Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Mol Cancer Res ; 21(12): 1356-1365, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37707375

ABSTRACT

Stage IA gastric adenocarcinoma, characterized by foci of intramucosal signet ring cells (SRC), is found in nearly all asymptomatic patients with germline pathogenic CDH1 variants and hereditary diffuse gastric cancer syndrome (HDGC). The molecular steps involved in initiating malignant transformation and promoting SRC dormancy in HDGC are unknown. Here, whole-exome bulk RNA sequencing (RNA-seq) of SRCs and adjacent non-SRC epithelium (NEP) was performed on laser-capture microdissected (LCM) regions of interest found in risk-reducing total gastrectomy specimens from patients with HDGC (Clinicaltrials.gov ID: NCT03030404). In total, 20 patients (6 male, 14 female) with confirmed HDGC were identified. Analysis of differentially expressed genes (DEG) demonstrated upregulation of certain individual EMT and proliferation genes. However, no oncogenic pathways were found to be upregulated in SRCs. Rather, SRC regions had significant enrichment in pathways involved in T-cell signaling. CIBERSORTx predicted significant increases in the presence of regulatory T cells (Treg) specific to SRC regions. IHC confirmed an increase in FOXP3+ cells in SRC foci, as well as elevations in CD4+ T cells and HLA-DR staining. In summary, the tumor immune microenvironment is microscopically inseparable from stage IA gastric SRCs using a granular isolation technique. An elevation in CD4+ T cells within SRC regions correlates with clinically observed SRC dormancy, while Treg upregulation represents a potential immune escape mechanism. IMPLICATIONS: Characterization of the tumor-immune microenvironment in HDGC underscores the potential for the immune system to shape the transcriptional profile of the earliest tumors, which suggests immune-directed therapy as a potential cancer interception strategy in diffuse-type gastric cancer.


Subject(s)
Adenocarcinoma , Carcinoma, Signet Ring Cell , Stomach Neoplasms , Humans , Male , Female , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Genetic Predisposition to Disease , Gastrectomy , Germ-Line Mutation , Carcinogenesis/genetics , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Carcinoma, Signet Ring Cell/surgery , Cadherins/genetics , Tumor Microenvironment , Antigens, CD
2.
HPB (Oxford) ; 25(11): 1420-1428, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37573232

ABSTRACT

BACKGROUND: The incidence of intrahepatic cholangiocarcinoma (ICC) continues to rise, and hepatectomy is the only cure. Perioperative outcomes following hepatectomy for colorectal liver metastases (CRLM) and hepatocellular carcinoma (HCC) are better described than for ICC. The aim was to compare post-hepatectomy outcomes for ICC to CRLM and HCC. METHODS: The 2014-2020 ACS NSQIP hepatectomy PUF was utilized. Patients with ICC, CRLM, and HCC were identified and others excluded. Demographic, disease, and procedural characteristics were collected. Univariable and multivariable analyses (Chi-Square for categorical variables; Kruskal-Wallis for continuous variables) were performed for mortality, serious morbidity, bile leak, post-hepatectomy liver failure (PHLF), and 30-day readmission. RESULTS: 17,789 patients underwent hepatectomy including 2377 for ICC, 10,195 for CRLM, and 5217 for HCC. Patients undergoing hepatectomy for ICC vs. HCC vs. CRLM were noted to have higher 30-day mortality (4.8% vs. 2.5% vs. 1.0%, respectively p < 0.05). ICC was associated with higher overall and serious morbidity, bile leak, severe PHLF, and readmission. Multivariable analyses confirmed higher odds ratios for mortality and morbidity (p < 0.05) in patients with ICC. CONCLUSION: Hepatectomy for ICC is associated with worse short-term outcomes than for CRLM or HCC. Surgeons should be aware of these risks during surgical planning.

