Male , Gene Expression Profiling
3.
Cancer Res Commun ; 4(8): 2133-2146, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38994676

ABSTRACT

Immunotherapies have demonstrated limited clinical efficacy in malignant mesothelioma treatment. We conducted multiplex immunofluorescence analyses on tissue microarrays (n = 3) from patients with malignant pleural mesothelioma (MPM, n = 88) and malignant peritoneal mesothelioma (MPeM, n = 25). Our study aimed to elucidate spatial distributions of key immune cell populations and their association with lymphocyte activation gene 3 (LAG3), BRCA1-associated protein 1 (BAP1), neurofibromatosis type 2 (NF2), and methylthioadenosine phosphorylase (MTAP), with MTAP serving as a cyclin-dependent kinase inhibitor 2A/2B (CDKN2A/B) surrogate marker. Additionally, we examined the relationship between the spatial distribution of major immune cell types and prognosis and clinical characteristics of patients with malignant mesothelioma. We observed a higher degree of interaction between immune cells and tumor cells in MPM compared with MPeM. Notably, within MPM tumors, we detected a significantly increased interaction between tumor cells and CD8+ T cells in tumors with low BAP1 expression compared with those with high BAP1 expression. To support the broader research community, we have developed The Human Spatial Atlas of Malignant Mesothelioma, containing hematoxylin and eosin and multiplex immunofluorescence images with corresponding metadata. SIGNIFICANCE: Considering the limited therapeutic options available to patients with malignant mesothelioma, there is substantial translational potential in understanding the correlation between the spatial architecture of the malignant mesothelioma tumor immune microenvironment and tumor biology. Our investigation reveals critical cell-cell interactions that may influence the immune response against malignant mesothelioma tumors, potentially contributing to the differential behaviors observed in MPM and MPeM. These findings represent a valuable resource for the malignant mesothelioma cancer research community.


Subject(s)
Mesothelioma, Malignant , Mesothelioma , Peritoneal Neoplasms , Pleural Neoplasms , Purine-Nucleoside Phosphorylase , Tumor Microenvironment , Ubiquitin Thiolesterase , Humans , Tumor Microenvironment/immunology , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/pathology , Pleural Neoplasms/immunology , Pleural Neoplasms/pathology , Male , Female , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/pathology , Purine-Nucleoside Phosphorylase/metabolism , Purine-Nucleoside Phosphorylase/genetics , Middle Aged , Ubiquitin Thiolesterase/metabolism , Mesothelioma/immunology , Mesothelioma/pathology , Aged , Tumor Suppressor Proteins/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Neurofibromin 2/genetics , Biomarkers, Tumor , Prognosis , Antigens, CD , CD8-Positive T-Lymphocytes/immunology
4.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38955417

ABSTRACT

BACKGROUND: Tertiary lymphoid structures (TLSs) are thought to stimulate antitumor immunity and positively impact prognosis and response to immune checkpoint blockade. In gastric cancers (GCs), however, TLSs are predominantly found in GC with poor prognosis and limited treatment response. We, therefore, hypothesize that immune cell composition and function of TLS depends on tumor location and the tumor immune environment. METHODS: Spatial transcriptomics and immunohistochemistry were used to characterize the phenotype of CD45+ immune cells inside and outside of TLS using archival resection specimens from GC primary tumors and peritoneal metastases. RESULTS: We identified significant intrapatient and interpatient diversity of the cellular composition and maturation status of TLS in GC. Tumor location (primary vs metastatic site) accounted for the majority of differences in TLS maturity, as TLS in peritoneal metastases were predominantly immature. This was associated with higher levels of tumor-infiltrating macrophages and Tregs and less plasma cells compared with tumors with mature TLS. Furthermore, mature TLSs were characterized by overexpression of antitumor immune pathways such as B cell-related pathways, MHC class II antigen presentation while immature TLS were associated with protumor pathways, including T cell exhaustion and enhancement of DNA repair pathways in the corresponding cancer. CONCLUSION: The observation that GC-derived peritoneal metastases often contain immature TLS which are associated with immune suppressive regulatory tumor-infiltrating leucocytes, is in keeping with the lack of response to immune checkpoint blockade and the poor prognostic features of peritoneal metastatic GC, which needs to be taken into account when optimizing immunomodulatory strategies for metastatic GC.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Tertiary Lymphoid Structures , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/immunology , Tertiary Lymphoid Structures/immunology , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/immunology , Male , Female , Tumor Microenvironment
5.
Cancer Biol Med ; 21(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026438

