Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Surg Endosc ; 38(2): 902-907, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37845533

ABSTRACT

INTRODUCTION: Adoption of robotic liver resections has been gradually increasing throughout the HPB surgical community over the past decade. Currently there is limited literature which demonstrates a significant benefit of robotic surgery for major hepatectomies over open or laparoscopic. As one of the first centers to develop a robotic HPB program, we have experienced improved outcomes over time with increasing utilization of robotics. Herein, we present our 10-year experience and outcomes for major robotic liver resections. METHODS: From 2012 to 2022, 361 robotic liver procedures were performed, including 100 major hepatectomies. A retrospective data review of the electronic medical record was performed evaluating outcomes after robotic major hepatectomy. Outcomes for the first 50 cases (Group A) and second 50 cases (Group B) were compared to identify any improvements in practice. Demographic and clinical outcome variables were analyzed. Data were assessed for normality, and Wilcoxon rank-sum, χ2 tests, and a logistic regression model were performed appropriate for the data. Stata v.17 was utilized, and significance was set as p < .05. RESULTS: There was no difference in median operative time (258 vs 256 min), EBL (500 vs 500 mL), median LOS (5 vs 3.5 days), 90-day readmission (14% vs 24%), major complications (14% vs 20%), and 90-day mortality (6% vs 4%) between early and late cases, respectively. ICU admissions and conversion rates were significantly lower in group B (14.0% vs 48.0%), while expert level difficulty indices were higher (82% vs 58%). CONCLUSION: Development of a robotic liver program with good outcomes is feasible over time. Our data suggest that our institutional learning curve for robotic major hepatectomy plateaued at approximately 50 cases.


Subject(s)
Laparoscopy , Liver Neoplasms , Robotic Surgical Procedures , Humans , Hepatectomy/methods , Retrospective Studies , Liver Neoplasms/surgery , Treatment Outcome , Length of Stay , Blood Loss, Surgical , Robotic Surgical Procedures/methods , Laparoscopy/methods , Operative Time , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Complications/surgery
2.
Surg Endosc ; 37(12): 9591-9600, 2023 12.
Article in English | MEDLINE | ID: mdl-37749202

ABSTRACT

BACKGROUND: Robotic pancreaticoduodenectomy (RPD) is an emerging alternative to open pancreaticoduodenectomy (OPD). Although RPD offers various theoretical advantages, it is used in less than 10% of all pancreaticoduodenectomies. The aim of this study was to report our 10-year experience and compare RPD outcomes with international benchmarks for OPD. METHODS: A retrospective review of a prospectively maintained institutional database was performed of consecutive patients who underwent RPD between January 2011 and December 2021. Patients were categorized into low-risk and high-risk groups according to the selection criteria set by the benchmark study. Their outcomes were compared to the international benchmark cut off values. Outcomes were then evaluated over time to identify improvements in practice and establish a learning curve. RESULTS: Of 201 RPDs, 36 were low-risk and 165 high-risk patients. Compared to the OPD benchmarks, outcomes of low-risk patients were within the cutoff values. High-risk patients were outside the cutoff for blood transfusions (26% vs. ≤ 23%), overall complications (78% vs. ≤ 73%), grade I-II complications (68% vs. ≤ 62%), and readmissions (22% vs ≤ 21%). Oncologic outcomes for high-risk patients were within benchmark cutoffs. Cases at the end of the learning curve included more pancreatic cancer (42% from 17%) and fewer low-risk patients (10% from 24%) than those at the beginning. After 41 RPD there was a decline in conversion rates and operative time. Between 95 and 143 cases operative time, transfusion rates, and LOS declined significantly. Complications did not differ over time. CONCLUSION: RPD yields results comparable to the established benchmarks in OPD in both low- and high-risk patients. Along the learning curve, RPD evolved with the inclusion of more high-risk cases while outcomes remained within benchmarks. Addition of a robotic HPB surgery fellowship did not compromise outcomes. These results suggest that RPD may be an option for high-risk patients at specialized centers.