3.
J Surg Educ ; 80(11): 1529-1535, 2023 11.
Article in English | MEDLINE | ID: mdl-37479647

ABSTRACT

PURPOSE: Professional coaching has been utilized in the military and private sector with a long track record of optimizing efficiency, improving high-functioning team performance, and creating greater satisfaction among the workforce. Recent studies in physician populations have suggested that coaching may protect healthcare providers from burnout and improve quality of life and resilience. The aims of the current study were to describe our single-institution experience with the introduction of a leadership coaching program among surgical residents and to characterize the nature of the common reasons for referral for coaching. METHODS: Upon identification by program faculty that a resident would benefit from coaching, an email was sent from the program director to the coach to establish contact between the coach and resident, with a brief synopsis of the factors that lead to the resident being referred for coaching. The study team obtained deidentified, simplified synopses of the initial email correspondences from program leadership reaching out to residents to refer them to coaching. Common recurring themes were quantified. Later, coaches reviewed their notes kept during coaching sessions and, in a similar fashion, identified topics discussed and skills developed in coaching sessions for each resident. Topics were summarized for each resident, and a deidentified list of residents and the themes reviewed in coaching were provided to the study team, who quantified these topics. Baseline demographic information on the resident cohort, including training level, gender, and number of repeat referrals were summarized to delineate differences in patterns of repeat referral and attrition. RESULTS: This study was conducted within the general surgery residency program at a single academic medical center, composed of 43 categorical and 8 preliminary residents. Over a 2.5-year period, 21 residents were referred: 5 chief, 8 senior, 5 midlevel, and 3 junior (1 preliminary) residents. Male residents represented 2/3 and female residents 1/3 of the total number of referrals. There were 3 repeat referrals, 2 male and 1 female. We identified 2 overarching reasons for which residents were referred for coaching: request for structured leadership training and request for communication training. Six themes were identified upon review of referrals for coaching. Among these were the need for improved communication, methods to improve team integration, tools to balance professional and personal responsibilities, and practices to improve confidence and assertiveness. Through the coaching relationship, residents reviewed deficits and received an individualized plan to address newly identified problem areas. Upon initiation of coaching, new themes like anxiety management, emotional intelligence, and cultural acclimatization were identified. During each coaching session, residents participate in exercises designed to build habits of effective listening, communication, and conflict resolution. CONCLUSIONS: Coaching in surgical residency provides a structured program for residents to develop skills in planning and orchestrating team operations, listening and communicating effectively, mitigating conflict, and managing professional and personal responsibilities. Follow-up studies will focus on the long-term effects of professional coaching, evaluating survey data from self-assessments and professional evaluations.


Subject(s)
Internship and Residency , Mentoring , Humans , Male , Female , Mentoring/methods , Leadership , Quality of Life , Health Personnel
4.
Am Surg ; 89(5): 1989-1996, 2023 May.
Article in English | MEDLINE | ID: mdl-34974741

ABSTRACT

Traumatic duodenal injuries are rare and often challenging to diagnose and treat. Management of these injuries remains controversial and continues to evolve. Here, we performed a review of the literature and guidelines for the diagnosis and management of traumatic duodenal injuries.A common recommendation in more recent literature is primary, tension-free repair of duodenal injuries when possible if surgical repair is necessary. Conversely, if duodenal injuries are unamenable to primary repair, more complex procedures such as Roux-en-Y duodenojejunostomy or pancreaticoduodenectomy may be necessary. Regardless of injury grade or type of surgical repair, the literature continues to support wide extraluminal drainage. Over time, the management of complex duodenal injuries has evolved to favor simple primary repair whenever possible. According to recent studies, more complex procedures are associated with higher rates of post-operative complications and should be reserved for severe injuries when primary repair is not possible.


Subject(s)
Abdominal Injuries , Wounds, Penetrating , Humans , Retrospective Studies , Duodenum/surgery , Duodenum/injuries , Pancreaticoduodenectomy , Wounds, Penetrating/surgery , Anastomosis, Surgical/methods , Abdominal Injuries/diagnosis , Abdominal Injuries/surgery
5.
Cancer Cell ; 40(9): 986-998.e5, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36055226

ABSTRACT

Platelets, the often-overlooked component of the immune system, have been shown to promote tumor growth. Non-alcoholic fatty liver disease (NAFLD) is a common disease in the Western world and rising risk for hepatocellular carcinoma (HCC). Unexpectedly, we observed that platelets can inhibit the growth of established HCC in NAFLD mice. Through pharmacological inhibition and genetic depletion of P2Y12 as well as in vivo transfusion of wild-type (WT) or CD40L-/- platelets, we demonstrate that the anti-tumor function of platelets is mediated through P2Y12-dependent CD40L release, which leads to CD8+ T cell activation by the CD40 receptor. Unlike P2Y12 inhibition, blocking platelets with aspirin does not prevent platelet CD40L release nor accelerate HCC in NAFLD mice. Similar findings were observed in liver metastasis models. All together, our study reveals a complex role of platelets in tumor regulation. Anti-platelet treatment without inhibiting CD40L release could be considered for liver cancer patients with NAFLD.


