Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Ann Surg Oncol ; 31(2): 1252-1263, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38006531

ABSTRACT

BACKGROUND: Discontinuation of the Codman 3000 pump in 2018 left no Food and Drug Administration (FDA)-approved hepatic artery infusion (HAI) device for unresectable colorectal liver metastases (uCLM) and intrahepatic cholangiocarcinoma (uIHC). Historically, HAI has been performed at academic medical centers in large metropolitan areas, which are often inaccessible to rural patients. Consequently, feasibility of dissemination of HAI to rural populations is unknown. PATIENTS AND METHODS: Under an FDA investigational device exemption, we opened the only HAI program in Kentucky and enrolled patients with uCLM and uIHC in a phase I clinical trial. The trial examined the safety of the hybrid Codman catheter/Medtronic SynchroMed II pump (hCMP) combination, defined as successful completion of one cycle of HAI chemotherapy. Rural feasibility was assessed by number of missed pump fills appointments. RESULTS: A total of 21 patients (n = 17 uCLM, n = 4 uIHC) underwent hCMP implantation before accrual was stopped early owing to FDA approval of the Intera 3000 pump. 20/21 (95%) patients met the primary safety endpoint. Serious adverse events (AEs) included a grade 5 coronavirus disease 2019 (COVID-19) infection (n = 1) and a grade 3 catheter erosion into the bowel (n = 1). Biliary sclerosis developed in two patients (9.5%). Median distance to infusion center was 47.6 miles (2-138 miles), and 62% were from Appalachia, yet there were no missed pump fill appointments. The 2-year overall survival was 82.4% (uCLM) and 50% (uIHC). CONCLUSIONS: The hCMP device had an acceptable safety profile. Despite the complexity of starting a new HAI program, early results showed feasibility for HAI delivery in a rural catchment area and comparable outcomes to larger urban-based HAI centers.


Subject(s)
Bile Duct Neoplasms , Colorectal Neoplasms , Liver Neoplasms , Vascular Access Devices , Humans , Colorectal Neoplasms/pathology , Hepatic Artery/pathology , Feasibility Studies , Antineoplastic Combined Chemotherapy Protocols , Liver Neoplasms/secondary , Infusions, Intra-Arterial , Bile Ducts, Intrahepatic/pathology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/etiology
3.
Cancers (Basel) ; 15(11)2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37296998

ABSTRACT

Gastric adenocarcinoma (GAd) is the third leading cause of cancer-related deaths worldwide. Most patients require perioperative chemotherapy, yet methods to accurately predict responses to therapy are lacking. Thus, patients may be unnecessarily exposed to considerable toxicities. Here, we present a novel methodology using patient-derived organoids (PDOs) that rapidly and accurately predicts the chemotherapy efficacy for GAd patients. Methods:Endoscopic GAd biopsies were obtained from 19 patients, shipped overnight, and PDOs were developed within 24 h. Drug sensitivity testing was performed on PDO single-cells with current standard-of-care systemic GAd regimens and cell viability was measured. Whole exome sequencing was used to confirm the consistency of tumor-related gene mutations and copy number alterations between primary tumors, PDOs, and PDO single-cells. Results:Overall, 15 of 19 biopsies (79%) were appropriate for PDO creation and single-cell expansion within 24 h of specimen collection and overnight shipment. With our PDO single-cell technique, PDOs (53%) were successfully developed. Subsequently, two PDO lines were subjected to drug sensitivity testing within 12 days from initial biopsy procurement. Drug sensitivity assays revealed unique treatment response profiles for combination drug regimens in both of the two unique PDOs, which corresponded with the clinical response. Conclusions:The successful creation of PDOs within 24 h of endoscopic biopsy and rapid drug testing within 2 weeks demonstrate the feasibility of our novel approach for future applications in clinical decision making. This proof of concept sets the foundation for future clinical trials using PDOs to predict clinical responses to GAd therapies.