Subject(s)
Pancreatic Neoplasms , Robotic Surgical Procedures , Robotics , Humans , Pancreaticoduodenectomy/methods , Benchmarking , Robotic Surgical Procedures/methods , Pancreatic Neoplasms/surgery , Retrospective Studies , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Complications/surgery
3.
Surg Endosc ; 37(9): 7288-7294, 2023 09.
Article in English | MEDLINE | ID: mdl-37558825

ABSTRACT

INTRODUCTION: The Japanese difficulty score (JDS) categorizes laparoscopic hepatectomy into low, intermediate, and high complexity procedures, and correlates with operative and postoperative outcomes. We sought to perform a validation study to determine if the JDS correlates with operative and postoperative indicators of surgical complexity for patients undergoing robotic-assisted hepatectomy. METHODS: Retrospective review of 657 minimally invasive hepatectomy procedures was performed between January 2008 through March 2019. Outcomes included operative time, estimated blood loss (EBL), blood transfusion, complications, post-hepatectomy liver failure (PHLF), length of stay, 30-day readmission, and 30-day and 90-day mortality. Patients were grouped based on JDS defined as: low (< 4), intermediate (4-6), and high (7 +) complexity procedures. Statistical comparisons were analyzed by ANOVA or χ2 test. RESULTS: 241 of 657 patients underwent robotic-assisted resection. Of these patients, 137 were included in the analysis based on JDS: 25 low, 58 intermediate, and 54 high. High JDS was associated with more major resections (≥ 4 contiguous segments) versus minor resections (median JDS 8 vs. 5, P < 0.0001). High JDS was associated with significantly longer operative times, higher EBL, and more blood transfusions. High JDS was associated with higher rates of PHLF at 16.7%, compared with 5.2% intermediate and 0.0% low, (P = 0.018). Complication rates, 30-day readmissions, and mortality rates were similar between groups. Median LOS was longer in patients with high JDS compared with intermediate and low (4 days vs. 3 days vs. 2 days; P = 0.0005). DISCUSSION: Higher JDS was associated with multiple indicators of operative complexity, including greater extent of resection, increased operative time, EBL, blood transfusion, PHLF, and LOS. This validation study supports the ability of the JDS to categorize patients undergoing robotic-assisted hepatectomy by complexity.


Subject(s)
Hepatic Insufficiency , Laparoscopy , Liver Failure , Liver Neoplasms , Robotic Surgical Procedures , Humans , Hepatectomy/adverse effects , Hepatectomy/methods , Robotic Surgical Procedures/adverse effects , Robotic Surgical Procedures/methods , East Asian People , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Complications/surgery , Length of Stay , Laparoscopy/adverse effects , Laparoscopy/methods , Operative Time , Retrospective Studies , Treatment Outcome
4.
HPB (Oxford) ; 25(7): 813-819, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37045742

ABSTRACT

BACKGROUND: Pancreatic necrosectomy with concomitant internal drainage is a single-stage treatment option for walled-off pancreatic necrosis (WOPN). However, an optimal minimally invasive technique has not been established. We evaluated the safety and single-intervention success rate of robotic pancreatic necrosectomy and internal drainage. METHODS: Patients with WOPN undergoing robotic pancreatic necrosectomy and internal drainage at a single institution from 2011-2022 were identified. The primary outcome was the rate of clinical symptom resolution following the index surgical intervention. RESULTS: 57 patients underwent robotic pancreatic necrosectomy and internal drainage, consisting of robotic cystgastrostomy (RCG, n = 37), robotic cystjejunostomy (RCJ, n = 13) and robotic fistulojejunostomy (RFJ, n = 7). Surgery was performed a median of 102 (range 28-1153) days following the onset of necrotizing pancreatitis. The median operative time was 187 (91-344) minutes and there were 2 (3.5%) conversions. The median length of hospital stay was 4 (2-38) days. Postoperative morbidity was 11%, and there was one (1.8%) 90-day mortality. At a median follow-up of 5.5 months, 53 (93%) patients had clinical symptom resolution after their index procedure and did not require any reintervention. CONCLUSION: In select patients, robotic pancreatic necrosectomy and internal drainage is safe and achieves a high single-intervention success rate.