ABSTRACT

Pseudomyxoma peritonei (PMP) is an indolent malignant syndrome. The standard treatment for PMP is cytoreductive surgery combined with intraperitoneal hyperthermic chemotherapy (CRS + HIPEC). However, the high recurrence rate and latent clinical symptoms and signs are major obstacles to further improving clinical outcomes. Moreover, patients in advanced stages receive little benefit from CRS + HIPEC due to widespread intraperitoneal metastases. Another challenge in PMP treatment involves the progressive sclerosis of PMP cell-secreted mucus, which is often increased due to activating mutations in the gene coding for guanine nucleotide-binding protein alpha subunit (GNAS). Consequently, the development of other PMP therapies is urgently needed. Several immune-related therapies have shown promise, including the use of bacterium-derived non-specific immunogenic agents, radio-immunotherapeutic agents, and tumor cell-derived neoantigens, but a well-recognized immunotherapy has not been established. In this review the roles of GNAS mutations in the promotion of mucin secretion and disease development are discussed. In addition, the immunologic features of the PMP microenvironment and immune-associated treatments are discussed to summarize the current understanding of key features of the disease and to facilitate the development of immunotherapies.


Subject(s)
Immunotherapy , Peritoneal Neoplasms , Pseudomyxoma Peritonei , Tumor Microenvironment , Humans , Pseudomyxoma Peritonei/therapy , Pseudomyxoma Peritonei/immunology , Pseudomyxoma Peritonei/genetics , Pseudomyxoma Peritonei/pathology , Tumor Microenvironment/immunology , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/genetics , Immunotherapy/methods , Cytoreduction Surgical Procedures , Mutation , Hyperthermic Intraperitoneal Chemotherapy
7.
Front Immunol ; 15: 1415457, 2024.
Article in English | MEDLINE | ID: mdl-39044825

ABSTRACT

Background: The occurrence of peritoneal metastasis (PM) in patients with colorectal cancer (CRC) has a dismal prognosis. There is often limited response to systemic- and immunotherapy, even in microsatellite unstable (MSI) CRC. To overcome therapy resistance, it is critical to understand local immune environment in the peritoneal cavity, and to develop models to study anti-tumor immune responses. Here, we defined the peritoneal immune system (PerIS) in PM-CRC patients and evaluate the pre-clinical potential of a humanized immune system (HIS) mouse model for PM-CRC. Methods: We studied the human PerIS in PM-CRC patients (n=20; MSS 19/20; 95%) and in healthy controls (n=3). HIS mice (NODscid gamma background; n=18) were generated, followed by intraperitoneal injection of either saline (HIS control; n=3) or human MSS/MSI CRC cell lines HUTU80, MDST8 and HCT116 (HIS-PM, n=15). Immune cells in peritoneal fluid and peritoneal tumors were analyzed using cytometry by time of flight (CyTOF). Results: The human and HIS mouse homeostatic PerIS was equally populated by NK cells and CD4+- and CD8+ T cells, however differences were observed in macrophage and B cell abundance. In HIS mice, successful peritoneal engraftment of both MSI and MSS tumors was observed (15/15; 100%). Both in human PM-CRC and in the HIS mouse PM-CRC model, we observed that MSS PM-CRC triggered a CD4+ Treg response in the PerIS, while MSI PM-CRC drives CD8+ TEMs responses. Conclusion: In conclusion, T cell responses in PM-CRC in HIS mice mirror those in human PM-CRC, making this model suitable to study antitumor T cell responses in PM-CRC.


Subject(s)
Colorectal Neoplasms , Disease Models, Animal , Peritoneal Neoplasms , Animals , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/immunology , Humans , Mice , Male , Female , Cell Line, Tumor , Mice, Inbred NOD , Mice, SCID , Middle Aged , Aged , Tumor Microenvironment/immunology , Killer Cells, Natural/immunology
8.
J Control Release ; 372: 362-371, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909698

