Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell ; 185(11): 1924-1942.e23, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35525247

ABSTRACT

For many solid malignancies, lymph node (LN) involvement represents a harbinger of distant metastatic disease and, therefore, an important prognostic factor. Beyond its utility as a biomarker, whether and how LN metastasis plays an active role in shaping distant metastasis remains an open question. Here, we develop a syngeneic melanoma mouse model of LN metastasis to investigate how tumors spread to LNs and whether LN colonization influences metastasis to distant tissues. We show that an epigenetically instilled tumor-intrinsic interferon response program confers enhanced LN metastatic potential by enabling the evasion of NK cells and promoting LN colonization. LN metastases resist T cell-mediated cytotoxicity, induce antigen-specific regulatory T cells, and generate tumor-specific immune tolerance that subsequently facilitates distant tumor colonization. These effects extend to human cancers and other murine cancer models, implicating a conserved systemic mechanism by which malignancies spread to distant organs.


Subject(s)
Lymph Nodes , Melanoma , Animals , Immune Tolerance , Immunotherapy , Lymphatic Metastasis/pathology , Melanoma/pathology , Mice
2.
Int J Mol Sci ; 22(12)2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34198548

ABSTRACT

Inflammation in the tumor microenvironment has been shown to promote disease progression in pancreatic ductal adenocarcinoma (PDAC); however, the role of macrophage metabolism in promoting inflammation is unclear. Using an orthotopic mouse model of PDAC, we demonstrate that macrophages from tumor-bearing mice exhibit elevated glycolysis. Macrophage-specific deletion of Glucose Transporter 1 (GLUT1) significantly reduced tumor burden, which was accompanied by increased Natural Killer and CD8+ T cell activity and suppression of the NLRP3-IL1ß inflammasome axis. Administration of mice with a GLUT1-specific inhibitor reduced tumor burden, comparable with gemcitabine, the current standard-of-care. In addition, we observe that intra-tumoral macrophages from human PDAC patients exhibit a pronounced glycolytic signature, which reliably predicts poor survival. Our data support a key role for macrophage metabolism in tumor immunity, which could be exploited to improve patient outcomes.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cytoprotection , Glycolysis , Macrophages/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Cell Proliferation/drug effects , Cytoprotection/drug effects , Drug Resistance, Neoplasm/drug effects , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Humans , Hydroxybenzoates/pharmacology , Inflammation/pathology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Macrophages/drug effects , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pancreatic Neoplasms/immunology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/drug effects , Pancreatic Neoplasms
3.
Nat Cancer ; 2(1): 18-33, 2021 01.
Article in English | MEDLINE | ID: mdl-35121890

ABSTRACT

Innate pattern recognition receptor agonists, including Toll-like receptors (TLRs), alter the tumor microenvironment and prime adaptive antitumor immunity. However, TLR agonists present toxicities associated with widespread immune activation after systemic administration. To design a TLR-based therapeutic suitable for systemic delivery and capable of safely eliciting tumor-targeted responses, we developed immune-stimulating antibody conjugates (ISACs) comprising a TLR7/8 dual agonist conjugated to tumor-targeting antibodies. Systemically administered human epidermal growth factor receptor 2 (HER2)-targeted ISACs were well tolerated and triggered a localized immune response in the tumor microenvironment that resulted in tumor clearance and immunological memory. Mechanistically, ISACs required tumor antigen recognition, Fcγ-receptor-dependent phagocytosis and TLR-mediated activation to drive tumor killing by myeloid cells and subsequent T-cell-mediated antitumor immunity. ISAC-mediated immunological memory was not limited to the HER2 ISAC target antigen since ISAC-treated mice were protected from rechallenge with the HER2- parental tumor. These results provide a strong rationale for the clinical development of ISACs.