Subject(s)
Pancreatitis, Acute Necrotizing , Robotic Surgical Procedures , Humans , Pancreatitis, Acute Necrotizing/diagnostic imaging , Pancreatitis, Acute Necrotizing/surgery , Robotic Surgical Procedures/adverse effects , Drainage/adverse effects , Drainage/methods , Treatment Outcome , Retrospective Studies , Necrosis
5.
J Gastrointest Surg ; 26(3): 623-634, 2022 03.
Article in English | MEDLINE | ID: mdl-34757511

ABSTRACT

BACKGROUND: The use of minimally invasive approaches for pancreatoduodenectomy has increased in recent years, but the risk of postoperative VTE is undefined. We aimed to compare venous thromboembolism (VTE) rates after open and minimally invasive pancreatoduodenectomy using an administrative dataset. METHODS: Patients who underwent pancreatoduodenectomy within the National Surgical Quality Improvement Program targeted pancreatectomy database (2016-2018) were identified. VTE was compared between patients who underwent open or minimally invasive pancreatoduodenectomy directly and after propensity score matching 1:1 for demographics, comorbidities, and peri-/intra-operative factors. RESULTS: A total of 12,227 patients underwent pancreatoduodenectomy during the study period (open: n = 11,217; minimally invasive: n = 1010). Before matching, the VTE rate was higher among patients who underwent minimally invasive pancreatoduodenectomy (5.2% vs. 3.8%, p = 0.033), and minimally invasive resection was independently associated with VTE (OR = 1.46, 95%CI = 1.09-2.06). After matching, there were 916 patients per group without differences in demographics or comorbidities. Patients who underwent minimally invasive pancreatoduodenectomy had longer median operative times (422 vs. 348 min). The VTE rate remained higher following minimally invasive pancreatoduodenectomy after matching (5.1% vs. 2.9%, p = 0.018), mainly driven by a higher DVT rate (3.9% vs. 1.7%, p = 0.005). CONCLUSIONS: Minimally invasive pancreatoduodenectomy is associated with a higher postoperative VTE rate compared to open pancreatoduodenectomy.


Subject(s)
Pulmonary Embolism , Venous Thromboembolism , Humans , Pancreaticoduodenectomy/adverse effects , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Propensity Score , Risk Factors , Venous Thromboembolism/epidemiology , Venous Thromboembolism/etiology
6.
Ann Surg Oncol ; 28(6): 3125-3134, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33051739

ABSTRACT

BACKGROUND: Modern chemotherapeutics have led to improved systemic disease control for patients with locally advanced pancreatic cancer (LAPC). Surgical strategies such as distal pancreatectomy with celiac axis resection (DP-CAR) are increasingly entertained. Herein we review procedure-specific outcomes and assess biologic rationale for DP-CAR. METHODS: A prospectively maintained single-institution database of all pancreatectomies was queried for patients undergoing DP-CAR. We excluded all patients for whom complete data were not available and those who were not treated with contemporary multi-agent therapy. Data were supplemented with dedicated chart review and outreach for long-term oncologic outcomes. RESULTS: Fifty-four patients underwent DP-CAR between 2008 and 2018. The median age was 62.7 years. Ninety-eight percent received induction chemotherapy. Arterial reconstruction was performed in 17% and concomitant visceral resection in 30%. The R0 resection rate was 87%. Postoperative complications were common (43%) with chyle leak being the most frequent (17%). Length of stay was 8 days, readmission occurred in one-third, and 90-day mortality was 2%. Disease recurrence occurred in 74% during a median follow up of 17.4 months. Median recurrence-free (RFS) and overall survival (OS) were 9 and 25 months, respectively. CONCLUSIONS: Following modern induction paradigms, DP-CAR can be performed with low mortality, manageable morbidity, and excellent rates of margin-negative resection in high-volume settings. The profile of complications of DP-CAR is distinct from pancreaticoduodenectomy and simple distal pancreatectomy. OS and RFS are similar to those undergoing resection of borderline resectable and resectable disease. Improved systemic disease control will likely lead to increasing utilization of aggressive surgical approaches to LAPC.