4.
Surgery ; 174(1): 101-105, 2023 07.
Article in English | MEDLINE | ID: mdl-37137740

ABSTRACT

Implementation of a successful hepatic artery infusion pump program requires numerous factors to be in place, and the lack of any of these may lead to program failure. First and foremost, hepatic artery infusion pump programs must have adequate surgical expertise in the complex technical aspects of hepatic artery infusion pump implantation and postoperative management. Most new hepatic artery infusion pump programs are initiated by a surgeon and led in conjunction with a medical oncologist. Medical oncology experience in floxuridine dosing is critical in maximizing the treatment doses and the number of cycles administered while avoiding biliary toxicity. This is facilitated by collaboration with an engaged pharmacy team. To have adequate patient volume for a successful program, internal and external stakeholders must have buy-in, including surgical and medical oncology colleagues unfamiliar with hepatic artery infusion pumps, colorectal surgery, and other referring providers. Programmatic support must be obtained from the hospital, cancer center, and department administration. Day-to-day pump access for chemotherapy and maintenance saline fills must be performed by appropriately trained infusion nurses to avoid complications. Nuclear and diagnostic radiology experience is key to identifying extrahepatic perfusion and hepatic artery infusion pump-specific complications. Additionally, skilled interventional radiologists and gastroenterologists are necessary to identify and treat rare complications rapidly. Finally, given the current rapid expansion of hepatic artery infusion pump programs, new programs must identify engaged mentors to help guide patient selection, navigate the nuanced issues that may arise, and provide advice in the case of complications. Although hepatic artery infusion pump dissemination outside of several major tertiary centers previously had stalled, establishing a successful and active hepatic artery infusion pump is feasible with appropriate training, mentorship, and thoughtful assembly of a dedicated multidisciplinary team.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Humans , Hepatic Artery , Liver Neoplasms/surgery , Floxuridine/therapeutic use , Infusion Pumps, Implantable , Infusions, Intra-Arterial
9.
J Gastrointest Surg ; 25(7): 1885-1895, 2021 07.
Article in English | MEDLINE | ID: mdl-32989690

ABSTRACT

The microbiome plays a major role in human physiology by influencing obesity, inducing inflammation, and impacting cancer therapies. During the 60th Annual Meeting of the Society of the Alimentary Tract (SSAT) at the State-of-the-Art Conference, experts in the field discussed the influence of the microbiome. This paper is a summary of the influence of the microbiome on obesity, inflammatory bowel disease, pancreatic cancer, cancer therapies, and gastrointestinal optimization. This review shows how the microbiome plays an important role in the development of diseases and surgical complications. Future studies are needed in targeting the gut microbiome to develop individualized therapies.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Microbiota , Humans , Inflammatory Bowel Diseases/therapy , Obesity
12.
Surgery ; 168(2): 222-225, 2020 08.
Article in English | MEDLINE | ID: mdl-32600881

ABSTRACT

BACKGROUND: The coronavirus disease 2019 pandemic has negatively affected the training of general surgery chief residents during the last trimester of their residency. Our goal was to evaluate the educational concerns of graduating general surgery chief residents during the coronavirus disease 2019 pandemic. METHODS: An anonymous web-based survey was distributed between March 31 and April 7, 2020 to all current general surgery chief residents from 6 academic medical centers in Boston, Massachusetts. Interviews were also conducted with attending surgeons from participating institutions. RESULTS: A total of 24 of 39 general surgery chief residents participated in our survey (61.5% response rate). General surgery chief residents were most concerned about the potential delay in the date of board examinations, followed by not feeling adequately prepared for the board examinations and a possible delay in the graduation date. Whereas not having enough cases to feel ready for fellowship or job and not achieving a sufficient number of cases to meet the requirements for graduation were only moderately concerning to chief residents, attending surgeons stressed a greater importance on the loss of the operative experience as nearly all (93.3%) of them suggested a personalized approach for additional general surgery training during fellowship or job onboarding. CONCLUSION: In addition to the dramatic impact on public health, the coronavirus disease 2019 outbreak has also caused unprecedented changes to surgical education. Therefore, creative interventions are needed to help general surgery chief residents successfully transition into the next phase of their surgical career.


Subject(s)
Betacoronavirus , Clinical Competence , Coronavirus Infections/epidemiology , Education, Medical, Graduate , Internship and Residency , Pneumonia, Viral/epidemiology , Surgeons , Academic Medical Centers , Adult , Attitude of Health Personnel , Boston/epidemiology , COVID-19 , Educational Measurement , Female , Humans , Male , Middle Aged , Pandemics , SARS-CoV-2 , Specialty Boards , Surveys and Questionnaires
13.
Nat Immunol ; 21(4): 442-454, 2020 04.
Article in English | MEDLINE | ID: mdl-32152508