Subject(s)
Immunotherapy , Neoplasms , Adaptive Immunity , Animals , Antigens, Neoplasm , Immunotherapy/methods , Mice , Neoplasms/drug therapy , Tumor Microenvironment
4.
Front Oncol ; 10: 595892, 2020.
Article in English | MEDLINE | ID: mdl-33282743

ABSTRACT

Enteric glia are a distinct population of peripheral glial cells in the enteric nervous system that regulate intestinal homeostasis, epithelial barrier integrity, and gut defense. Given these unique attributes, we investigated the impact of enteric glia depletion on tumor development in azoxymethane/dextran sodium sulfate (AOM/DSS)-treated mice, a classical model of colorectal cancer (CRC). Depleting GFAP+ enteric glia resulted in a profoundly reduced tumor burden in AOM/DSS mice and additionally reduced adenomas in the ApcMin /+ mouse model of familial adenomatous polyposis, suggesting a tumor-promoting role for these cells at an early premalignant stage. This was confirmed in further studies of AOM/DSS mice, as enteric glia depletion did not affect the properties of established malignant tumors but did result in a marked reduction in the development of precancerous dysplastic lesions. Surprisingly, the protective effect of enteric glia depletion was not dependent on modulation of anti-tumor immunity or intestinal inflammation. These findings reveal that GFAP+ enteric glia play a critical pro-tumorigenic role during early CRC development and identify these cells as a potential target for CRC prevention.

5.
Immunity ; 48(2): 350-363.e7, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29426701

ABSTRACT

Despite evidence that γδ T cells play an important role during malaria, their precise role remains unclear. During murine malaria induced by Plasmodium chabaudi infection and in human P. falciparum infection, we found that γδ T cells expanded rapidly after resolution of acute parasitemia, in contrast to αß T cells that expanded at the acute stage and then declined. Single-cell sequencing showed that TRAV15N-1 (Vδ6.3) γδ T cells were clonally expanded in mice and had convergent complementarity-determining region 3 sequences. These γδ T cells expressed specific cytokines, M-CSF, CCL5, CCL3, which are known to act on myeloid cells, indicating that this γδ T cell subset might have distinct functions. Both γδ T cells and M-CSF were necessary for preventing parasitemic recurrence. These findings point to an M-CSF-producing γδ T cell subset that fulfills a specialized protective role in the later stage of malaria infection when αß T cells have declined.


Subject(s)
Macrophage Colony-Stimulating Factor/physiology , Malaria/prevention & control , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocyte Subsets/immunology , Animals , Female , Humans , Lymphocyte Activation , Malaria/immunology , Mice , Parasitemia/prevention & control , Recurrence
6.
Angew Chem Int Ed Engl ; 57(12): 3137-3142, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29370452

ABSTRACT

The C-type lectins dectin-1 and dectin-2 contribute to innate immunity against microbial pathogens by recognizing their foreign glycan structures. These receptors are promising targets for vaccine development and cancer immunotherapy. However, currently available agonists are heterogeneous glycoconjugates and polysaccharides from natural sources. Herein, we designed and synthesized the first chemically defined ligands for dectin-1 and dectin-2. They comprised glycopolypeptides bearing mono-, di-, and trisaccharides and were built through polymerization of glycosylated N-carboxyanhydrides. Through this approach, we achieved glycopolypeptides with high molecular weights and low dispersities. We identified structures that elicit a pro-inflammatory response through dectin-1 or dectin-2 in antigen-presenting cells. With their native proteinaceous backbones and natural glycosidic linkages, these agonists are attractive for translational applications.


Subject(s)
Anhydrides/metabolism , Antigen-Presenting Cells/metabolism , Glycopeptides/metabolism , Lectins, C-Type/metabolism , Anhydrides/chemistry , Cells, Cultured , Glycopeptides/chemistry , Humans , Lectins, C-Type/chemistry , Ligands , Molecular Structure , Polymerization
7.
Cancer Res ; 77(15): 4158-4170, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28611041

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) after complete surgical resection is often followed by distant metastatic relapse for reasons that remain unclear. In this study, we investigated how the immune response at secondary sites affects tumor spread in murine models of metastatic PDAC. Early metastases were associated with dense networks of CD11b+CD11c+MHC-II+CD24+CD64lowF4/80low dendritic cells (DC), which developed from monocytes in response to tumor-released GM-CSF. These cells uniquely expressed MGL2 and PD-L2 in the metastatic microenvironment and preferentially induced the expansion of T regulatory cells (Treg) in vitro and in vivo Targeted depletion of this DC population in Mgl2DTR hosts activated cytotoxic lymphocytes, reduced Tregs, and inhibited metastasis development. Moreover, blocking PD-L2 selectively activated CD8 T cells at secondary sites and suppressed metastasis, suggesting that the DCs use this particular pathway to inhibit CD8 T-cell-mediated tumor immunity. Phenotypically similar DCs accumulated at primary and secondary sites in other models and in human PDAC. These studies suggest that a discrete DC subset both expands Tregs and suppresses CD8 T cells to establish an immunosuppressive microenvironment conducive to metastasis formation. Therapeutic strategies to block the accumulation and immunosuppressive activity of such cells may help prevent PDAC progression and metastatic relapse after surgical resection. Cancer Res; 77(15); 4158-70. ©2017 AACR.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Dendritic Cells/immunology , Neoplasm Invasiveness/immunology , Pancreatic Neoplasms/pathology , Tumor Escape/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Disease Models, Animal , Flow Cytometry , Mice , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/immunology
8.
Immunity ; 45(3): 641-655, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27590114