Subject(s)
Pancreatectomy , Pancreatic Neoplasms , Celiac Artery/surgery , Humans , Middle Aged , Neoplasm Recurrence, Local/surgery , Pancreatic Neoplasms/surgery , Retrospective Studies
7.
HPB (Oxford) ; 23(4): 575-586, 2021 04.
Article in English | MEDLINE | ID: mdl-32900612

ABSTRACT

BACKGROUND: Lymph node (LN) metastasis is associated with decreased survival following resection for pancreatic ductal adenocarcinoma (PDAC). In N0 disease, increasing total evaluated LN (ELN) correlates with improved outcomes suggesting patients may be understaged when LNs are undersampled. We aim to assess the optimal number of examined lymph nodes (ELN) following pancreatectomy. METHODS: Data from 1837 patients undergoing surgery were prospectively collected. The binomial probability law was utilized to analyze the minimum number of examined LNs (minELN) and accurately characterize each histopathologic stage. LN ratio (LNR) was compared to American Joint Committee on Cancer (AJCC) guidelines. RESULTS: As ELN total increased, the likelihood of finding node positive disease increased. An evaluation based upon the binomial probability law suggested an optimal minELN of 12 for accurate AJCC N staging. As the number of ELNs increased, the discriminatory capacity of alternative strategies to characterize LN disease exceeded that offered by AJCC N stage. CONCLUSION: This is the first study dedicated to optimizing histopathologic staging in PDAC using models of minELN informed by the binomial probability law. This study highlights two separate cutoffs for ELNs depending upon prognostic goal and validates that 12 LNs are adequate to determine AJCC N stage for the majority of patients.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Humans , Lymph Node Excision , Lymph Nodes/pathology , Lymph Nodes/surgery , Neoplasm Staging , Pancreas/pathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Prognosis , Survival Rate
8.
J Pancreat Cancer ; 6(1): 85-95, 2020.
Article in English | MEDLINE | ID: mdl-32999955

ABSTRACT

Background: Underutilization of operative management of early stage pancreatic cancer is associated with sociodemographic variables, including age, race, facility type, insurance, and education. It is currently unclear how these variables are associated with survival in patients who undergo surgery. Methods: Patients with clinical stage I pancreatic adenocarcinoma were identified within the National Cancer Database (2010-2016). Utilization of surgery and nonoperative management was determined. Nonclinical factors associated with nonoperative management were identified by multivariable analysis. The association between nonclinical factors and survival was assessed in patients who received operative management. Results: A total of 17,833 patients with clinical stage I pancreatic cancer were identified, and 41.2% underwent operative intervention. Approximately 46% of nonoperatively managed patients lacked a contraindication. Operatively managed patients had longer overall survival (OS) than those who were nonoperatively managed or untreated (25.1 months vs. 11.1 months vs. 5.1 months, p < 0.0001). Factors associated with nonoperative management included age, black/Hispanic race, nonacademic facilities, nonprivate health insurance, lower education level, and lower income. In operatively managed patients, nonclinical factors associated with lower OS included Medicaid (hazard ratio [HR] 1.27) and treatment at nonacademic facilities (HR 1.20-1.22). Patients on Medicaid received less adjuvant therapy and had higher 30- and 90-day mortality rates. Patients treated at nonacademic facilities received less neoadjuvant therapy, had worse pathologic outcomes, and had higher 30- and 90-day mortality rates. Conclusions: Surgical management is underutilized in clinical stage I pancreatic cancer. Primary insurance payor and facility type appear to be associated with OS in patients who undergo operative management.

9.
Curr Oncol Rep ; 21(8): 67, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31236716

ABSTRACT

PURPOSE OF REVIEW: Gastric adenocarcinoma is the fifth most common and the third most lethal cancer worldwide. Surgery is the only chance of cure, but recurrence is common, even with complete resection. RECENT FINDINGS: Advances in diagnosis and staging, genomic classification, surgical resection and treatment of peritoneal disease, systemic chemotherapy and chemoradiation, and targeted and immune therapies have led to the current multidisciplinary approach to gastric adenocarcinoma. Treatment of gastric cancer is rapidly evolving in an effort to combat this challenging disease.