ABSTRACT

Programmed cell death protein 1 (PD-1) ligation delimits immunogenic responses in T cells. However, the consequences of programmed cell death 1 ligand 1 (PD-L1) ligation in T cells are uncertain. We found that T cell expression of PD-L1 in cancer was regulated by tumor antigen and sterile inflammatory cues. PD-L1+ T cells exerted tumor-promoting tolerance via three distinct mechanisms: (1) binding of PD-L1 induced STAT3-dependent 'back-signaling' in CD4+ T cells, which prevented activation, reduced TH1-polarization and directed TH17-differentiation. PD-L1 signaling also induced an anergic T-bet-IFN-γ- phenotype in CD8+ T cells and was equally suppressive compared to PD-1 signaling; (2) PD-L1+ T cells restrained effector T cells via the canonical PD-L1-PD-1 axis and were sufficient to accelerate tumorigenesis, even in the absence of endogenous PD-L1; (3) PD-L1+ T cells engaged PD-1+ macrophages, inducing an alternative M2-like program, which had crippling effects on adaptive antitumor immunity. Collectively, we demonstrate that PD-L1+ T cells have diverse tolerogenic effects on tumor immunity.


Subject(s)
B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Macrophages/immunology , Self Tolerance/immunology , Animals , Cell Differentiation/immunology , Cell Line, Tumor , Female , Humans , Interferon-gamma/immunology , Male , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology
14.
Hepatology ; 71(2): 477-494, 2020 02.
Article in English | MEDLINE | ID: mdl-31529720

ABSTRACT

BACKGROUND AND AIMS: The recruitment and activation of inflammatory cells in the liver delineates the transition from hepatic steatosis to steatohepatitis (SH). APPROACH AND RESULTS: We found that in SH, γδT cells are recruited to the liver by C-C chemokine receptor (CCR) 2, CCR5, and nucleotide-binding oligomerization domain-containing protein 2 signaling and are skewed toward an interleukin (IL)-17A+ phenotype in an inducible costimulator (ICOS)/ICOS ligand-dependent manner. γδT cells exhibit a distinct Vγ4+ , PD1+ , Ly6C+ CD44+ phenotype in SH. Moreover, γδT cells up-regulate both CD1d, which is necessary for lipid-based antigens presentation, and the free fatty acid receptor, CD36. γδT cells are stimulated to express IL-17A by palmitic acid and CD1d ligation. Deletion, depletion, and targeted interruption of γδT cell recruitment protects against diet-induced SH and accelerates disease resolution. CONCLUSIONS: We demonstrate that hepatic γδT cells exacerbate SH, independent of IL-17 expression, by mitigating conventional CD4+ T-cell expansion and modulating their inflammatory program by CD1d-dependent vascular endothelial growth factor expression.


Subject(s)
Adaptive Immunity/physiology , Fatty Liver/etiology , Immunity, Innate/physiology , Intraepithelial Lymphocytes/physiology , Animals , Female , Male , Mice
15.
Nature ; 574(7777): 264-267, 2019 10.
Article in English | MEDLINE | ID: mdl-31578522

ABSTRACT

Bacterial dysbiosis accompanies carcinogenesis in malignancies such as colon and liver cancer, and has recently been implicated in the pathogenesis of pancreatic ductal adenocarcinoma (PDA)1. However, the mycobiome has not been clearly implicated in tumorigenesis. Here we show that fungi migrate from the gut lumen to the pancreas, and that this is implicated in the pathogenesis of PDA. PDA tumours in humans and mouse models of this cancer displayed an increase in fungi of about 3,000-fold compared to normal pancreatic tissue. The composition of the mycobiome of PDA tumours was distinct from that of the gut or normal pancreas on the basis of alpha- and beta-diversity indices. Specifically, the fungal community that infiltrated PDA tumours was markedly enriched for Malassezia spp. in both mice and humans. Ablation of the mycobiome was protective against tumour growth in slowly progressive and invasive models of PDA, and repopulation with a Malassezia species-but not species in the genera Candida, Saccharomyces or Aspergillus-accelerated oncogenesis. We also discovered that ligation of mannose-binding lectin (MBL), which binds to glycans of the fungal wall to activate the complement cascade, was required for oncogenic progression, whereas deletion of MBL or C3 in the extratumoral compartment-or knockdown of C3aR in tumour cells-were both protective against tumour growth. In addition, reprogramming of the mycobiome did not alter the progression of PDA in Mbl- (also known as Mbl2) or C3-deficient mice. Collectively, our work shows that pathogenic fungi promote PDA by driving the complement cascade through the activation of MBL.