ABSTRACT

Although all-trans-retinoic acid (atRA) is a key regulator of intestinal immunity, its role in colorectal cancer (CRC) is unknown. We found that mice with colitis-associated CRC had a marked deficiency in colonic atRA due to alterations in atRA metabolism mediated by microbiota-induced intestinal inflammation. Human ulcerative colitis (UC), UC-associated CRC, and sporadic CRC specimens have similar alterations in atRA metabolic enzymes, consistent with reduced colonic atRA. Inhibition of atRA signaling promoted tumorigenesis, whereas atRA supplementation reduced tumor burden. The benefit of atRA treatment was mediated by cytotoxic CD8(+) T cells, which were activated due to MHCI upregulation on tumor cells. Consistent with these findings, increased colonic expression of the atRA-catabolizing enzyme, CYP26A1, correlated with reduced frequencies of tumoral cytotoxic CD8(+) T cells and with worse disease prognosis in human CRC. These results reveal a mechanism by which microbiota drive colon carcinogenesis and highlight atRA metabolism as a therapeutic target for CRC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Microbiota/immunology , Tretinoin/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/immunology , Colon/immunology , Colon/metabolism , Colorectal Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Retinoic Acid 4-Hydroxylase/metabolism , Signal Transduction/immunology , Up-Regulation/immunology
9.
Cancer Immunol Res ; 4(11): 917-926, 2016 11.
Article in English | MEDLINE | ID: mdl-27638841

ABSTRACT

Chronic intestinal inflammation accompanies familial adenomatous polyposis (FAP) and is a major risk factor for colorectal cancer in patients with this disease, but the cause of such inflammation is unknown. Because retinoic acid (RA) plays a critical role in maintaining immune homeostasis in the intestine, we hypothesized that altered RA metabolism contributes to inflammation and tumorigenesis in FAP. To assess this hypothesis, we analyzed RA metabolism in the intestines of patients with FAP as well as APCMin/+ mice, a model that recapitulates FAP in most respects. We also investigated the impact of intestinal RA repletion and depletion on tumorigenesis and inflammation in APCMin/+ mice. Tumors from both FAP patients and APCMin/+ mice displayed striking alterations in RA metabolism that resulted in reduced intestinal RA. APCMin/+ mice placed on a vitamin A-deficient diet exhibited further reductions in intestinal RA with concomitant increases in inflammation and tumor burden. Conversely, restoration of RA by pharmacologic blockade of the RA-catabolizing enzyme CYP26A1 attenuated inflammation and diminished tumor burden. To investigate the effect of RA deficiency on the gut immune system, we studied lamina propria dendritic cells (LPDC) because these cells play a central role in promoting tolerance. APCMin/+ LPDCs preferentially induced Th17 cells, but reverted to inducing Tregs following restoration of intestinal RA in vivo or direct treatment of LPDCs with RA in vitro These findings demonstrate the importance of intestinal RA deficiency in tumorigenesis and suggest that pharmacologic repletion of RA could reduce tumorigenesis in FAP patients. Cancer Immunol Res; 4(11); 917-26. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Enterocolitis/genetics , Genes, APC , Tretinoin/pharmacology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Adenomatous Polyposis Coli/complications , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enterocolitis/drug therapy , Enterocolitis/metabolism , Enterocolitis/pathology , Humans , Mice , Phenotype , Th17 Cells/immunology , Th17 Cells/metabolism , Tretinoin/metabolism , Tumor Burden , Vitamin A/metabolism , Vitamin A Deficiency/metabolism
10.
Nature ; 521(7550): 99-104, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25924063