ABSTRACT

Peritoneal carcinomatosis (PC) is characterized by a high recurrence rate and mortality following cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC), primarily due to incomplete cancer elimination. To enhance the standard of care for PC, we developed two cationic liposomal formulations aimed at localizing a toll-like receptor agonist, resiquimod (R848), in the peritoneal cavity to activate the immune system locally to specifically eradicate residual tumor cells. These formulations effectively extended R848 retention in the peritoneum by >10-fold, resulting in up to a 2-fold increase in interferon α (IFN-α) induction in the peritoneal fluid, without increasing the plasma levels. In a CT26 colon cancer model with peritoneal metastases, these liposomal R848 formulations, when combined with oxaliplatin (OXA)-an agent used in HIPEC that induces immunogenic cell death-increased tumor infiltration of effector immune cells, including DCs, CD4, and CD8 T cells. This led to the complete elimination of PC in 60-70% of the mice, while the control mice reached humane endpoints by 30 days. The cured mice developed specific antitumor immunity, as re-challenging them with the same tumor cells did not result in tumor establishment. However, inoculation with a different tumor line led to tumor development. Additionally, exposing CT26 tumor antigens to the splenocytes isolated from the cured mice induced the expansion of CD4 and CD8 T cells and the release of IFN-γ, demonstrating long-term immune memory to the specific tumor. The anti-tumor efficacy of these liposomal R848 formulations was mediated via CD8 T cells with different levels of involvement of CD4 and B cells, and the combination with an anti-PD-1 antibody achieved a cure rate of 90%.


Subject(s)
Imidazoles , Liposomes , Mice, Inbred BALB C , Oxaliplatin , Peritoneal Neoplasms , Animals , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/immunology , Imidazoles/administration & dosage , Cell Line, Tumor , Female , Oxaliplatin/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cations , Mice , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/drug therapy
9.
Gastric Cancer ; 27(5): 971-985, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38805119

ABSTRACT

BACKGROUND: Cancer immunotherapy aims to unleash the immune system's potential against cancer cells, providing sustained relief for tumors responsive to immune checkpoint inhibitors (ICIs). While promising in gastric cancer (GC) trials, the efficacy of ICIs diminishes in the context of peritoneal dissemination. Our objective is to identify strategies to enhance the impact of ICI treatment specifically for cases involving peritoneal dissemination in GC. METHODS: The therapeutic efficacy of anti-PD1, CTLA4 treatment alone, or in combination was assessed using the YTN16 peritoneal dissemination tumor model. Peritoneum and peritoneal exudate cells were collected for subsequent analysis. Immunohistochemical staining, flow cytometry, and bulk RNA-sequence analyses were conducted to evaluate the tumor microenvironment (TME). A Janus kinase inhibitor (JAKi) was introduced based on the pathway analysis results. RESULTS: Anti-PD1 and anti-CTLA4 combination treatment (dual ICI treatment) demonstrated therapeutic efficacy in certain mice, primarily mediated by CD8 + T cells. However, in mice resistant to dual ICI treatment, even with CD8 + T cell infiltration, most of the T cells exhibited an exhaustion phenotype. Notably, resistant tumors displayed abnormal activation of the Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) pathway compared to the untreated group, with observed infiltration of macrophages, neutrophils, and Tregs in the TME. The concurrent administration of JAKi rescued CD8 + T cells function and reshaped the immunosuppressive TME, resulting in enhanced efficacy of the dual ICI treatment. CONCLUSION: Dual ICI treatment exerts its anti-tumor effects by increasing tumor-specific CD8 + T cell infiltration, and the addition of JAKi further improves ICI resistance by reshaping the immunosuppressive TME.


Subject(s)
Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors , Janus Kinase Inhibitors , Mice, Inbred C57BL , Peritoneal Neoplasms , Stomach Neoplasms , Tumor Microenvironment , Animals , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/immunology , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Tumor Microenvironment/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CTLA-4 Antigen/antagonists & inhibitors , Humans , Female , Programmed Cell Death 1 Receptor/antagonists & inhibitors
10.
Int J Surg ; 110(6): 3504-3517, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38502852

ABSTRACT

BACKGROUND: Patients with peritoneal metastasis (PM) from gastric cancer (GC) exhibit poor prognosis. Chemoimmunotherapy offers promising clinical benefits; however, its efficacy and predictive biomarkers in a conversion therapy setting remain unclear. The authors aimed to retrospectively evaluate chemoimmunotherapy efficacy in a conversion therapy setting for GC patients with PM and establish a prediction model for assessing clinical benefits. MATERIALS AND METHODS: A retrospective evaluation of clinical outcomes encompassed 55 GC patients with PM who underwent chemoimmunotherapy in a conversion therapy setting. Baseline PM specimens were collected for genomic and transcriptomic profiling. Clinicopathological factors, gene signatures, and tumor immune microenvironment were evaluated to identify predictive markers and develop a prediction model. RESULTS: Chemoimmunotherapy achieved a 41.8% objective response rate and 72.4% R0 resection rate in GC patients with PM. Patients with conversion surgery showed better overall survival (OS) than those without the surgery (median OS: not reached vs 7.82 m, P <0.0001). Responders to chemoimmunotherapy showed higher ERBB2 and ERBB3 mutation frequencies, CTLA4 and HLA-DQB1 expression, and CD8+ T cell infiltration, but lower CDH1 mutation and naïve CD4+ T cell infiltration, compared to nonresponders. A prediction model was established integrating CDH1 and ERBB3 mutations, HLA-DQB1 expression, and naïve CD4+ T cell infiltration (AUC=0.918), which were further tested using an independent external cohort (AUC=0.785). CONCLUSION: This exploratory study comprehensively evaluated clinicopathological, genomic, and immune features and developed a novel prediction model, providing a rational basis for the selection of GC patients with PM for chemoimmunotherapy-involved conversion therapy.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Tumor Microenvironment , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/therapy , Retrospective Studies , Male , Female , Tumor Microenvironment/immunology , Middle Aged , Aged , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/drug therapy , Immunotherapy/methods , Adult , Treatment Outcome , Genomics
11.
BMC Cancer ; 22(1): 28, 2022 Jan 03.
Article in English | MEDLINE | ID: mdl-34980026