Subject(s)
Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Carcinogenesis , Carcinoma, Pancreatic Ductal/microbiology , Carcinoma, Pancreatic Ductal/pathology , Gastrointestinal Microbiome/immunology , Mannose-Binding Lectin/immunology , Mycobiome/immunology , Adenocarcinoma/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Case-Control Studies , Complement Activation , Complement C3/deficiency , Complement C3/immunology , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred C57BL
16.
Cancer Discov ; 9(9): 1288-1305, 2019 09.
Article in English | MEDLINE | ID: mdl-31266770

ABSTRACT

Unconventional T-lymphocyte populations are emerging as important regulators of tumor immunity. Despite this, the role of TCRαß+CD4-CD8-NK1.1- innate αß T cells (iαßT) in pancreatic ductal adenocarcinoma (PDA) has not been explored. We found that iαßTs represent ∼10% of T lymphocytes infiltrating PDA in mice and humans. Intratumoral iαßTs express a distinct T-cell receptor repertoire and profoundly immunogenic phenotype compared with their peripheral counterparts and conventional lymphocytes. iαßTs comprised ∼75% of the total intratumoral IL17+ cells. Moreover, iαßT-cell adoptive transfer is protective in both murine models of PDA and human organotypic systems. We show that iαßT cells induce a CCR5-dependent immunogenic macrophage reprogramming, thereby enabling marked CD4+ and CD8+ T-cell expansion/activation and tumor protection. Collectively, iαßTs govern fundamental intratumoral cross-talk between innate and adaptive immune populations and are attractive therapeutic targets. SIGNIFICANCE: We found that iαßTs are a profoundly activated T-cell subset in PDA that slow tumor growth in murine and human models of disease. iαßTs induce a CCR5-dependent immunogenic tumor-associated macrophage program, T-cell activation and expansion, and should be considered as novel targets for immunotherapy.See related commentary by Banerjee et al., p. 1164.This article is highlighted in the In This Issue feature, p. 1143.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Macrophages/immunology , Pancreatic Neoplasms/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/immunology , Animals , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Female , Humans , Immunity, Innate , Immunotherapy, Adoptive , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Pancreatic Neoplasms/therapy , T-Lymphocytes/transplantation , Tumor Microenvironment
17.
Nat Commun ; 10(1): 1424, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926808

ABSTRACT

The drivers and the specification of CD4+ T cell differentiation in the tumor microenvironment and their contributions to tumor immunity or tolerance are incompletely understood. Using models of pancreatic ductal adenocarcinoma (PDA), we show that a distinct subset of tumor-infiltrating dendritic cells (DC) promotes PDA growth by directing a unique TH-program. Specifically, CD11b+CD103- DC predominate in PDA, express high IL-23 and TGF-ß, and induce FoxP3neg tumor-promoting IL-10+IL-17+IFNγ+ regulatory CD4+ T cells. The balance between this distinctive TH program and canonical FoxP3+ TREGS is unaffected by pattern recognition receptor ligation and is modulated by DC expression of retinoic acid. This TH-signature is mimicked in human PDA where it is associated with immune-tolerance and diminished patient survival. Our data suggest that CD11b+CD103- DC promote CD4+ T cell tolerance in PDA which may underscore its resistance to immunotherapy.


Subject(s)
Dendritic Cells/immunology , Interleukin-10/metabolism , Interleukin-17/metabolism , Pancreatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Disease Progression , Forkhead Transcription Factors , Gene Expression Regulation, Neoplastic , Humans , Lectins, C-Type/metabolism , Mice, Inbred C57BL , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Signal Transduction , Th17 Cells/immunology , Toll-Like Receptor 2/metabolism , Tretinoin/metabolism , Pancreatic Neoplasms
18.
Oncogene ; 38(23): 4512-4526, 2019 06.
Article in English | MEDLINE | ID: mdl-30742098

ABSTRACT

Liver fibrosis and fibrosis-associated hepatocarcinogenesis are driven by chronic inflammation and are leading causes of morbidity and death worldwide. SYK signaling regulates critical processes in innate and adaptive immunity, as well as parenchymal cells. We discovered high SYK expression in the parenchymal hepatocyte, hepatic stellate cell (HSC), and the inflammatory compartments in the fibrotic liver. We postulated that targeting SYK would mitigate hepatic fibrosis and oncogenic progression. We found that inhibition of SYK with the selective small molecule inhibitors Piceatannol and PRT062607 markedly protected against toxin-induced hepatic fibrosis, associated hepatocellular injury and intra-hepatic inflammation, and hepatocarcinogenesis. SYK inhibition resulted in increased intra-tumoral expression of the p16 and p53 but decreased expression of Bcl-xL and SMAD4. Further, hepatic expression of genes regulating angiogenesis, apoptosis, cell cycle regulation, and cellular senescence were affected by targeting SYK. We found that SYK inhibition mitigated both HSC trans-differentiation and acquisition of an inflammatory phenotype in T cells, B cells, and myeloid cells. However, in vivo experiments employing selective targeted deletion of SYK indicated that only SYK deletion in the myeloid compartment was sufficient to confer protection against fibrogenic progression. Targeting SYK promoted myeloid cell differentiation into hepato-protective TNFαlow CD206hi phenotype downregulating mTOR, IL-8 signaling and oxidative phosphorylation. Collectively, these data suggest that SYK is an attractive target for experimental therapeutics in treating hepatic fibrosis and oncogenesis.