Subject(s)
Blood Platelets/immunology , Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Receptors, Purinergic P2Y12/metabolism , Animals , CD40 Ligand/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics
6.
J Hepatol ; 77(3): 748-760, 2022 09.
Article in English | MEDLINE | ID: mdl-35378172

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic steatohepatitis (NASH) represents the fastest growing underlying cause of hepatocellular carcinoma (HCC) and has been shown to impact immune effector cell function. The standard of care for the treatment of advanced HCC is immune checkpoint inhibitor (ICI) therapy, yet NASH may negatively affect the efficacy of ICI therapy in HCC. The immunologic mechanisms underlying the impact of NASH on ICI therapy remain unclear. METHODS: Herein, using multiple murine NASH models, we analysed the influence of NASH on the CD8+ T-cell-dependent anti-PD-1 responses against liver cancer. We characterised CD8+ T cells' transcriptomic, functional, and motility changes in mice receiving a normal diet (ND) or a NASH diet. RESULTS: NASH blunted the effect of anti-PD-1 therapy against liver cancers in multiple murine models. NASH caused a proinflammatory phenotypic change of hepatic CD8+ T cells. Transcriptomic analysis revealed changes related to NASH-dependent impairment of hepatic CD8+ T-cell metabolism. In vivo imaging analysis showed reduced motility of intratumoural CD8+ T cells. Metformin treatment rescued the efficacy of anti-PD-1 therapy against liver tumours in NASH. CONCLUSIONS: We discovered that CD8+ T-cell metabolism is critically altered in the context of NASH-related liver cancer, impacting the effectiveness of ICI therapy - a finding which has therapeutic implications in patients with NASH-related liver cancer. LAY SUMMARY: Non-alcoholic steatohepatitis represents the fastest growing cause of hepatocellular carcinoma. It is also associated with reduced efficacy of immunotherapy, which is the standard of care for advanced hepatocellular carcinoma. Herein, we show that non-alcoholic steatohepatitis is associated with impaired motility, metabolic function, and response to anti-PD-1 treatment in hepatic CD8+ T cells, which can be rescued by metformin treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Metformin , Non-alcoholic Fatty Liver Disease , Animals , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Liver/pathology , Liver Neoplasms/etiology , Metformin/pharmacology , Metformin/therapeutic use , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism
7.
iScience ; 25(2): 103847, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35198900

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) has become an important etiology leading to liver cancer. NAFLD alters adaptive T cell immunity and has a profound influence on liver cancer development. However, it is unclear how NAFLD affects tumor antigen-specific T cell response. In this study, we generated a doxycycline-inducible MHC-I and -II antigen-expressing HCC cell line which allowed us to investigate tumor antigen-specific T cell response in two NAFLD mouse models. The system proved to be an effective and efficient way to study tumor antigen-specific T cells. Using this model, it was found that NAFLD impairs antigen-specific CD8+ T cell immunity against HCC. The effect was not due to reduced generation or intrinsic functional changes of tumor antigen-specific CD8+ T cells but caused by accumulated macrophages in the liver environment. The findings suggest that targeting macrophages in NAFLD-driven HCC may improve therapeutic outcomes.

8.
Gut ; 71(6): 1161-1175, 2022 06.
Article in English | MEDLINE | ID: mdl-34340996

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) represents a typical inflammation-associated cancer. Tissue resident innate lymphoid cells (ILCs) have been suggested to control tumour surveillance. Here, we studied how the local cytokine milieu controls ILCs in HCC. DESIGN: We performed bulk RNA sequencing of HCC tissue as well as flow cytometry and single-cell RNA sequencing of enriched ILCs from non-tumour liver, margin and tumour core derived from 48 patients with HCC. Simultaneous measurement of protein and RNA expression at the single-cell level (AbSeq) identified precise signatures of ILC subgroups. In vitro culturing of ILCs was used to validate findings from in silico analysis. Analysis of RNA-sequencing data from large HCC cohorts allowed stratification and survival analysis based on transcriptomic signatures. RESULTS: RNA sequencing of tumour, non-tumour and margin identified tumour-dependent gradients, which were associated with poor survival and control of ILC plasticity. Single-cell RNA sequencing and flow cytometry of ILCs from HCC livers identified natural killer (NK)-like cells in the non-tumour tissue, losing their cytotoxic profile as they transitioned into tumour ILC1 and NK-like-ILC3 cells. Tumour ILC composition was mediated by cytokine gradients that directed ILC plasticity towards activated tumour ILC2s. This was liver-specific and not seen in ILCs from peripheral blood mononuclear cells. Patients with high ILC2/ILC1 ratio expressed interleukin-33 in the tumour that promoted ILC2 generation, which was associated with better survival. CONCLUSION: Our results suggest that the tumour cytokine milieu controls ILC composition and HCC outcome. Specific changes of cytokines modify ILC composition in the tumour by inducing plasticity and alter ILC function.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/metabolism , Cytokines/metabolism , Humans , Immunity, Innate , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear , Liver Neoplasms/metabolism , Lymphocytes , RNA/metabolism , Tumor Microenvironment
9.
Cancer Immunol Res ; 9(9): 1024-1034, 2021 09.
Article in English | MEDLINE | ID: mdl-34193462