ABSTRACT

BACKGROUND: PHI-101 is an orally available, selective checkpoint kinase 2 (Chk2) inhibitor. PHI-101 has shown anti-tumour activity in ovarian cancer cell lines and impaired DNA repair pathways in preclinical experiments. Furthermore, the in vivo study suggests the synergistic effect of PHI-101 through combination with PARP inhibitors for ovarian cancer treatment. The primary objective of this study is to evaluate the safety and tolerability of PHI-101 in platinum-resistant recurrent ovarian cancer. METHODS: Chk2 inhibitor for Recurrent EpitheliAl periToneal, fallopIan, or oVarian cancEr (CREATIVE) trial is a prospective, multi-centre, phase IA dose-escalation study. Six cohorts of dose levels are planned, and six to 36 patients are expected to be enrolled in this trial. Major inclusion criteria include ≥ 19 years with histologically confirmed epithelial ovarian cancer, fallopian tube carcinoma, or primary peritoneal cancer. Also, patients who showed disease progression during platinum-based chemotherapy or disease progression within 24 weeks from completion of platinum-based chemotherapy will be included, and prior chemotherapy lines of more than five will be excluded. The primary endpoint of this study is to determine the dose-limiting toxicity (DLT) and maximum tolerated dose (MTD) of PHI-101. DISCUSSION: PHI-101 is the first orally available Chk2 inhibitor, expected to show effectiveness in treating recurrent ovarian cancer. Through this CREATIVE trial, DLT and MTD of this new targeted therapy can be confirmed to find the recommended dose for the phase II clinical trial. This study may contribute to developing a new combination regimen for the treatment of ovarian cancer. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04678102 .


Subject(s)
Antineoplastic Agents, Immunological , Checkpoint Kinase 2 , Immune Checkpoint Inhibitors , Neoplasm Recurrence, Local , Ovarian Neoplasms , Adult , Female , Humans , Middle Aged , Antineoplastic Agents, Immunological/administration & dosage , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/immunology , Checkpoint Kinase 2/antagonists & inhibitors , Dose-Response Relationship, Drug , Fallopian Tube Neoplasms/drug therapy , Fallopian Tube Neoplasms/immunology , Immune Checkpoint Inhibitors/administration & dosage , Maximum Tolerated Dose , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/immunology , Prospective Studies
12.
Theranostics ; 12(2): 875-890, 2022.
Article in English | MEDLINE | ID: mdl-34976218