Subject(s)
Liver Cirrhosis/pathology , Myeloid Cells/metabolism , Signal Transduction , Syk Kinase/metabolism , Animals , Carcinogenesis , Carcinoma, Hepatocellular/metabolism , Cell Transdifferentiation , Cyclohexylamines/pharmacology , Female , Fibrosis , Hepatic Stellate Cells/cytology , Humans , Interleukin-8/metabolism , Lectins, C-Type/metabolism , Liver/metabolism , Liver Neoplasms/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Neoplasms, Experimental , Oxidative Phosphorylation , Phenotype , Pyrimidines/pharmacology , Receptors, Cell Surface/metabolism , Stilbenes/pharmacology , Syk Kinase/antagonists & inhibitors , Transcriptome
19.
Cancer Cell ; 34(5): 757-774.e7, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30423296

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance and immunotherapeutic resistance. We discovered upregulation of receptor-interacting serine/threonine protein kinase 1 (RIP1) in tumor-associated macrophages (TAMs) in PDA. To study its role in oncogenic progression, we developed a selective small-molecule RIP1 inhibitor with high in vivo exposure. Targeting RIP1 reprogrammed TAMs toward an MHCIIhiTNFα+IFNγ+ immunogenic phenotype in a STAT1-dependent manner. RIP1 inhibition in TAMs resulted in cytotoxic T cell activation and T helper cell differentiation toward a mixed Th1/Th17 phenotype, leading to tumor immunity in mice and in organotypic models of human PDA. Targeting RIP1 synergized with PD1-and inducible co-stimulator-based immunotherapies. Tumor-promoting effects of RIP1 were independent of its co-association with RIP3. Collectively, our work describes RIP1 as a checkpoint kinase governing tumor immunity.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Immune Tolerance/immunology , Macrophages/immunology , Pancreatic Neoplasms/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , Immune Tolerance/genetics , L Cells , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , STAT1 Transcription Factor/metabolism , Th1 Cells/cytology , Th17 Cells/cytology
20.
Cancer Discov ; 8(4): 403-416, 2018 04.
Article in English | MEDLINE | ID: mdl-29567829

ABSTRACT

We found that the cancerous pancreas harbors a markedly more abundant microbiome compared with normal pancreas in both mice and humans, and select bacteria are differentially increased in the tumorous pancreas compared with gut. Ablation of the microbiome protects against preinvasive and invasive pancreatic ductal adenocarcinoma (PDA), whereas transfer of bacteria from PDA-bearing hosts, but not controls, reverses tumor protection. Bacterial ablation was associated with immunogenic reprogramming of the PDA tumor microenvironment, including a reduction in myeloid-derived suppressor cells and an increase in M1 macrophage differentiation, promoting TH1 differentiation of CD4+ T cells and CD8+ T-cell activation. Bacterial ablation also enabled efficacy for checkpoint-targeted immunotherapy by upregulating PD-1 expression. Mechanistically, the PDA microbiome generated a tolerogenic immune program by differentially activating select Toll-like receptors in monocytic cells. These data suggest that endogenous microbiota promote the crippling immune-suppression characteristic of PDA and that the microbiome has potential as a therapeutic target in the modulation of disease progression.Significance: We found that a distinct and abundant microbiome drives suppressive monocytic cellular differentiation in pancreatic cancer via selective Toll-like receptor ligation leading to T-cell anergy. Targeting the microbiome protects against oncogenesis, reverses intratumoral immune tolerance, and enables efficacy for checkpoint-based immunotherapy. These data have implications for understanding immune suppression in pancreatic cancer and its reversal in the clinic. Cancer Discov; 8(4); 403-16. ©2018 AACR.See related commentary by Riquelme et al., p. 386This article is highlighted in the In This Issue feature, p. 371.


Subject(s)
Carcinogenesis , Microbiota , Monocytes/physiology , Pancreatic Neoplasms/microbiology , Toll-Like Receptors/metabolism , Animals , Bacteria , Cell Differentiation , Female , Humans , Male , Mice , Monocytes/immunology , Monocytes/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...