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are MR1-restricted innate-like T cells that recognize non-peptide antigens including riboflavin derivates. Although in vitro-activated MAIT cells show antitumor activity, the in vivo role of MAIT cells in cancer is still unclear. Here, we have shown that MAIT cells have antitumor function in vivo when activated by a combination of the synthetic riboflavin synthesis pathway-derived antigen 5-OP-RU [5-(2-oxopropylideneamino)-6-D-ribitylaminouracil] and the Toll-like receptor 9 (TLR9) agonist CpG. Coadministration of 5-OP-RU and CpG induced strong systemic in vivo expansion and activation of MAIT cells with high CD69 expression, pronounced effector memory phenotype, and upregulated levels of effector molecules including IFNγ, granzyme B, and perforin. Activated and expanded MAITs induced a potent and broad antitumor immune response in murine models of liver metastasis and hepatocellular carcinoma, lung metastasis, and subcutaneous tumors in two different mouse strains. Such tumor inhibition was absent in MAIT-deficient Mr1 -/- mice. CRISPR/Cas9-mediated MR1 knockout in tumor cells did not affect efficacy of this MAIT-directed immunotherapy, pointing toward an indirect mechanism of action. Our findings suggest that MAIT cells are an attractive target for cancer immunotherapy.See related Spotlight by Lantz, p. 996.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Lymphocyte Activation/immunology , Minor Histocompatibility Antigens/metabolism , Mucosal-Associated Invariant T Cells/drug effects , Neoplasms/drug therapy , Animals , Antigens, CD , Antigens, Differentiation, T-Lymphocyte , CRISPR-Cas Systems , Cell Line, Tumor , Female , Histocompatibility Antigens Class I/genetics , Humans , Lectins, C-Type , Male , Mice , Minor Histocompatibility Antigens/genetics , Mucosal-Associated Invariant T Cells/metabolism , Neoplasms/metabolism , Ribitol/administration & dosage , Ribitol/analogs & derivatives , Riboflavin/biosynthesis , Riboflavin/chemistry , Riboflavin/pharmacology , Uracil/administration & dosage , Uracil/analogs & derivatives
10.
Mol Cancer Ther ; 20(8): 1469-1480, 2021 08.
Article in English | MEDLINE | ID: mdl-34088830

ABSTRACT

Approximately 20,000 patients per year are diagnosed with esophageal adenocarcinoma (EAC) and malignant pleural mesothelioma (MPM); fewer than 20% survive 5 years. Effective therapeutic strategies are limited although patients receive a combination of chemotherapeutics. These tumors harbor thousands of mutations that contribute to tumor development. Downstream of oncogenic driving mutations, altered tumor mitochondria promote resistance to apoptosis. Dynamic Bcl-2 homology-3 profiling (DBP) is a functional assay of live cells that identifies the mitochondrial proteins responsible for resistance to apoptosis. We hypothesized that DBP will predict which protein to target to overcome resistance thereby enhancing combinatorial therapy.DBP predicted that targeting either Mcl-1 or Bcl-xL increases the efficacy of the chemotherapeutic agent, cisplatin, whereas targeting Bcl-2 does not. We performed these assays by treating EAC and MPM cells with a combination of Bcl-2 homology-3 (BH3) mimetics and cisplatin. Following treatments, we performed efficacy assessments including apoptosis assays, IC50 calculations, and generation of a combinatorial index. DBP confirmed that targeting mitochondria with BH3 mimetics alters the threshold of apoptosis. These apoptotic effects were abolished when the mitochondrial pathway was disrupted. We validated our findings by developing knockdown models of antiapoptotic proteins Mcl-1, Bcl-xL, and the mitochondrial effector proteins Bax/Bak. Knockdown of Mcl-1 or Bcl-xL recapitulated the results of BH3 mimetics. In addition, we report an approach for BH3 profiling directly from patient tumor samples. We demonstrate that the DBP assay on living tumor cells measures the dynamic changes of resistance mechanisms, assesses response to combinatorial therapy, and provides results in a clinically feasible time frame.


Subject(s)
Biomarkers, Tumor/metabolism , Biomimetics/methods , Cisplatin/pharmacology , Esophageal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Mesothelioma, Malignant/drug therapy , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Drug Synergism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , Mesothelioma, Malignant/genetics , Mesothelioma, Malignant/metabolism , Mesothelioma, Malignant/pathology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Tumor Cells, Cultured , bcl-2-Associated X Protein/antagonists & inhibitors
11.
Cell Mol Gastroenterol Hepatol ; 12(3): 1166-1178, 2021.
Article in English | MEDLINE | ID: mdl-34033968