ABSTRACT

Background: 5-aza-2'-deoxycytidine (5Aza), a DNA methyltransferase (DNMT) inhibitor, could activate tumor adaptive immunity to inhibit tumor progression. However, the molecular mechanisms by which 5Aza regulates tumor immune microenvironment are still not fully understood. Methods: The role of 5Aza in immune microenvironment of peritoneal carcinomatosis (PC) of colorectal cancer (CRC) was investigated. The effects of 5Aza on macrophage activation were studied by flow cytometry, real-time PCR, Western blotting assays, and Drug Affinity Responsive Target Stability (DARTS). The effects of 5Aza on tumor immunity were validated in stromal macrophages and T cells from CRC patients. Results: 5Aza could stimulate the activation of macrophages toward an M1-like phenotype and subsequent activation of T cells in premetastatic fat tissues, and ultimately suppress CRC-PC in immune-competent mouse models. Mechanistically, 5Aza stimulated primary mouse macrophages toward to a M1-like phenotype characterized by the increase of p65 phosphorylation and IL-6 expression. Furthermore, we screened and identified ATP-binding cassette transporter A9 (ABC A9) as a binding target of 5Aza. 5Aza induced cholesterol accumulation, p65 phosphorylation and IL-6 expression in an ABC A9-dependent manner. Pharmacological inhibition of NF-κB, or genetic depletion of IL-6 abolished the antitumor effect of 5Aza in mice. In addition, the antitumor effect of 5Aza was synergistically potentiated by conventional chemotherapeutic drugs 5-Fu or OXP. Finally, we validated the reprogramming role of 5Aza in antitumor immunity in stromal macrophages and T cells from CRC patients. Conclusions: Taken together, our findings showed for the first time that 5Aza suppressed CRC-PC by regulating macrophage-dependent T cell activation in premetastatic microenvironment, meanwhile uncovered a DNA methylation-independent mechanism of 5Aza in regulating ABC A9-associated cholesterol metabolism and macrophage activation.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cholesterol/metabolism , Colorectal Neoplasms/immunology , Decitabine/pharmacology , Macrophages, Peritoneal/drug effects , Neoplasm Metastasis/immunology , Peritoneal Neoplasms/immunology , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Humans , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peritoneal Neoplasms/diet therapy , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
13.
Cell Rep ; 37(11): 110111, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34910922

ABSTRACT

Blockade of the inhibitory checkpoint SIRPα-CD47 promotes phagocytosis of cancer cells by macrophages and is a promising avenue in anti-cancer therapy. Productive phagocytosis is strictly predicated on co-engagement of pro-phagocytic receptors-namely, Fc receptors (FcRs), integrin CD11b, or SLAMF7-by their ligands on cancer cells. Here, we examine whether additional pro-phagocytic receptors could be harnessed to broaden the scope of phagocytosis. Inflammatory stimuli, including multiple cytokines and Toll-like receptor (TLR) ligands, augment phagocytosis efficiency and fully alleviate the requirement of FcRs, CD11b, and SLAMF7 for phagocytosis. These effects are mediated by the unconventional pro-phagocytic integrins CD11a and CD11c, which act with CD18 to initiate actin polarization, leading to phagocytosis. Some inflammatory stimuli enable phagocytosis even in the absence of SIRPα-CD47 blockade. Higher CD11c expression in macrophage-enriched tumors correlates with improved survival in clinical studies. Thus, inflammatory macrophages exploit unconventional pro-phagocytic integrins for improved phagocytosis and anti-tumor immunity.


Subject(s)
CD11a Antigen/metabolism , CD11c Antigen/metabolism , Inflammation/immunology , Macrophages/immunology , Peritoneal Neoplasms/prevention & control , Phagocytosis , Signaling Lymphocytic Activation Molecule Family/physiology , Animals , CD11a Antigen/genetics , CD11c Antigen/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology
14.
Immunotherapy ; 13(18): 1457-1463, 2021 12.
Article in English | MEDLINE | ID: mdl-34664999

ABSTRACT

Determining the most appropriate management strategy for patients with large tumor masses is a very challenging issue. Unconventional radiotherapy modalities, such as spatially fractionated radiation therapy (SFRT), are associated with dramatic responses. Recent studies have suggested that systemic immune activation may be triggered by SFRT delivery to primary tumor lesion. This report describes the case of a patient treated with a novel form of immune-sparing partially ablative irradiation (ISPART) for a bulky peritoneal metastasis from renal cell cancer, refractory to anti-PD-1 therapy (nivolumab) as third-line therapy after sequential therapy with sunitinib and cabozantinib. The observed response suggests that there may be a synergistic effect between ISPART and immunotherapy. This case report supports the inclusion of ISPART in patients presenting with bulky lesions treated with checkpoint inhibitors .


Lay abstract Managing large tumor masses is a very challenging issue. In recent years, radiotherapy methods have been linked with good responses, which may be due to the activation of immune mechanisms. We describe the case of a patient with a large tumor mass from renal cell cancer. The patient had already been treated with anti-PD-1 therapy, after treatment with sunitinib and cabozantinib, along with radiotherapy. The results suggest that radiotherapy together with immunotherapy is very effective in enhancing immune response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Renal Cell , Dose Fractionation, Radiation , Immunotherapy , Kidney Neoplasms , Peritoneal Neoplasms , Anilides/administration & dosage , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Female , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Middle Aged , Neoplasm Metastasis , Nivolumab/administration & dosage , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/therapy , Pyridines/administration & dosage , Sunitinib/administration & dosage
15.
Cancer Res ; 81(22): 5706-5719, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34561272