ABSTRACT

Whereas cancers grow within host tissues and evade host immunity through immune-editing and immunosuppression, tumours are rarely transmissible between individuals. Much like transplanted allogeneic organs, allogeneic tumours are reliably rejected by host T cells, even when the tumour and host share the same major histocompatibility complex alleles, the most potent determinants of transplant rejection. How such tumour-eradicating immunity is initiated remains unknown, although elucidating this process could provide the basis for inducing similar responses against naturally arising tumours. Here we find that allogeneic tumour rejection is initiated in mice by naturally occurring tumour-binding IgG antibodies, which enable dendritic cells (DCs) to internalize tumour antigens and subsequently activate tumour-reactive T cells. We exploited this mechanism to treat autologous and autochthonous tumours successfully. Either systemic administration of DCs loaded with allogeneic-IgG-coated tumour cells or intratumoral injection of allogeneic IgG in combination with DC stimuli induced potent T-cell-mediated antitumour immune responses, resulting in tumour eradication in mouse models of melanoma, pancreas, lung and breast cancer. Moreover, this strategy led to eradication of distant tumours and metastases, as well as the injected primary tumours. To assess the clinical relevance of these findings, we studied antibodies and cells from patients with lung cancer. T cells from these patients responded vigorously to autologous tumour antigens after culture with allogeneic-IgG-loaded DCs, recapitulating our findings in mice. These results reveal that tumour-binding allogeneic IgG can induce powerful antitumour immunity that can be exploited for cancer immunotherapy.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Immunoglobulin G/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Neoplasm/administration & dosage , CD40 Antigens/metabolism , Disease Models, Animal , Female , Immunoglobulin G/administration & dosage , Isoantibodies/administration & dosage , Isoantibodies/immunology , Lymphocyte Activation/immunology , Male , Mice , Neoplasm Metastasis , Neoplasm Transplantation/immunology , Neoplasms/pathology , Receptors, IgG/immunology , Tumor Necrosis Factor-alpha/immunology
11.
PLoS One ; 8(10): e76258, 2013.
Article in English | MEDLINE | ID: mdl-24098455

ABSTRACT

Two critical functions of dendritic cells (DC) are to activate and functionally polarize T cells. Activated T cells can, in turn, influence DC maturation, although their effect on de novo DC development is poorly understood. Here we report that activation of T cells in mice, with either an anti-CD3 antibody or super antigen, drives the rapid formation of CD209(+)CD11b(+)CD11c(+) MHC II(+) DC from monocytic precursors (Mo-DC). GM-CSF is produced by T cells following activation, but surprisingly, it is not required for the formation of CD209(+) Mo-DC. CD40L, however, is critical for the full induction of Mo-DC following T cell activation. T cell induced CD209(+) Mo-DC are comparable to conventional CD209(-) DC in their ability to stimulate T cell proliferation. However, in contrast to conventional CD209(-) DC, CD209(+) Mo-DC fail to effectively polarize T cells, as indicated by a paucity of T cell cytokine production. The inability of CD209(+) Mo-DC to polarize T cells is partly explained by increased expression of PDL-2, since blockade of this molecule restores some polarizing capacity to the Mo-DC. These findings expand the range of signals capable of driving Mo-DC differentiation in vivo beyond exogenous microbial factors to include endogenous factors produced following T cell activation.


Subject(s)
Cell Adhesion Molecules/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocyte Subsets/immunology , Animals , CD40 Ligand/metabolism , Cell Differentiation/immunology , Cell Movement , Dendritic Cells/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lymph Nodes/immunology , Mice , Mice, Knockout , Monocytes/immunology , T-Lymphocyte Subsets/metabolism
12.
J Immunol ; 191(3): 1175-87, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23794631