ABSTRACT

BACKGROUND & AIMS: Intrahepatic cholangiocarcinoma (iCCA) accounts for a fraction of primary liver cancers but has a 5-year survival rate of only 10%. Immune checkpoint inhibitors are effective in treating many solid cancers, but immune checkpoint inhibitor monotherapy has no clear benefit in iCCA. Mitogen-activated kinase (MEK) inhibitors, such as trametinib, have shown promising results in preclinical studies for iCCA by inhibiting cell proliferation and modifying the tumor microenvironment. This study aimed to show the potential benefit of combining trametinib with anti-programmed cell death protein 1 (PD-1) therapy in different iCCA mouse models. METHODS: Here, we assessed the in vitro cytotoxicity of trametinib in mouse (SB1 and LD-1) and human (EGI-1) cholangiocarcinoma cell lines. We examined the efficacy of single-agent trametinib, anti-PD-1, and a combination of both in subcutaneous, orthotopic, and plasmid-induced iCCA mouse models. Flow cytometry analysis was used to elucidate changes in the tumor immune microenvironment upon treatment. Whole-exome sequencing (WES) was performed on the SB1 tumor cell line to correlate this preclinical model with iCCAs in patients. RESULTS: Trametinib reduced tumor cell growth of SB1, LD-1, and EGI-1 tumor cells in vitro. Trametinib treatment led to up-regulation of major histocompatibility complex (MHC-I) and programmed cell death ligand 1 (PD-L-1) (programmed cell death ligand 1) on tumor cells in vitro. The combination of trametinib and anti-PD-1 reduced tumor burden in several iCCA tumor models and improved survival in SB1 tumor-bearing mice compared with either agent alone. Immunoprofiling of tumor-bearing mice showed an increase of hepatic effector memory CD8+ and CD4+ T cells, as well as an increased degranulation of CD8+ T cells, indicating enhanced cytotoxicity. WES and somatic mutational analysis showed no mutations of KRAS, BRAF, and ERK in SB1 tumor cells, and showed a similar genetic signature of SB1 found in a cohort of patients with iCCA. CONCLUSIONS: Altogether, our study shows that trametinib improves the immunogenicity of tumor cells by up-regulating MHC-I surface expression. The combination with anti-PD-1 results in optimal treatment efficacy for iCCA. WES of SB1 cells suggests that KRAS wild-type iCCAs also respond to this combination therapy.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Immune Checkpoint Inhibitors/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Animals , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/mortality , Drug Synergism , Female , High-Throughput Nucleotide Sequencing , Humans , Immune Checkpoint Inhibitors/pharmacology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidinones/pharmacology , Exome Sequencing , Xenograft Model Antitumor Assays
12.
J Thorac Dis ; 13(4): 2177-2183, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34012568

ABSTRACT

BACKGROUND: Malignant pleural effusions (MPEs) are common manifestations of metastatic cancers and are associated with a dismal prognosis. Talc pleurodesis has been proven to be effective in the management of MPEs, however, class-action lawsuits linking talc to ovarian adenocarcinoma have rendered it unavailable at many institutions. As a result, surgeons have resorted to less effective chemical pleurodesis as an alternative to indwelling pleural drainage catheters. Given the absence of talc, we explored the effectiveness of video-assisted thoracoscopic surgery (VATS) partial pleurectomy (VPP) for treating MPEs. METHODS: We performed a retrospective review of patients with MPEs managed after talc became unavailable at our institution. Between 2016 and 2018, we identified five patients who refused pleural drainage catheters and underwent VPP. Symptoms at presentation included fatigue, dyspnea, and pleuritic chest pain. All had unilateral MPEs (left n=3, right n=2). VPP included removal of parietal surfaces of the pleura other than the pleura overlying the subclavian vessels, the mediastinum, and the lung viscera. RESULTS: There were no significant perioperative adverse events and post-operative pain was well controlled. Chest tubes were removed between post-operative day (POD) 3 and 7. Follow-up time ranged from four to 36 weeks. All patients had symptomatic relief and radiographic evidence of improved MPEs. No patients required re-interventions. One patient expired six months after surgery while the remaining four were alive at last follow-up. CONCLUSIONS: VPP offers an effective alternative to chemical pleurodesis for managing MPEs in patients who prefer to avoid pleural drainage catheters.