Subject(s)
Adenocarcinoma/therapy , Stomach Neoplasms/therapy , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Chemoradiotherapy , Chemotherapy, Adjuvant , General Surgery , Humans , Immunotherapy , Molecular Targeted Therapy , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
10.
PLoS One ; 14(5): e0216275, 2019.
Article in English | MEDLINE | ID: mdl-31042769

ABSTRACT

INTRODUCTION: Murine Kupffer cells (KCs) comprise CD11bhi and F4/80hi subsets. Tissue-resident macrophages are known to express the tyrosine kinase receptors colony-stimulating factor 1 receptor (Csf1r) and Mer. However, the expression of Csf1r and Mer on KC subsets and the importance of these tyrosine kinases during liver regeneration (LR) are unknown. METHODS: KCs from wild-type and Csf1r-GFP mice were characterized by flow cytometry. Partial hepatectomy (PH) was performed in mice treated with clodronate liposomes, a Csf1r small molecule inhibitor or depleting antibody, or a small molecule Mer inhibitor. Sera and livers were analyzed. The function of sorted KC subsets was tested in vitro. RESULTS: Mer was specifically expressed on tissue-resident F4/80hi KCs, 55% of which also expressed Csf1r. Mer+Csf1r+ and Mer+Csf1r- KCs had distinct expression of macrophage markers. Csf1r inhibition in mice reduced F4/80hi KCs by approximately 50%, but did not affect CD11bhi KCs. Clodronate liposomes depleted F4/80hi KCs, but also altered levels of other intrahepatic leukocytes. Csf1r inhibition delayed LR, as demonstrated by a 20% reduction in liver-to-body weight ratios 7 days after PH. At 36h after PH, Csf1r inhibition increased serum ALT and histological liver injury, and decreased liver cell proliferation. A small molecule inhibitor of Mer did not alter the percentage of KCs or their proliferation and just modestly delayed LR. In vitro, Csf1r or Mer inhibition did not decrease KC viability, but did attenuate their cytokine response to stimulation. CONCLUSIONS: F4/80hi KCs are Mer+ and can be subdivided based on Csf1r expression. Csf1r or Mer inhibition each reduces KC cytokine production and delays LR.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Kupffer Cells/metabolism , Liver Regeneration/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Immunologic/metabolism , Animals , Antigens, Differentiation/analysis , Hepatectomy , Kupffer Cells/cytology , Kupffer Cells/drug effects , Macrophages/metabolism , Mice , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors
11.
J Surg Oncol ; 120(1): 23-29, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31001875

ABSTRACT

Breaking bad news and navigating difficult conversations is challenging and a frequent occurrence in surgical oncology practice. Cancer communication recommendations are guided by ethical principles, legal precedence, and emerging evidence regarding patient preference and efficacy of communication tools. Evidence continues to mount supporting a patient-centered communication approach and a model of shared decision making. Likewise, physician training in effective patient-centered cancer communication continues to evolve.


Subject(s)
Neoplasms/therapy , Patient-Centered Care , Physician-Patient Relations , Truth Disclosure , Clinical Protocols , Communication , Decision Making/ethics , Education, Medical , Humans , Neoplasms/psychology , Palliative Care , Patient Acceptance of Health Care , Patient Participation/legislation & jurisprudence , Patient-Centered Care/ethics , Patient-Centered Care/legislation & jurisprudence , Physician-Patient Relations/ethics , Prognosis , Terminal Care , Truth Disclosure/ethics
12.
Cancer Immunol Res ; 6(4): 434-447, 2018 04.
Article in English | MEDLINE | ID: mdl-29467128

ABSTRACT

Tyrosine kinase inhibition of gastrointestinal stromal tumors (GIST) is effective but typically culminates in resistance and is rarely curative. Immunotherapy has potential application to GIST, as we previously showed that T-cell checkpoint blockade increases the antitumor effects of imatinib. Here, we showed that ligation of CD40 using an agonistic antibody (anti-CD40) activated tumor-associated macrophages (TAMs) in vivo in a knock-in mouse model of GIST harboring a germline mutation in Kit exon 11. Activated TAMs had greater TNFα production and NFκB signaling and directly inhibited tumor cells in vitro Anti-CD40 required concomitant therapy with imatinib for efficacy and depended on TAMs, and to a lesser extent CD8+ T cells, but not on CD4+ T cells or B cells. In an analysis of 50 human GIST specimens by flow cytometry, we found that CD40 was expressed on human TAMs and tumor cells yet was downregulated after response to imatinib. CD40 ligation did not have a direct inhibitory effect on human GIST cells. Our findings provide the rationale for combining anti-CD40 and tyrosine kinase inhibition to treat human GIST. Cancer Immunol Res; 6(4); 434-47. ©2018 AACR.