ABSTRACT

The tumor microenvironment evolves during malignant progression, with major changes in nonmalignant cells, cytokine networks, and the extracellular matrix (ECM). In this study, we aimed to understand how the ECM changes during neoplastic transformation of serous tubal intraepithelial carcinoma lesions (STIC) into high-grade serous ovarian cancers (HGSOC). Analysis of the mechanical properties of human fallopian tubes (FT) and ovaries revealed that normal FT and fimbria had a lower tissue modulus, a measure of stiffness, than normal or diseased ovaries. Proteomic analysis of the matrisome fraction between FT, fimbria, and ovaries showed significant differences in the ECM protein TGF beta induced (TGFBI, also known as ßig-h3). STIC lesions in the fimbria expressed high levels of TGFBI, which was predominantly produced by CD163-positive macrophages proximal to STIC epithelial cells. In vitro stimulation of macrophages with TGFß and IL4 induced secretion of TGFBI, whereas IFNγ/LPS downregulated macrophage TGFBI expression. Immortalized FT secretory epithelial cells carrying clinically relevant TP53 mutations stimulated macrophages to secrete TGFBI and upregulated integrin αvß3, a putative TGFBI receptor. Transcriptomic HGSOC datasets showed a significant correlation between TGFBI expression and alternatively activated macrophage signatures. Fibroblasts in HGSOC metastases expressed TGFBI and stimulated macrophage TGFBI production in vitro. Treatment of orthotopic mouse HGSOC tumors with an anti-TGFBI antibody reduced peritoneal tumor size, increased tumor monocytes, and activated ß3-expressing unconventional T cells. In conclusion, TGFBI may favor an immunosuppressive microenvironment in STICs that persists in advanced HGSOC. Furthermore, TGFBI may be an effector of the tumor-promoting actions of TGFß and a potential therapeutic target. SIGNIFICANCE: Analysis of ECM changes during neoplastic transformation reveals a role for TGFBI secreted by macrophages in immunosuppression in early ovarian cancer.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Extracellular Matrix/pathology , Macrophages/immunology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment , Animals , Apoptosis , Cell Proliferation , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/metabolism , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Female , Humans , Immunosuppressive Agents , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/metabolism , Prognosis , Transforming Growth Factor beta1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
J Cancer Res Clin Oncol ; 147(11): 3333-3341, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34476575

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the third most common malignancy worldwide, but the key driver to distant metastases is still unknown. This study aimed to elucidate the link between immunosurveillance and organotropism of metastases in CRC by evaluating different gene signatures and pathways. MATERIAL AND METHODS: CRC patients undergoing surgery at the Department of General, Visceral and Transplantation Surgery at the Ludwig-Maximilian University Hospital Munich (Munich, Germany) were screened and categorized into M0 (no distant metastases), HEP (liver metastases) and PER (peritoneal carcinomatosis) after a 5-year follow-up. Six patients of each group were randomly selected to conduct a NanoString analysis, which includes 770 genes. Subsequently, all genes were further analyzed by gene set enrichment analysis (GSEA) based on seven main cancer-associated databases. RESULTS: Comparing HEP vs. M0, the gene set associated with the Toll-like receptor (TLR) cascade defined by the Reactome database was significantly overrepresented in HEP. HSP90B1, MAPKAPK3, PPP2CB, PPP2R1A were identified as the core enrichment genes. The immunologic signature pathway GSE6875_TCONV_VS_FOXP3_KO_TREG_DN with FOXP3 as downstream target was significantly overexpressed in M0. RB1, TMEM 100, CFP, ZKSCAN5, DDX50 were the core enrichment genes. Comparing PER vs. M0 no significantly differentially expressed gene signatures were identified. CONCLUSION: Chronic inflammation might enhance local tumor growth. This is the first study identifying immune related gene sets differentially expressed between patients with either liver or peritoneal metastases. The present findings suggest that the formation of liver metastases might be associated with TLR-associated pathways. In M0, a high expression of FOXP3 + tumor infiltrating lymphocytes (TILs) seemed to prevent at least in part metastases. Thus, these correlative findings lay the cornerstone to further studies elucidating the underlying mechanisms of organotropism of metastases.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Aged , Aged, 80 and over , Colorectal Neoplasms/genetics , Female , Humans , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Neoplasm Staging , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/secondary
17.
PLoS One ; 16(7): e0255049, 2021.
Article in English | MEDLINE | ID: mdl-34293030