13.
Cancer Genet ; 256-257: 21-25, 2021 08.
Article in English | MEDLINE | ID: mdl-33836455

ABSTRACT

BACKGROUND: Adrenocortical carcinoma (ACC) is a rare malignancy arising from the adrenal cortex. ACC carries a dismal prognosis and surgery offers the only chance for a cure. Germline pathogenic variants among certain oncogenes have been implicated in ACC. Here, we report the first case of ACC in a patient with a pathogenic variant in the Ataxia Telangiectasia Mutated (ATM) gene. PATIENTS AND METHODS: A 56-year-old Caucasian woman with biopsy proven ACC deemed unresectable and treated with etoposide, doxorubicin and cisplatin (EDP), and mitotane presented to our institution for evaluation. The tumor specimen was examined pathologically, and genetic analyses were performed on the tumor and germline using next-generation sequencing. RESULTS: Pathologic evaluation revealed an 18.0 × 14.0 × 9.0 cm low-grade ACC with tumor free resection margins. Immunohistochemistry stained for inhibin, melan-A, and chromogranin. ClinOmics analysis revealed a germline pathogenic deletion mutation of one nucleotide in ATM is denoted as c.1215delT at the cDNA level and p.Asn405LysfsX15 (N405KfsX15) at the protein level. Genomic analysis of the tumor showed loss of heterozygosity (LOH) of chromosome 11 on which the ATM resides. CONCLUSION: ACC is an aggressive malignancy for which surgical resection currently offers the only curative option. Here we report a heterozygous loss-of-function mutation in germline DNA and LOH of ATM in tumor in an ACC patient, a classic two-hit scenario in a well-known cancer suppresser gene, suggesting a pathogenic role of the ATM gene in certain ACC cases.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Germ-Line Mutation/genetics , Adrenal Cortex Neoplasms/diagnostic imaging , Adrenal Cortex Neoplasms/pathology , Female , Genomics , Humans , Middle Aged , Multimodal Imaging
14.
Nat Commun ; 12(1): 1455, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674593

ABSTRACT

T-cell exhaustion denotes a hypofunctional state of T lymphocytes commonly found in cancer, but how tumor cells drive T-cell exhaustion remains elusive. Here, we find T-cell exhaustion linked to overall survival in 675 hepatocellular carcinoma (HCC) patients with diverse ethnicities and etiologies. Integrative omics analyses uncover oncogenic reprograming of HCC methionine recycling with elevated 5-methylthioadenosine (MTA) and S-adenosylmethionine (SAM) to be tightly linked to T-cell exhaustion. SAM and MTA induce T-cell dysfunction in vitro. Moreover, CRISPR-Cas9-mediated deletion of MAT2A, a key SAM producing enzyme, results in an inhibition of T-cell dysfunction and HCC growth in mice. Thus, reprogramming of tumor methionine metabolism may be a viable therapeutic strategy to improve HCC immunity.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Methionine/metabolism , T-Lymphocytes/metabolism , Animals , Biomarkers, Tumor , CD8-Positive T-Lymphocytes , CRISPR-Cas Systems , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Methionine Adenosyltransferase/blood , Methionine Adenosyltransferase/genetics , Methionine Adenosyltransferase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , S-Adenosylmethionine/metabolism , Transcriptome
15.
iScience ; 24(1): 101990, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33490900

ABSTRACT

Hydroxychloroquine (HCQ) is a well-known anti-inflammatory drug but is also known as an anti-inflammatory drug. Here, we evaluate the influence of HCQ treatment on the effect of anti-PD1 tumor immunotherapy. Anti-PD1 therapy-sensitive tumor lines MC38, CT26, and RIL-175 were used to investigate the impact of HCQ on anti-PD1 therapy efficacy. In vitro assays demonstrated that HCQ directly inhibited tumor cell growth in all the tested tumor cell lines. HCQ treatment impaired both antigen-specific and nonspecific T-cell production of TNFα and IFNγ in vitro and in vivo. Importantly, in all the three tumor models, HCQ treatment significantly impaired the response to anti-PD1 treatment, accompanying diminished in vivo T-cell activation and reduced tumor-infiltrating, antigen-specific CD8+ T cells. This study shows that HCQ treatment can result in immunotherapy failure due to its immunosuppressive effects that offset both increased MHC-I expression by tumor cell and direct cytotoxicity.

16.
Cancer Discov ; 11(5): 1248-1267, 2021 05.
Article in English | MEDLINE | ID: mdl-33323397

ABSTRACT

Gut dysbiosis is commonly observed in patients with cirrhosis and chronic gastrointestinal disorders; however, its effect on antitumor immunity in the liver is largely unknown. Here we studied how the gut microbiome affects antitumor immunity in cholangiocarcinoma. Primary sclerosing cholangitis (PSC) or colitis, two known risk factors for cholangiocarcinoma which promote tumor development in mice, caused an accumulation of CXCR2+ polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC). A decrease in gut barrier function observed in mice with PSC and colitis allowed gut-derived bacteria and lipopolysaccharide to appear in the liver and induced CXCL1 expression in hepatocytes through a TLR4-dependent mechanism and an accumulation of CXCR2+ PMN-MDSCs. In contrast, neomycin treatment blocked CXCL1 expression and PMN-MDSC accumulation and inhibited tumor growth even in the absence of liver disease or colitis. Our study demonstrates that the gut microbiome controls hepatocytes to form an immunosuppressive environment by increasing PMN-MDSCs to promote liver cancer. SIGNIFICANCE: MDSCs have been shown to be induced by tumors and suppress antitumor immunity. Here we show that the gut microbiome can control accumulation of MDSCs in the liver in the context of a benign liver disease or colitis.See related commentary by Chagani and Kwong, p. 1014.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Cholangiocarcinoma/pathology , Gram-Negative Bacteria/physiology , Hepatocytes/physiology , Liver Neoplasms/pathology , Myeloid-Derived Suppressor Cells/physiology , Animals , Disease Models, Animal , Gastrointestinal Microbiome , Humans , Mice
17.
Am Surg ; 87(7): 1163-1170, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33345554