Subject(s)
CD40 Antigens/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Stromal Tumors/immunology , Gastrointestinal Stromal Tumors/metabolism , Imatinib Mesylate/pharmacology , Macrophages/drug effects , Macrophages/immunology , Animals , Biomarkers , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Drug Synergism , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Gene Expression , Humans , Immunophenotyping , Immunotherapy , Macrophages/metabolism , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Tumor Burden , Xenograft Model Antitumor Assays
13.
Mol Cancer Ther ; 16(9): 1954-1966, 2017 09.
Article in English | MEDLINE | ID: mdl-28611108

ABSTRACT

Gastrointestinal stromal tumor (GIST) is the most common type of sarcoma and usually harbors either a KIT or PDGFRA mutation. However, the molecular basis for tumor malignancy is not well defined. Although the Wnt/ß-catenin signaling pathway is important in a variety of cancers, its role in GIST is uncertain. Through analysis of nearly 150 human GIST specimens, we found that some human GISTs expressed ß-catenin and contained active, dephosphorylated nuclear ß-catenin. Furthermore, advanced human GISTs expressed reduced levels of the Wnt antagonist DKK4. Accordingly, in human GIST T1 cells, Wnt stimulation increased ß-catenin-mediated transcriptional activity in a reporter assay as well as transcription of the downstream target genes Axin2 and CCND1 In contrast, DKK4 overexpression in GIST T1 cells reduced Wnt/ß-catenin signaling. In addition, we showed that nuclear ß-catenin stability was partially regulated by the E3 ligase COP1, as demonstrated with coimmunoprecipitation and COP1 knockdown. Three molecular inhibitors of the Wnt/ß-catenin pathway demonstrated antitumor efficacy in various GIST models, both in vitro and in vivo Notably, the tankyrase inhibitor G007-LK alone had substantial activity against tumors of genetically engineered KitV558Δ/+ mice, and the effect was increased by the addition of the Kit inhibitor imatinib mesylate. Collectively, our findings demonstrate that Wnt/ß-catenin signaling is a novel therapeutic target for selected untreated or imatinib-resistant GISTs. Mol Cancer Ther; 16(9); 1954-66. ©2017 AACR.


Subject(s)
Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Wnt Signaling Pathway , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Disease Models, Animal , Gastrointestinal Stromal Tumors/drug therapy , Humans , Imatinib Mesylate/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Staging , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway/drug effects , Wnt3A Protein/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
14.
Clin Cancer Res ; 23(2): 454-465, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27470968

ABSTRACT

PURPOSE: Tyrosine kinase inhibitors are effective in gastrointestinal stromal tumors (GISTs) but often are of transient benefit as resistance commonly develops. Immunotherapy, particularly blockade of the inhibitory receptor programmed death 1 (PD-1) or the ligand programmed death ligand 1 (PD-L1), has shown effectiveness in a variety of cancers. The functional effects of PD-1/PD-L1 blockade are unknown in GISTs. EXPERIMENTAL DESIGN: We analyzed tumor and matched blood samples from 85 patients with GISTs and determined the expression of immune checkpoint molecules using flow cytometry. We investigated the combination of imatinib with PD-1/PD-L1 blockade in KitV558Δ/+ mice that develop GISTs. RESULTS: The inhibitory receptors PD-1, lymphocyte activation gene 3, and T-cell immunoglobulin mucin-3 were upregulated on tumor-infiltrating T cells compared with T cells from matched blood. PD-1 expression on T cells was highest in imatinib-treated human GISTs. Meanwhile, intratumoral PD-L1 expression was variable. In human GIST cell lines, treatment with imatinib abrogated the IFNγ-induced upregulation of PD-L1 via STAT1 inhibition. In KitV558Δ/+ mice, imatinib downregulated IFNγ-related genes and reduced PD-L1 expression on tumor cells. PD-1 and PD-L1 blockade in vivo each had no efficacy alone but enhanced the antitumor effects of imatinib by increasing T-cell effector function in the presence of KIT and IDO inhibition. CONCLUSIONS: PD-1/PD-L1 blockade is a promising strategy to improve the effects of targeted therapy in GISTs. Collectively, our results provide the rationale to combine these agents in human GISTs. Clin Cancer Res; 23(2); 454-65. ©2016 AACR.