ABSTRACT

In chronically inflamed tissues, such as those affected by autoimmune disease, activated Th cells often colocalize with monocytes. We investigate in this study how murine Th cells influence the phenotype and function of monocytes. The data demonstrate that Th1, Th2, and Th17 subsets promote the differentiation of autologous monocytes into MHC class II(+), CD11b(+), CD11c(+) DC that we call DCTh. Although all Th subsets induce the formation of DCTh, activated Th17 cells uniquely promote the formation of IL-12/IL-23-producing DCTh (DCTh17) that can polarize both naive and Th17 cells to a Th1 phenotype. In the inflamed CNS of mice with Th17-mediated experimental autoimmune encephalomyelitis, Th cells colocalize with DC, as well as monocytes, and the Th cells obtained from these lesions drive the formation of DCTh that are phenotypically indistinguishable from DCTh17 and polarize naive T cells toward a Th1 phenotype. These results suggest that DCTh17 are critical in the interplay of Th17- and Th1-mediated responses and may explain the previous finding that IL-17-secreting Th cells become IFN-γ-secreting Th1 cells in experimental autoimmune encephalomyelitis and other autoimmune disorders.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , Inflammation/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cell Differentiation , Cell Movement , Cell Polarity , Cells, Cultured , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/metabolism , Interleukin-12/biosynthesis , Interleukin-17/metabolism , Interleukin-23/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology
13.
Nat Med ; 17(5): 610-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21499269

ABSTRACT

Chronic inflammation characterized by T cell and macrophage infiltration of visceral adipose tissue (VAT) is a hallmark of obesity-associated insulin resistance and glucose intolerance. Here we show a fundamental pathogenic role for B cells in the development of these metabolic abnormalities. B cells accumulate in VAT in diet-induced obese (DIO) mice, and DIO mice lacking B cells are protected from disease despite weight gain. B cell effects on glucose metabolism are mechanistically linked to the activation of proinflammatory macrophages and T cells and to the production of pathogenic IgG antibodies. Treatment with a B cell-depleting CD20 antibody attenuates disease, whereas transfer of IgG from DIO mice rapidly induces insulin resistance and glucose intolerance. Moreover, insulin resistance in obese humans is associated with a unique profile of IgG autoantibodies. These results establish the importance of B cells and adaptive immunity in insulin resistance and suggest new diagnostic and therapeutic modalities for managing the disease.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin G/biosynthesis , Insulin Resistance/immunology , T-Lymphocytes/immunology , Animals , Autoantibodies/biosynthesis , Autoantigens/immunology , Autoimmunity , Dietary Fats/adverse effects , Glucose/metabolism , Humans , Immunoglobulin mu-Chains/genetics , Inflammation/immunology , Intra-Abdominal Fat/immunology , Lymphocyte Activation , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/immunology , Obesity/metabolism
14.
Clin Cancer Res ; 16(14): 3684-95, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20534740

ABSTRACT

PURPOSE: The most common preclinical models of pancreatic adenocarcinoma utilize human cells or tissues that are xenografted into immunodeficient hosts. Several immunocompetent, genetically engineered mouse models of pancreatic cancer exist; however, tumor latency and disease progression in these models are highly variable. We sought to develop an immunocompetent, orthotopic mouse model of pancreatic cancer with rapid and predictable growth kinetics. EXPERIMENTAL DESIGN: Cell lines with epithelial morphology were derived from liver metastases obtained from Kras(G12D/+);LSL-Trp53(R172H/+);Pdx-1-Cre mice. Tumor cells were implanted in the pancreas of immunocompetent, histocompatible B6/129 mice, and the mice were monitored for disease progression. Relevant tissues were harvested for histologic, genomic, and immunophenotypic analysis. RESULTS: All mice developed pancreatic tumors by two weeks. Invasive disease and liver metastases were noted by six to eight weeks. Histologic examination of tumors showed cytokeratin-19-positive adenocarcinoma with regions of desmoplasia. Genomic analysis revealed broad chromosomal changes along with focal gains and losses. Pancreatic tumors were infiltrated with dendritic cells, myeloid-derived suppressor cells, macrophages, and T lymphocytes. Survival was decreased in RAG(-/-) mice, which are deficient in T cells, suggesting that an adaptive immune response alters the course of disease in wild-type mice. CONCLUSIONS: We have developed a rapid, predictable orthotopic model of pancreatic adenocarcinoma in immunocompetent mice that mimics human pancreatic cancer with regard to genetic mutations, histologic appearance, and pattern of disease progression. This model highlights both the complexity and relevance of the immune response to invasive pancreatic cancer and may be useful for the preclinical evaluation of new therapeutic agents.


Subject(s)
Adenocarcinoma/immunology , Disease Models, Animal , Immunocompetence , Pancreatic Neoplasms/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Disease Progression , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...