ABSTRACT

INTRODUCTION: Major hepatectomies are utilized to manage primary hepatic malignancies. Reports from high-volume centers (HVCs) with minimal perioperative mortality focus on multiple aspects of perioperative care, although patient-specific factors remain unelucidated. We identified patient factors associated with outcomes and examined whether these contribute to survival differences. METHODS: We queried the National Cancer Database (2006-2015) for patients with primary liver malignancies managed with major hepatectomy. Facilities were dichotomized by volume (high volume: >15 hepatectomies/year). Perioperative outcomes were compared based on patient demographic and clinical characteristics as well as center volume. RESULTS: 4263 patients were included with 78.5% receiving care in low-volume centers (LVCs). 90-day postoperative mortality was higher in LVCs vs. HVCs (12% vs. 7.5%; P < .001). Factors associated with undergoing surgery in LVCs included: living in areas with lower income (P = .006) and education (P < .001), having nonprivate insurance (P < .001), residing near the care center (P < .001), and having a comorbidity score (CDS) >1 (P = .014). Patients with CDS ≤ 1 had higher 90-day mortality in LVCs (11.3% vs. 6.6%; P < .001) and had similar outcomes in LVCs and HVCs (15.6% vs. 13.7% P = .6). Patients with CDS > 1 were more likely to receive care in LVCs (16.3% vs. 12.7%; P < .001). CONCLUSION: Reduced perioperative mortality following major hepatectomy in HVCs is driven by optimal management of patients with low CDS. However, nearly 1 in 5 patients who undergo major hepatectomies have a high CDS and approximately 15% of them succumb in the perioperative period irrespective of the treating centers' experience.


Subject(s)
Hepatectomy/adverse effects , Hepatectomy/mortality , Hospitals, High-Volume , Hospitals, Low-Volume , Liver Diseases/complications , Liver Diseases/surgery , Aged , Databases, Factual , Female , Hospital Mortality , Humans , Liver Diseases/mortality , Male , Middle Aged , Retrospective Studies , Socioeconomic Factors , Treatment Outcome , United States
18.
Ann Thorac Surg ; 112(6): 2070-2075, 2021 12.
Article in English | MEDLINE | ID: mdl-33378696

ABSTRACT

BACKGROUND: Thoracic surgery (TS) residency positions are in high demand. There is no study describing the nationwide attributes of successful matriculants in this specialty. We examined the characteristics of TS resident applicants and identified factors associated with acceptance. METHODS: Applicant data from 2014 to 2017 application cycles was extracted from the Electronic Residency Application System and stratified by matriculation status. Medical education, type of general surgery residency, and research achievements were analyzed. The number of peer-reviewed publications and the corresponding impact factor for the journals where they were published were quantified. RESULTS: There were 492 applicants and 358 matriculants. The overall population was primarily male (79.5%), white (55.1%), educated at United States allopathic medical schools (66.5%), and trained at university-based general surgery residencies (59.6%). Education at United States allopathic schools (odds ratio [OR], 2.54; P < .0001), being a member of the American Osteopathic Association (OR, 3.27; P = .021), general surgery residency affiliation with a TS residency (OR, 2.41; P = .0003) or National Cancer Institute designated Comprehensive Cancer Center (OR, 1.76; P = .0172), and being a first-time applicant (OR, 4.71, P < .0001) were independently associated with matriculation. Matriculants published a higher number of manuscripts than nonmatriculants (median of 3 vs 2, P < .0001) and more frequently published in higher impact journals (P < .0001). CONCLUSIONS: Our study includes objective and quantifiable data from recent application cycles and represents an in-depth examination of applicants to TS residency. The type of medical school and residency, as well as academic productivity, correlate with successful matriculation.