Subject(s)
B7-H1 Antigen/immunology , Gastrointestinal Stromal Tumors/therapy , Programmed Cell Death 1 Receptor/immunology , STAT1 Transcription Factor/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/immunology , Gastrointestinal Stromal Tumors/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate/administration & dosage , Immunotherapy , Lymphocyte Activation/drug effects , Mice , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , STAT1 Transcription Factor/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
15.
Cancer Res ; 75(10): 2061-70, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25836719

ABSTRACT

Gastrointestinal stromal tumors (GIST) are the most common adult sarcomas and the oncogenic driver is usually a KIT or PDGFRA mutation. Although GISTs are often initially sensitive to imatinib or other tyrosine kinase inhibitors, resistance generally develops, necessitating backup strategies for therapy. In this study, we determined that a subset of human GIST specimens that acquired imatinib resistance acquired expression of activated forms of the MET oncogene. MET activation also developed after imatinib therapy in a mouse model of GIST (KitV558del/+ mice), where it was associated with increased tumor hypoxia. MET activation also occurred in imatinib-sensitive human GIST cell lines after imatinib treatment in vitro. MET inhibition by crizotinib or RNA interference was cytotoxic to an imatinib-resistant human GIST cell population. Moreover, combining crizotinib and imatinib was more effective than imatinib alone in imatinib-sensitive GIST models. Finally, cabozantinib, a dual MET and KIT small-molecule inhibitor, was markedly more effective than imatinib in multiple preclinical models of imatinib-sensitive and imatinib-resistant GIST. Collectively, our findings showed that activation of compensatory MET signaling by KIT inhibition may contribute to tumor resistance. Furthermore, our work offered a preclinical proof of concept for MET inhibition by cabozantinib as an effective strategy for GIST treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Stromal Tumors/drug therapy , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Pyrimidines/pharmacology , Anilides/pharmacology , Animals , Cell Line, Tumor , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate , Indoles/pharmacology , Inhibitory Concentration 50 , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-kit/genetics , Pyridines/pharmacology , Pyrroles/pharmacology , Signal Transduction , Sunitinib , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
Clin Cancer Res ; 20(12): 3146-58, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24718867

ABSTRACT

PURPOSE: Malignant peripheral nerve sheath tumor (MPNST) is a highly aggressive tumor type that is resistant to chemotherapy and there are no effective therapies. MPNSTs have been shown to have gene amplification for receptor tyrosine kinases (RTK), PDGFR and c-Kit. We tested the c-Kit inhibitor, imatinib, and PLX3397, a selective c-Fms and c-Kit inhibitor, to evaluate their efficacy against MPNST cells in vitro and in vivo. EXPERIMENTAL DESIGN: We tested the efficacy of imatinib or PLX3397 either alone or in combination with TORC1 inhibitor rapamycin in a cell proliferation assay in vitro and by immunoblotting to determine target inhibition. Immunoblotting and immunohistochemical analysis was further carried out using xenograft samples in vivo. RESULTS: Our in vitro studies show that imatinib and PLX3397 similarly inhibit cell growth and this can be enhanced with rapamycin with comparable target specificity. However, in vivo studies clearly demonstrate that compared with imatinib, PLX3397 results in sustained blockade of c-Kit, c-Fms, and PDGFRß, resulting in significant suppression of tumor growth. Moreover, staining for Iba-1, a marker for macrophages, indicates that PLX3397 results in significant depletion of macrophages in the growing tumors. The combination of PLX3397 and rapamycin results in even greater macrophage depletion with continued growth suppression, even when the drug treatment is discontinued. CONCLUSIONS: Taken together, our data strongly suggest that PLX3397 is superior to imatinib in the treatment of MPNSTs, and the combination of PLX3397 with a TORC1 inhibitor could provide a new therapeutic approach for the treatment of this disease.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Macrophages/drug effects , Neurilemmoma/drug therapy , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrroles/pharmacology , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzamides/administration & dosage , Blotting, Western , Cell Proliferation/drug effects , Humans , Imatinib Mesylate , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred ICR , Mice, SCID , Neurilemmoma/metabolism , Neurilemmoma/pathology , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Sirolimus/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...