ABSTRACT

BACKGROUND: Tumor-infiltrating lymphocytes (TILs) have been reported to reflect the anti-tumor immune status. However, recent investigations have demonstrated that intratumoral fibrosis is important as a factor affecting the infiltration of TILs. This study investigated the organ specificities of TIL infiltration and intratumoral fibrosis in primary colorectal cancer and distant metastases, as well as the relationship between the distribution of TILs and intratumoral fibrosis. METHODS: Patients who underwent resection of primary tumors or distant metastases for colorectal cancer with distant metastases were enrolled. We evaluated the TIL infiltration by immunohistochemical staining with CD3&CD8 and intratumoral fibrosis by immunohistochemical staining with α-SMA positive cancer-associated fibroblasts and Masson's trichrome staining against collagen fibers. The "ImageJ" was used to evaluate fibrosis, and the density of TILs in the dense and sparse areas of fibrosis was calculated. The Immunoscore (IS) was obtained based on the density of CD3+/CD8+TILs in the tumor center and invasive margin of the primary tumor. RESULTS: The degree of CD3+/CD8+TIL infiltration in peritoneal metastases was significantly lower than that in liver and lung metastases. The area ratio of α-SMA positive cancer-associated fibroblasts and collagen fibers in peritoneal metastases was significantly higher than that of liver and lung metastases. Furthermore, the density of TILs in the high-fibrosis area was significantly lower than that in the low-fibrosis area. In the high-IS group of primary tumors, the degree of TIL infiltration in distant metastases was significantly higher than that in the low-IS group. CONCLUSION: The infiltration of T lymphocytes into tumors is prevented in peritoneal metastases of colorectal cancer due to the high intratumoral fibrosis, which may lead to treatment resistance and a poor prognosis.


Subject(s)
CD8-Positive T-Lymphocytes , Collagen/immunology , Colorectal Neoplasms , Lymphocytes, Tumor-Infiltrating , Neoplasm Proteins/immunology , Peritoneal Neoplasms , Adult , Aged , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Female , Fibrosis , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Metastasis , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Retrospective Studies
18.
Mol Cancer Ther ; 20(9): 1508-1520, 2021 09.
Article in English | MEDLINE | ID: mdl-34210826

ABSTRACT

Advanced peritoneal carcinomatosis including high-grade ovarian cancer has poor prognoses and a poor response rate to current checkpoint inhibitor immunotherapies; thus, there is an unmet need for effective therapeutics that would provide benefit to these patients. Here we present the preclinical development of SENTI-101, a cell preparation of bone marrow-derived mesenchymal stromal (also known as stem) cells (MSC), which are engineered to express two potent immune-modulatory cytokines, IL12 and IL21. Intraperitoneal administration of SENTI-101 results in selective tumor-homing and localized and sustained cytokine production in murine models of peritoneal cancer. SENTI-101 has extended half-life, reduced systemic distribution, and improved antitumor activity when compared with recombinant cytokines, suggesting that it is more effective and has lower risk of systemic immunotoxicities. Treatment of tumor-bearing immune-competent mice with a murine surrogate of SENTI-101 (mSENTI-101) results in a potent and localized immune response consistent with increased number and activation of antigen presenting cells, T cells and B cells, which leads to antitumor response and memory-induced long-term immunity. Consistent with this mechanism of action, co-administration of mSENTI-101 with checkpoint inhibitors leads to synergistic improvement in antitumor response. Collectively, these data warrant potential clinical development of SENTI-101 for patients with peritoneal carcinomatosis and high-grade ovarian cancer.Graphical abstract: SENTI-101 schematic and mechanism of actionSENTI-101 is a novel cell-based immunotherapeutic consisting of bone marrow-derived mesenchymal stromal cells (BM-MSC) engineered to express IL12 and IL21 intended for the treatment of peritoneal carcinomatosis including high-grade serous ovarian cancer. Upon intraperitoneal administration, SENTI-101 homes to peritoneal solid tumors and secretes IL12 and IL21 in a localized and sustained fashion. The expression of these two potent cytokines drives tumor infiltration and engagement of multiple components of the immune system: antigen-presenting cells, T cells, and B cells, resulting in durable antitumor immunity in preclinical models of cancer.