Subject(s)
Education, Medical, Graduate/methods , Internship and Residency/methods , Program Evaluation/methods , Schools, Medical , Surgeons/education , Thoracic Surgery/education , Thoracic Surgical Procedures/education , Adult , Female , Humans , Male , Retrospective Studies , United States
19.
J Gastrointest Surg ; 25(3): 609-622, 2021 03.
Article in English | MEDLINE | ID: mdl-32705611

ABSTRACT

BACKGROUND: Locally advanced gastric cancer (LAGC) presents a therapeutic dilemma, particularly as it often involves adjacent organs through desmoplasia or true pathologic invasion. To obtain a margin-negative resection, these tumors require en bloc gastrectomy with multivisceral resection (G+MVR), and contention remains regarding its safety and oncologic benefit. METHODS: We used the National Cancer Database to retrospectively evaluate the short- and long-term outcomes of patients with LAGC treated in the USA between 2004 and 2016. Associations with margin status and perioperative outcomes were calculated using logistic regression. Survival was estimated using Cox proportional hazards regression and the Kaplan-Meier method. RESULTS: Overall, 785 pathologic stage T4b (pT4b) patients diagnosed with LAGC underwent gastrectomy (n = 438) or G+MVR (n = 347). There was no association between G+MVR and short- or long-term mortality. Positive resection margins (HR 1.68, 95% CI 1.40-2.03), the presence of nodal disease (HRs 1.46-1.50), treatment at a high-volume center (HR 0.76, 95% CI 0.68-0.85), and the receipt of adjuvant chemotherapy (HR 0.64, 95% CI 0.51-0.80) were independently associated with overall survival. Diffuse-type histology was associated with higher rates of an R1 resection (OR 3.60, 95% CI 2.20-5.87). Perioperative and long-term survival metrics were comparable between patients with pT4a and pT4b LAGC who underwent a margin-negative G+MVR. Undergoing a margin-negative G+MVR imparted a 6-month survival benefit over non-curative gastrectomy alone (p < 0.001). CONCLUSIONS: Our study demonstrates the safety and long-term feasibility of G+MVR for disease clearance in well-selected patients with LAGC, and we advocate for their referral to high-volume centers for optimal care.


Subject(s)
Stomach Neoplasms , Chemotherapy, Adjuvant , Gastrectomy , Humans , Retrospective Studies , Stomach Neoplasms/surgery
20.
J Hepatol ; 74(5): 1145-1154, 2021 05.
Article in English | MEDLINE | ID: mdl-33276030

ABSTRACT

BACKGROUND & AIMS: While cholangiocarcinomas (CCAs) commonly express programmed cell death 1 (PD-1) and its ligand (PD-L1), they respond poorly to immune checkpoint inhibitors (ICIs). We aimed to determine whether stimulating antigen-presenting cells, including macrophages and dendritic cells, using a CD40 agonist could improve this response. METHODS: We compared treatment responses in subcutaneous, orthotopic, and 2 plasmid-based murine intrahepatic CCA (iCCA) models. Mice were treated for 4 weeks with weekly IgG control, a CD40 agonistic antibody, anti-PD-1, or the combination of both (anti-CD40/PD-1). Flow cytometric (FACS) analysis of lymphocytes and myeloid cell populations (including activation status) was performed. We used dendritic cell knockout mice, and macrophage, CD4+ and CD8+ T cell depletion models to identify effector cells. Anti-CD40/PD-1 was combined with chemotherapy (gemcitabine/cisplatin) to test for improved therapeutic efficacy. RESULTS: In all 4 models, anti-PD-1 alone was minimally efficacious. Mice exhibited a moderate response to CD40 agonist monotherapy. Combination anti-CD40/PD-1 therapy led to a significantly greater reduction in tumor burden. FACS demonstrated increased number and activation of CD4+ and CD8+ T cells, natural killer cells, and myeloid cells in tumor and non-tumor liver tissue of tumor-bearing mice treated with anti-CD40/PD-1. Depletion of macrophages, dendritic cells, CD4+ T cells, or CD8+ T cells abrogated treatment efficacy. Combining anti-CD40/PD-1 with gemcitabine/cisplatin resulted in a significant survival benefit compared to gemcitabine/cisplatin alone. CONCLUSION: CD40-mediated activation of macrophages and dendritic cells in iCCA significantly enhances response to anti-PD-1 therapy. This regimen may enhance the efficacy of first-line chemotherapy. LAY SUMMARY: Checkpoint inhibition, a common form of immune therapy, is generally ineffective for the treatment of cholangiocarcinoma. These tumors suppress the infiltration and function of surrounding immune cells. Stimulating immune cells such as macrophages and dendritic cells via the CD40 receptor activates downstream immune cells and enhances the response to checkpoint inhibitors.


Subject(s)
CD40 Antigens/agonists , Cholangiocarcinoma , Immune Checkpoint Inhibitors/pharmacology , Liver Neoplasms , Macrophage Activation/immunology , Tumor Microenvironment , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Cholangiocarcinoma/immunology , Cholangiocarcinoma/pathology , Cisplatin/pharmacology , Dendritic Cells/immunology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Collateral Sensitivity , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/drug effects , Macrophage-Activating Factors/immunology , Mice , Mice, Knockout , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...