Subject(s)
Interleukin-12/metabolism , Interleukins/metabolism , Melanoma, Experimental/immunology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Neoplasms/immunology , Peritoneal Neoplasms/immunology , Animals , Apoptosis , Cell Proliferation , Female , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Nutrients ; 13(7)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201458

ABSTRACT

The effect of preoperative immunonutrition intake on postoperative major complications in patients following cytoreductive surgery (CRS) with or without hyperthermic intraperitoneal chemotherapy (HIPEC) was assessed. The accuracy of C-Reactive Protein (CRP) for detecting postoperative complications was also analyzed. Patients treated within a peritoneal carcinomatosis program in which a complete or optimal cytoreduction was achieved were retrospectively analyzed. They were divided into two groups based on whether preoperative immunonutrition (IMN) or not (non-IMN) were administered. Clinical and surgical variables and postoperative complications were gathered. Predictive values of major morbidity of CRP during the first 3 postoperative days (POD) were also evaluated. A total of 107 patients were included, 48 belonging to the IMN group and 59 to the non-IMN group. In multivariate analysis immunonutrition (OR 0.247; 95%CI 0.071-0.859; p = 0.028), and the number of visceral resections (OR 1.947; 95%CI 1.086-3.488; p = 0.025) emerged as independent factors associated with postoperative major morbidity. CRP values above 103 mg/L yielded a negative predictive value of 84%. Preoperative intake of immunonutrition was associated with a decrease of postoperative major morbidity and might be recommended to patients with peritoneal carcinomatosis following CRS. Measuring CRP levels during the 3 first postoperative days is useful to rule out major morbidity.


Subject(s)
Cytoreduction Surgical Procedures/adverse effects , Hyperthermic Intraperitoneal Chemotherapy/adverse effects , Nutritional Physiological Phenomena , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/secondary , Postoperative Care , Preoperative Care , Aged , Area Under Curve , C-Reactive Protein/metabolism , Female , Humans , Logistic Models , Male , Middle Aged , Morbidity , Peritoneal Neoplasms/blood , Peritoneal Neoplasms/surgery , Postoperative Complications/etiology , ROC Curve
20.
Front Immunol ; 12: 651033, 2021.
Article in English | MEDLINE | ID: mdl-34054812

ABSTRACT

Background: Gastric cancer (GC) still represents the third leading cause of cancer-related death worldwide. Peritoneal relapse (PR) is the most frequent metastasis occurring among patients with advanced gastric cancer. Increasingly more evidence have clarified the tumor immune microenvironment (TIME) may predict survival and have clinical significance in GC. However, tumor-transcriptomics based immune signatures derived from immune profiling have not been established for predicting the peritoneal recurrence of the advanced GC. Methods: In this study, we depict the immune landscape of GC by using transcriptome profiling and clinical characteristics retrieved from GSE62254 of Gene Expression Omnibus (GEO). Immune cell infiltration score was evaluated via single-sample gene set enrichment (ssGSEA) analysis algorithm. The least absolute shrinkage and selection operator (LASSO) Cox regression algorithm was used to select the valuable immune cells and construct the final model for the prediction of PR. The receiver operating characteristic (ROC) curve and the Kaplan-Meier curve were used to check the accuracy of PRIs. Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed to explore the molecular pathways associated with PRIs. Results: A peritoneal recurrence related immune score (PRIs) with 10 immune cells was constructed. Compared to the low-PRIs group, the high-PRIs group had a greater risk. The upregulation of the focal adhesion signaling was observed in the high-PRIs subtype by GSEA and KEGG. Multivariate analysis found that both in the internal training cohort and the internal validation cohort, PRIs was a stable and independent predictor for PR. A nomogram that integrated clinicopathological features and PRIs to predict peritoneal relapse was constructed. Subgroup analysis indicated that the PRIs could obviously distinguish peritoneal recurrence in different molecular subtypes, pathological stages and Lauren subtypes, in which PRIs of Epithelial-Mesenchymal Transitions (EMT) subtype, III-IV stage and diffuse subtype are higher respectively. Conclusion: Overall, we performed a comprehensive evaluation of the immune landscape of GC and constructed a predictive PR model based on the immune cell infiltration. The PRIs represents novel promising feature of predicting peritoneal recurrence of GC and sheds light on the improvement of the personalized management of GC patients after surgery.


Subject(s)
Carcinoma/epidemiology , Gene Expression Regulation, Neoplastic/immunology , Peritoneal Neoplasms/epidemiology , Stomach Neoplasms/pathology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , Carcinoma/genetics , Carcinoma/immunology , Carcinoma/secondary , Chemotherapy, Adjuvant , Female , Follow-Up Studies , Gastrectomy , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Nomograms , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/secondary , Progression-Free Survival , ROC Curve , Retrospective Studies , Risk Assessment/methods , Stomach/immunology , Stomach/pathology , Stomach/surgery , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/therapy , Tumor Microenvironment/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL