151
|
Abstract
B cell receptor (BCR) signalling is crucial for normal B cell development and adaptive immunity. BCR signalling also supports the survival and growth of malignant B cells in patients with B cell leukaemias or lymphomas. The mechanism of BCR pathway activation in these diseases includes continuous BCR stimulation by microbial antigens or autoantigens present in the tissue microenvironment, activating mutations within the BCR complex or downstream signalling components and ligand-independent tonic BCR signalling. The most established agents targeting BCR signalling are Bruton tyrosine kinase (BTK) inhibitors and PI3K isoform-specific inhibitors, and their introduction into the clinic is rapidly changing how B cell malignancies are treated. B cells and BCR-related kinases, such as BTK, also play a role in the microenvironment of solid tumours, such as squamous cell carcinoma and pancreatic cancer, and therefore targeting B cells or BCR-related kinases may have anticancer activity beyond B cell malignancies.
Collapse
MESH Headings
- Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors
- Antineoplastic Agents/pharmacology
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Drug Resistance, Neoplasm
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Lymphoma, Mantle-Cell/drug therapy
- Lymphoma, Mantle-Cell/metabolism
- Molecular Targeted Therapy/methods
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptors, Antigen, B-Cell/antagonists & inhibitors
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction/drug effects
- Tumor Microenvironment
Collapse
Affiliation(s)
- Jan A Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Adrian Wiestner
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
152
|
Hessmann E, Patzak MS, Klein L, Chen N, Kari V, Ramu I, Bapiro TE, Frese KK, Gopinathan A, Richards FM, Jodrell DI, Verbeke C, Li X, Heuchel R, Löhr JM, Johnsen SA, Gress TM, Ellenrieder V, Neesse A. Fibroblast drug scavenging increases intratumoural gemcitabine accumulation in murine pancreas cancer. Gut 2018; 67:497-507. [PMID: 28077438 PMCID: PMC5868285 DOI: 10.1136/gutjnl-2016-311954] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 11/01/2016] [Accepted: 11/23/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Desmoplasia and hypovascularity are thought to impede drug delivery in pancreatic ductal adenocarcinoma (PDAC). However, stromal depletion approaches have failed to show clinical responses in patients. Here, we aimed to revisit the role of the tumour microenvironment as a physical barrier for gemcitabine delivery. DESIGN Gemcitabine metabolites were analysed in LSL-KrasG12D/+ ; LSL-Trp53R172H/+ ; Pdx-1-Cre (KPC) murine tumours and matched liver metastases, primary tumour cell lines, cancer-associated fibroblasts (CAFs) and pancreatic stellate cells (PSCs) by liquid chromatography-mass spectrometry/mass spectrometry. Functional and preclinical experiments, as well as expression analysis of stromal markers and gemcitabine metabolism pathways were performed in murine and human specimen to investigate the preclinical implications and the mechanism of gemcitabine accumulation. RESULTS Gemcitabine accumulation was significantly enhanced in fibroblast-rich tumours compared with liver metastases and normal liver. In vitro, significantly increased concentrations of activated 2',2'-difluorodeoxycytidine-5'-triphosphate (dFdCTP) and greatly reduced amounts of the inactive gemcitabine metabolite 2',2'-difluorodeoxyuridine were detected in PSCs and CAFs. Mechanistically, key metabolic enzymes involved in gemcitabine inactivation such as hydrolytic cytosolic 5'-nucleotidases (Nt5c1A, Nt5c3) were expressed at low levels in CAFs in vitro and in vivo, and recombinant expression of Nt5c1A resulted in decreased intracellular dFdCTP concentrations in vitro. Moreover, gemcitabine treatment in KPC mice reduced the number of liver metastases by >50%. CONCLUSIONS Our findings suggest that fibroblast drug scavenging may contribute to the clinical failure of gemcitabine in desmoplastic PDAC. Metabolic targeting of CAFs may thus be a promising strategy to enhance the antiproliferative effects of gemcitabine.
Collapse
Affiliation(s)
- E Hessmann
- Department Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Goettingen, Germany
| | - M S Patzak
- Department Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Goettingen, Germany
| | - L Klein
- Department Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Goettingen, Germany
| | - N Chen
- Department Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Goettingen, Germany
| | - V Kari
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Goettingen, Germany
| | - I Ramu
- Department Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Goettingen, Germany
| | - T E Bapiro
- Cancer Research UK Cambridge Institute, The University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Oncology iMED DMPK AstraZeneca UK Ltd, HODGKIN C/o B310 Cambridge Science Park, Cambridge, UK
| | - K K Frese
- The University of Manchester, Cancer Research UK Manchester Institute, Manchester, UK
| | - A Gopinathan
- Cancer Research UK Cambridge Institute, The University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - F M Richards
- Cancer Research UK Cambridge Institute, The University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - D I Jodrell
- Cancer Research UK Cambridge Institute, The University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - C Verbeke
- Department of Pathology, Karolinska University Hospital, Stockholm, Sweden
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - X Li
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - R Heuchel
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - J M Löhr
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - S A Johnsen
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Goettingen, Germany
| | - T M Gress
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps University Marburg, Marburg, Germany
| | - V Ellenrieder
- Department Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Goettingen, Germany
| | - A Neesse
- Department Gastroenterology and Gastrointestinal Oncology, University Medical Centre Goettingen, Goettingen, Germany
| |
Collapse
|
153
|
Abstract
The tumor microenvironment (TME) is defined as the structural and dynamic network of cellular and non-cellular interactions between malignant cells and the surrounding non-malignant matrix. Hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC) are two of the most challenging gastrointestinal malignancies. Despite clinical advancements in understanding tumor biology and growth of the chemotherapeutic industry, there have been no corresponding improvements in prognosis and overall survival of HCC and PDAC. Both of these cancers have a very intimate relationship with their surrounding environment; the TME is thought to actively participate in initiating and sustaining these malignancies. Individual TME constituents play a vital role in chemoresistance and recurrence after surgery and have been established as independent prognostic factors. This review article will highlight the diverse structural components, key signaling pathways, and extracellular matrices of HCC and PDAC and discuss their crosstalk with tumor cells to promote growth and metastasis. The article will also summarize the latest laboratory and clinical research based on therapeutic targets identified within the TME of both HCC and PDAC.
Collapse
Affiliation(s)
- Fathima Kamil
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Julie H Rowe
- Division of Oncology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
154
|
Tokarz P, Wiśniewska M, Kamiński MM, Dubin G, Grudnik P. Crystal structure of ADP-dependent glucokinase from Methanocaldococcus jannaschii in complex with 5-iodotubercidin reveals phosphoryl transfer mechanism. Protein Sci 2018; 27:790-797. [PMID: 29352744 DOI: 10.1002/pro.3377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 11/11/2022]
Abstract
ADP-dependent glucokinase (ADPGK) is an alternative novel glucose phosphorylating enzyme in a modified glycolysis pathway of hyperthermophilic Archaea. In contrast to classical ATP-dependent hexokinases, ADPGK utilizes ADP as a phosphoryl group donor. Here, we present a crystal structure of archaeal ADPGK from Methanocaldococcus jannaschii in complex with an inhibitor, 5-iodotubercidin, d-glucose, inorganic phosphate, and a magnesium ion. Detailed analysis of the architecture of the active site allowed for confirmation of the previously proposed phosphorylation mechanism and the crucial role of the invariant arginine residue (Arg197). The crystal structure shows how the phosphate ion, while mimicking a β-phosphate group, is positioned in the proximity of the glucose moiety by arginine and the magnesium ion, thus providing novel insights into the mechanism of catalysis. In addition, we demonstrate that 5-iodotubercidin inhibits human ADPGK-dependent T cell activation-induced reactive oxygen species (ROS) release and downstream gene expression, and as such it may serve as a model compound for further screening for hADPGK-specific inhibitors.
Collapse
Affiliation(s)
- Piotr Tokarz
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7, Krakow, 30-387, Poland.,Malopolska Center of Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7a, Krakow, 30-387, Poland
| | - Magdalena Wiśniewska
- Malopolska Center of Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7a, Krakow, 30-387, Poland
| | - Marcin M Kamiński
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee, 38105
| | - Grzegorz Dubin
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7, Krakow, 30-387, Poland.,Malopolska Center of Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7a, Krakow, 30-387, Poland
| | - Przemysław Grudnik
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7, Krakow, 30-387, Poland.,Malopolska Center of Biotechnology, Jagiellonian University in Krakow, ul. Gronostajowa 7a, Krakow, 30-387, Poland
| |
Collapse
|
155
|
Liudahl SM, Coussens LM. B cells as biomarkers: predicting immune checkpoint therapy adverse events. J Clin Invest 2018; 128:577-579. [PMID: 29309049 DOI: 10.1172/jci99036] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Immune checkpoint inhibitors are becoming a cornerstone of cancer immunotherapy as a result of their clinical success in relieving immune suppression and driving durable antitumor T cell responses in certain subsets of patients. Unfortunately, checkpoint inhibition is also associated with treatment-related toxicities that result in a myriad of side effects, ranging from mild and manageable to severe and debilitating. In this issue of the JCI, Das and colleagues report an association between early therapy-induced changes in circulating B cells and an increased risk of high-grade immune-related adverse events (IRAEs) in patients treated with checkpoint inhibitors that target cytotoxic T lymphocyte-associated antigen-4 (CTLA4) and programmed cell death protein 1 (PD1). These findings identify potential predictive biomarkers for high-grade IRAEs that may be leveraged to improve patient monitoring and may prompt new treatment strategies to prevent IRAEs.
Collapse
|
156
|
Prendergast GC, Malachowski WP, DuHadaway JB, Muller AJ. Discovery of IDO1 Inhibitors: From Bench to Bedside. Cancer Res 2018; 77:6795-6811. [PMID: 29247038 DOI: 10.1158/0008-5472.can-17-2285] [Citation(s) in RCA: 412] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/23/2017] [Accepted: 11/01/2017] [Indexed: 01/11/2023]
Abstract
Small-molecule inhibitors of indoleamine 2,3-dioxygenase-1 (IDO1) are emerging at the vanguard of experimental agents in oncology. Here, pioneers of this new drug class provide a bench-to-bedside review on preclinical validation of IDO1 as a cancer therapeutic target and on the discovery and development of a set of mechanistically distinct compounds, indoximod, epacadostat, and navoximod, that were first to be evaluated as IDO inhibitors in clinical trials. As immunometabolic adjuvants to widen therapeutic windows, IDO inhibitors may leverage not only immuno-oncology modalities but also chemotherapy and radiotherapy as standards of care in the oncology clinic. Cancer Res; 77(24); 6795-811. ©2017 AACR.
Collapse
Affiliation(s)
| | | | - James B DuHadaway
- Lankenau Institute for Medical Research (LIMR), Wynnewood, Pennsylvania
| | | |
Collapse
|
157
|
Abstract
Hypoxia (insufficient O2 availability) is involved in various biological processes, such as tumorigenesis and inflammation. Hypoxia results in stabilization of hypoxia-inducible factors (HIFs) including HIF1α and HIF2α. Here we describe a protocol to detect mouse and human tissue hypoxia by using Hypoxyprobe and immunohistochemical staining for HIF1α and HIF2α.
Collapse
Affiliation(s)
- Fuming Li
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kyoung Eun Lee
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA.
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
158
|
|
159
|
Inflammatory Cytokine Signaling during Development of Pancreatic and Prostate Cancers. J Immunol Res 2017; 2017:7979637. [PMID: 29379802 PMCID: PMC5742898 DOI: 10.1155/2017/7979637] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/31/2017] [Accepted: 11/08/2017] [Indexed: 02/07/2023] Open
Abstract
Inflammation is essential for many diseases including cancer. Activation and recruitment of immune cells during inflammation result in a cytokine- and chemokine-enriched cell environment, which affects cancer development. Since each type of cancer has its unique tumor environment, effects of cytokines from different sources such as tumor-infiltrating immune cells, stromal cells, endothelial cells, and cancer cells on cancer development can be quite complex. In this review, how immune cells contribute to tumorigenesis of pancreatic and prostate cancers through their secreted cytokines is discussed. In addition, the cytokine signaling that tumor cells of pancreatic and prostate cancers utilize to benefit their own survival is delineated.
Collapse
|
160
|
Boyle KA, Dwinell MB. GEMMs Are a Gem When it Comes to Defining the Role of HIF2α in Mucinous Cystic Neoplasms. Cell Mol Gastroenterol Hepatol 2017; 5:165-166. [PMID: 29693045 PMCID: PMC5904046 DOI: 10.1016/j.jcmgh.2017.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
| | - Michael B. Dwinell
- Correspondence Address correspondence to: Michael B. Dwinell, PhD, Department of Microbiology & Immunology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226.
| |
Collapse
|
161
|
Qian BZ. Inflammation fires up cancer metastasis. Semin Cancer Biol 2017; 47:170-176. [PMID: 28838845 DOI: 10.1016/j.semcancer.2017.08.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/16/2022]
Abstract
Metastatic disease is the major challenge of cancer that accounts for over 90% of total cancer lethality. Mounting clinical and preclinical data now indicate that inflammation, a potent immune and repair response, is indispensable for metastasis. In this review we describe our current understanding of how major inflammatory cells contribute to metastatic cascade with a focus on the primary tumour. We also discuss exciting new directions for future research and novel therapeutic approaches to tackle metastatic disease through targeting inflammation.
Collapse
Affiliation(s)
- Bin-Zhi Qian
- University of Edinburgh and MRC Centre for Reproductive Health, EH16 4TJ, Edinburgh, United Kingdom; Edinburgh Cancer Research UK Centre, EH16 4TJ, Edinburgh, United Kingdom.
| |
Collapse
|
162
|
Schofield HK, Tandon M, Park MJ, Halbrook CJ, Ramakrishnan SK, Kim EC, Shi J, Omary MB, Shah YM, Esni F, Pasca di Magliano M. Pancreatic HIF2α Stabilization Leads to Chronic Pancreatitis and Predisposes to Mucinous Cystic Neoplasm. Cell Mol Gastroenterol Hepatol 2017; 5:169-185.e2. [PMID: 29693047 PMCID: PMC5904051 DOI: 10.1016/j.jcmgh.2017.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Tissue hypoxia controls cell differentiation in the embryonic pancreas, and promotes tumor growth in pancreatic cancer. The cellular response to hypoxia is controlled by the hypoxia-inducible factor (HIF) proteins, including HIF2α. Previous studies of HIF action in the pancreas have relied on loss-of-function mouse models, and the effects of HIF2α expression in the pancreas have remained undefined. METHODS We developed several transgenic mouse models based on the expression of an oxygen-stable form of HIF2α, or indirect stabilization of HIF proteins though deletion of von Hippel-Lindau, thus preventing HIF degradation. Furthermore, we crossed both sets of animals into mice expressing oncogenic KrasG12D in the pancreas. RESULTS We show that HIF2α is not expressed in the normal human pancreas, however, it is up-regulated in human chronic pancreatitis. Deletion of von Hippel-Lindau or stabilization of HIF2α in mouse pancreata led to the development of chronic pancreatitis. Importantly, pancreatic HIF1α stabilization did not disrupt the pancreatic parenchyma, indicating that the chronic pancreatitis phenotype is specific to HIF2α. In the presence of oncogenic Kras, HIF2α stabilization drove the formation of cysts resembling mucinous cystic neoplasm (MCN) in humans. Mechanistically, we show that the pancreatitis phenotype is linked to expression of multiple inflammatory cytokines and activation of the unfolded protein response. Conversely, MCN formation is linked to activation of Wnt signaling, a feature of human MCN. CONCLUSIONS We show that pancreatic HIF2α stabilization disrupts pancreatic homeostasis, leading to chronic pancreatitis, and, in the context of oncogenic Kras, MCN formation. These findings provide new mouse models of both chronic pancreatitis and MCN, as well as illustrate the importance of hypoxia signaling in the pancreas.
Collapse
Affiliation(s)
- Heather K. Schofield
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan
| | - Manuj Tandon
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Min-Jung Park
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Christopher J. Halbrook
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Sadeesh K. Ramakrishnan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Esther C. Kim
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Jiaqi Shi
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - M. Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Yatrik M. Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Farzad Esni
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
- Department of Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
163
|
Stromnes IM, Hulbert A, Pierce RH, Greenberg PD, Hingorani SR. T-cell Localization, Activation, and Clonal Expansion in Human Pancreatic Ductal Adenocarcinoma. Cancer Immunol Res 2017; 5:978-991. [PMID: 29066497 PMCID: PMC5802342 DOI: 10.1158/2326-6066.cir-16-0322] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 07/21/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal malignancy resistant to most therapies, including immune checkpoint blockade. To elucidate mechanisms of immunotherapy resistance, we assessed immune parameters in resected human PDA. We demonstrate significant interpatient variability in T-cell number, localization, and phenotype. CD8+ T cells, Foxp3+ regulatory T cells, and PD-1+ and PD-L1+ cells were preferentially enriched in tertiary lymphoid structures that were found in most tumors compared with stroma and tumor cell nests. Tumors containing more CD8+ T cells also had increased granulocytes, CD163+ (M2 immunosuppressive phenotype) macrophages, and FOXP3+ regulatory T cells. PD-L1 was rare on tumor cells, but was expressed by CD163+ macrophages and an additional stromal cell subset commonly found clustered together adjacent to tumor epithelium. The majority of tumoral CD8+ T cells did not express molecules suggestive of recent T-cell receptor (TCR) signaling. However, 41BB+PD-1+ T cells were still significantly enriched in tumors compared with circulation. Tumoral CD8+PD-1+ T cells commonly expressed additional inhibitory receptors, yet were mostly T-BEThi and EOMESlo, consistent with a less terminally exhausted state. Analysis of gene expression and rearranged TCR genes by deep sequencing suggested most patients have a limited tumor-reactive T-cell response. Multiplex immunohistochemistry revealed variable T-cell infiltration based on abundance and location, which may result in different mechanisms of immunotherapy resistance. Overall, the data support the need for therapies that either induce endogenous, or provide engineered, tumor-specific T-cell responses, and concurrently relieve suppressive mechanisms operative at the tumor site. Cancer Immunol Res; 5(11); 978-91. ©2017 AACR.
Collapse
Affiliation(s)
- Ingunn M Stromnes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ayaka Hulbert
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Robert H Pierce
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Philip D Greenberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
- Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Division of Medical Oncology, University of Washington School of Medicine, Seattle, Washington
| | - Sunil R Hingorani
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
- Division of Medical Oncology, University of Washington School of Medicine, Seattle, Washington
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
164
|
Hypoxia inducible factor (HIF) in the tumor microenvironment: friend or foe? SCIENCE CHINA-LIFE SCIENCES 2017; 60:1114-1124. [PMID: 29039125 DOI: 10.1007/s11427-017-9178-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/18/2017] [Indexed: 02/06/2023]
Abstract
Hypoxia acts as an important regulator of physiological and pathological processes. Hypoxia inducible factors (HIFs) are the central players involved in the cellular adaptation to hypoxia and are regulated by oxygen sensing EGLN prolyl hydroxylases. Hypoxia affects many aspects of cellular growth through both redox effects and through the stabilization of HIFs. The HIF isoforms likely have differential effects on tumor growth via alteration of metabolism, growth, and self-renewal and are likely highly context-dependent. In some tumors such as renal cell carcinoma, the EGLN/HIF axis appears to drive tumorigenesis, while in many others HIF1 and HIF2 may actually have a tumor suppressive role. An emerging role of HIF biology is its effects on the tumor microenvironment. The EGLN/HIF axis plays a key role in regulating the function of the various components of the tumor microenvironment, which include cancer-associated fibroblasts, endothelial cells, immune cells, and the extracellular matrix (ECM). Here, we discuss hypoxia and the diverse roles of HIFs in the setting of tumorigenesis and the maintenance of the tumor microenvironment as well as possible future directions of the field.
Collapse
|
165
|
Seiller C, Dubois B. [Infiltrated B cells promote pancreatic ductal adenocarcinoma]. Med Sci (Paris) 2017; 33:859-862. [PMID: 28994380 DOI: 10.1051/medsci/20173310015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Carolane Seiller
- Master de cancérologie, module d'immunologie-virologie, université de Lyon, université Claude Bernard Lyon 1, Lyon, France
| | - Bertrand Dubois
- Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS 5286, centre Léon Bérard, centre de recherche en cancérologie de Lyon, 69008 Lyon, France
| |
Collapse
|
166
|
Somasundaram R, Zhang G, Fukunaga-Kalabis M, Perego M, Krepler C, Xu X, Wagner C, Hristova D, Zhang J, Tian T, Wei Z, Liu Q, Garg K, Griss J, Hards R, Maurer M, Hafner C, Mayerhöfer M, Karanikas G, Jalili A, Bauer-Pohl V, Weihsengruber F, Rappersberger K, Koller J, Lang R, Hudgens C, Chen G, Tetzlaff M, Wu L, Frederick DT, Scolyer RA, Long GV, Damle M, Ellingsworth C, Grinman L, Choi H, Gavin BJ, Dunagin M, Raj A, Scholler N, Gross L, Beqiri M, Bennett K, Watson I, Schaider H, Davies MA, Wargo J, Czerniecki BJ, Schuchter L, Herlyn D, Flaherty K, Herlyn M, Wagner SN. Tumor-associated B-cells induce tumor heterogeneity and therapy resistance. Nat Commun 2017; 8:607. [PMID: 28928360 PMCID: PMC5605714 DOI: 10.1038/s41467-017-00452-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/30/2017] [Indexed: 01/19/2023] Open
Abstract
In melanoma, therapies with inhibitors to oncogenic BRAFV600E are highly effective but responses are often short-lived due to the emergence of drug-resistant tumor subpopulations. We describe here a mechanism of acquired drug resistance through the tumor microenvironment, which is mediated by human tumor-associated B cells. Human melanoma cells constitutively produce the growth factor FGF-2, which activates tumor-infiltrating B cells to produce the growth factor IGF-1. B-cell-derived IGF-1 is critical for resistance of melanomas to BRAF and MEK inhibitors due to emergence of heterogeneous subpopulations and activation of FGFR-3. Consistently, resistance of melanomas to BRAF and/or MEK inhibitors is associated with increased CD20 and IGF-1 transcript levels in tumors and IGF-1 expression in tumor-associated B cells. Furthermore, first clinical data from a pilot trial in therapy-resistant metastatic melanoma patients show anti-tumor activity through B-cell depletion by anti-CD20 antibody. Our findings establish a mechanism of acquired therapy resistance through tumor-associated B cells with important clinical implications.Resistance to BRAFV600E inhibitors often occurs in melanoma patients. Here, the authors describe a potential mechanism of acquired drug resistance mediated by tumor-associated B cells-derived IGF-1.
Collapse
Affiliation(s)
| | - Gao Zhang
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | | | | | | | - Xiaowei Xu
- Department of Pathology and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Christine Wagner
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, A-1090, Austria
| | | | - Jie Zhang
- New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Tian Tian
- New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zhi Wei
- New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Qin Liu
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Kanika Garg
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, A-1090, Austria
| | - Johannes Griss
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, A-1090, Austria
| | - Rufus Hards
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Margarita Maurer
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, A-1090, Austria
| | - Christine Hafner
- Department of Dermatology and Venereology, Karl Landsteiner University of Health Sciences, St. Pölten, A-3100, Austria
| | - Marius Mayerhöfer
- Department of Radiology, Division of Nuclear Medicine, Medical University of Vienna, Vienna, A-1090, Austria
| | - Georgios Karanikas
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, A-1090, Austria
| | - Ahmad Jalili
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, A-1090, Austria
| | - Verena Bauer-Pohl
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, A-1090, Austria
| | - Felix Weihsengruber
- Department of Dermatology and Venereology, The Rudolfstiftung Hospital, Teaching Hospital of the Medical University Vienna, Vienna, A-1030, Austria
| | - Klemens Rappersberger
- Department of Dermatology and Venereology, The Rudolfstiftung Hospital, Teaching Hospital of the Medical University Vienna, Vienna, A-1030, Austria
| | - Josef Koller
- Department of Dermatology, Paracelsus Medical University Salzburg, Salzburg, A-5020, Austria
| | - Roland Lang
- Department of Dermatology, Paracelsus Medical University Salzburg, Salzburg, A-5020, Austria
| | - Courtney Hudgens
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77040, USA
| | - Guo Chen
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77040, USA
| | - Michael Tetzlaff
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77040, USA
| | - Lawrence Wu
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | | | - Richard A Scolyer
- Melanoma Institute of Australia, and The University of Sydney, Sydney, 2065, Australia
| | - Georgina V Long
- Melanoma Institute of Australia, and The University of Sydney, Sydney, 2065, Australia
| | | | | | - Leon Grinman
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Harry Choi
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | | | - Margaret Dunagin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Arjun Raj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nathalie Scholler
- Abramson Cancer Center, Hospital of University of Pennsylvania, Philadelphia, PA, 19104, USA
- SRI International, Menlo Park, CA, 94025, USA
| | - Laura Gross
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | | | - Keiryn Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, A-1090, Austria
| | - Ian Watson
- Department of Biochemistry, McGill University, Montreal, QC, Canada, H3A0G4
| | - Helmut Schaider
- Dermatology Research Center, University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, 4102, Australia
| | - Michael A Davies
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77040, USA
| | - Jennifer Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer, Center, Houston, TX, 77040, USA
| | - Brian J Czerniecki
- Abramson Cancer Center, Hospital of University of Pennsylvania, Philadelphia, PA, 19104, USA
- Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Lynn Schuchter
- Abramson Cancer Center, Hospital of University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Keith Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Stephan N Wagner
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, A-1090, Austria.
| |
Collapse
|
167
|
Zhang Z, Zhu Y, Wang Z, Zhang T, Wu P, Huang J. Yin-yang effect of tumor infiltrating B cells in breast cancer: From mechanism to immunotherapy. Cancer Lett 2017; 393:1-7. [DOI: 10.1016/j.canlet.2017.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 01/11/2023]
|
168
|
Anderson KG, Stromnes IM, Greenberg PD. Obstacles Posed by the Tumor Microenvironment to T cell Activity: A Case for Synergistic Therapies. Cancer Cell 2017; 31:311-325. [PMID: 28292435 PMCID: PMC5423788 DOI: 10.1016/j.ccell.2017.02.008] [Citation(s) in RCA: 502] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/13/2022]
Abstract
T cell dysfunction in solid tumors results from multiple mechanisms. Altered signaling pathways in tumor cells help produce a suppressive tumor microenvironment enriched for inhibitory cells, posing a major obstacle for cancer immunity. Metabolic constraints to cell function and survival shape tumor progression and immune cell function. In the face of persistent antigen, chronic T cell receptor signaling drives T lymphocytes to a functionally exhausted state. Here we discuss how the tumor and its microenvironment influences T cell trafficking and function with a focus on melanoma, and pancreatic and ovarian cancer, and discuss how scientific advances may help overcome these hurdles.
Collapse
Affiliation(s)
- Kristin G Anderson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Ingunn M Stromnes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Philip D Greenberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Mail Stop D3-100, P.O. Box 19024, Seattle, WA 98109, USA; Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
169
|
Goldman N, Valiuskyte K, Londregan J, Swider A, Somerville J, Riggs JE. Macrophage regulation of B cell proliferation. Cell Immunol 2017; 314:54-62. [PMID: 28238361 DOI: 10.1016/j.cellimm.2017.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/07/2017] [Accepted: 02/12/2017] [Indexed: 12/11/2022]
Abstract
Unlike organized lymphoid tissue, the tumor microenvironment (TME) often includes a high proportion of immunosuppressive macrophages. We model the TME by culturing peritoneal cavity (PerC) cells that naturally have a high macrophage to lymphocyte ratio. Prior studies revealed that, following TCR ligation, PerC T cell proliferation is suppressed due to IFNγ-triggered inducible nitric oxide synthase expression. In this study we assessed the ability of PerC B cells to respond to surrogate activating signals in the presence of high numbers of macrophages. Surface IgM (BCR) ligation led to cyclooxygenase-mediated, and TLR-4 ligation to IL10-mediated, suppression of PerC B cell proliferation. In contrast, PerC B cells had a robust response to CD40 ligation, which could overcome the suppression generated by the BCR or TLR-4 response. However, the CD40 response was suppressed by concurrent TCR ligation. These results reveal the challenges of promoting B and T cell responses in macrophage-rich conditions that model the TME.
Collapse
Affiliation(s)
- Naomi Goldman
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | | | | | - Adam Swider
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - John Somerville
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - James E Riggs
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA.
| |
Collapse
|
170
|
Epithelial Hypoxia-Inducible Factor 2α Facilitates the Progression of Colon Tumors through Recruiting Neutrophils. Mol Cell Biol 2017; 37:MCB.00481-16. [PMID: 27956697 DOI: 10.1128/mcb.00481-16] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 12/02/2016] [Indexed: 12/24/2022] Open
Abstract
Inflammation is a significant risk factor for colon cancer. Recent work has demonstrated essential roles for several infiltrating immune populations in the metaplastic progression following inflammation. Hypoxia and stabilization of hypoxia-inducible factors (HIFs) are hallmark features of inflammation and solid tumors. Previously, we demonstrated an important role for tumor epithelial HIF-2α in colon tumors; however, the function of epithelial HIF-2α as a critical link in the progression of inflammation to cancer has not been elucidated. In colitis-associated colon cancer models, epithelial HIF-2α was essential in tumor growth. Concurrently, epithelial disruption of HIF-2α significantly decreased neutrophils in the colon tumor microenvironment. Intestinal epithelial HIF-2α-overexpressing mice demonstrated that neutrophil recruitment was a direct response to increased epithelial HIF-2α signaling. High-throughput RNA sequencing (RNA-seq) analysis of HIF-2α-overexpressing mice in conjunction with data mining from the Cancer Genome Atlas showed that the neutrophil chemokine CXCL1 gene was highly upregulated in colon tumor epithelium in a HIF-2α-dependent manner. Using selective peptide inhibitors of the CXCL1-CXCR2 signaling axis identified HIF-2α-dependent neutrophil recruitment as an essential mechanism to increase colon carcinogenesis. These studies demonstrate that HIF-2α is a novel regulator of neutrophil recruitment to colon tumors and that it is essential in shaping the protumorigenic inflammatory microenvironment in colon cancer.
Collapse
|
171
|
Crosstalk between stromal cells and cancer cells in pancreatic cancer: New insights into stromal biology. Cancer Lett 2017; 392:83-93. [PMID: 28189533 DOI: 10.1016/j.canlet.2017.01.041] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/28/2017] [Accepted: 01/31/2017] [Indexed: 12/18/2022]
Abstract
Pancreatic cancer (PC) remains one of the most lethal malignancies worldwide. Increasing evidence has confirmed the pivotal role of stromal components in the regulation of carcinogenesis, invasion, metastasis, and therapeutic resistance in PC. Interaction between neoplastic cells and stromal cells builds a specific microenvironment, which further modulates the malignant properties of cancer cells. Instead of being a "passive bystander", stroma may play a role as a "partner in crime" in PC. However, the role of stromal components in PC is complex and requires further investigation. In this article, we review recent advances regarding the regulatory roles and mechanisms of stroma biology, especially the cellular components such as pancreatic stellate cells, macrophages, neutrophils, adipocytes, epithelial cells, pericytes, mast cells, and lymphocytes, in PC. Crosstalk between stromal cells and cancer cells is thoroughly investigated. We also review the prognostic value and molecular therapeutic targets of stroma in PC. This review may help us further understand the molecular mechanisms of stromal biology and its role in PC development and therapeutic resistance. Moreover, targeting stroma components may provide new therapeutic strategies for this stubborn disease.
Collapse
|
172
|
Halbrook CJ, Lyssiotis CA. Employing Metabolism to Improve the Diagnosis and Treatment of Pancreatic Cancer. Cancer Cell 2017; 31:5-19. [PMID: 28073003 DOI: 10.1016/j.ccell.2016.12.006] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/03/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma is on pace to become the second leading cause of cancer-related death. The high mortality rate results from a lack of methods for early detection and the inability to successfully treat patients once diagnosed. Pancreatic cancer cells have extensively reprogrammed metabolism, which is driven by oncogene-mediated cell-autonomous pathways, the unique physiology of the tumor microenvironment, and interactions with non-cancer cells. In this review, we discuss how recent efforts delineating rewired metabolic networks in pancreatic cancer have revealed new in-roads to develop detection and treatment strategies for this dreadful disease.
Collapse
Affiliation(s)
- Christopher J Halbrook
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
173
|
In vivo functional dissection of a context-dependent role for Hif1α in pancreatic tumorigenesis. Oncogenesis 2016; 5:e278. [PMID: 27941931 PMCID: PMC5177776 DOI: 10.1038/oncsis.2016.78] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/31/2016] [Indexed: 01/25/2023] Open
Abstract
Hypoxia-inducible factor 1α (Hif1α) is a key regulator of cellular adaptation and survival under hypoxic conditions. In pancreatic ductal adenocarcinoma (PDAC), it has been recently shown that genetic ablation of Hif1α accelerates tumour development by promoting tumour-supportive inflammation in mice, questioning its role as the key downstream target of many oncogenic signals of PDAC. Likely, Hif1α has a context-dependent role in pancreatic tumorigenesis. To further analyse this, murine PDAC cell lines with reduced Hif1α expression were generated using shRNA transfection. Cells were transplanted into wild-type mice through orthotopic or portal vein injection in order to test the in vivo function of Hif1α in two major tumour-associated biological scenarios: primary tumour growth and remote colonization/metastasis. Although Hif1α protects PDAC cells from stress-induced cell deaths in both scenarios-in line with the general function Hif1α-its depletion leads to different oncogenic consequences. Hif1α depletion results in rapid tumour growth with marked hypoxia-induced cell death, which potentially leads to a persistent tumour-sustaining inflammatory response. However, it simultaneously reduces tumour colonization and hepatic metastases by increasing the susceptibility to anoikis induced by anchorage-independent conditions. Taken together, the role of Hif1α in pancreatic tumorigenesis is context-dependent. Clinical trials of Hif1α inhibitors need to take this into account, targeting the appropriate scenario, for example palliative vs adjuvant therapy.
Collapse
|
174
|
Chang JH, Jiang Y, Pillarisetty VG. Role of immune cells in pancreatic cancer from bench to clinical application: An updated review. Medicine (Baltimore) 2016; 95:e5541. [PMID: 27930550 PMCID: PMC5266022 DOI: 10.1097/md.0000000000005541] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) remains difficult to treat, despite the recent advances in various anticancer therapies. Immuno-inflammatory response is considered to be a major risk factor for the development of PC in addition to a combination of genetic background and environmental factors. Although patients with PC exhibit evidence of systemic immune dysfunction, the PC microenvironment is replete with immune cells. METHODS We searched PubMed for all relevant English language articles published up to March 2016. They included clinical trials, experimental studies, observational studies, and reviews. Trials enrolled at Clinical trial.gov were also searched. RESULTS PC induces an immunosuppressive microenvironment, and intratumoral activation of immunity in PC is attenuated by inhibitory signals that limit immune effector function. Multiple types of immune responses can promote an immunosuppressive microenvironment; key regulators of the host tumor immune response are dendritic cells, natural killer cells, macrophages, myeloid derived suppressor cells, and T cells. The function of these immune cells in PC is also influenced by chemotherapeutic agents and the components in tumor microenvironment such as pancreatic stellate cells. Immunotherapy of PC employs monoclonal antibodies/effector cells generated in vitro or vaccination to stimulate antitumor response. Immune therapy in PC has failed to improve overall survival; however, combination therapies comprising immune checkpoint inhibitors and vaccines have been attempted to increase the response. CONCLUSION A number of studies have begun to elucidate the roles of immune cell subtypes and their capacity to function or dysfunction in the tumor microenvironment of PC. It will not be long before immune therapy for PC becomes a clinical reality.
Collapse
Affiliation(s)
- Jae Hyuck Chang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yongjian Jiang
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Venu G. Pillarisetty
- Department of Surgery, University of Washington Medical Center, Seattle, University of Washington, Seattle, WA
| |
Collapse
|
175
|
Diana A, Wang LM, D'Costa Z, Azad A, Silva MA, Soonawalla Z, Allen P, Liu S, McKenna WG, Muschel RJ, Fokas E. Prognostic role and correlation of CA9, CD31, CD68 and CD20 with the desmoplastic stroma in pancreatic ductal adenocarcinoma. Oncotarget 2016; 7:72819-72832. [PMID: 27637082 PMCID: PMC5341946 DOI: 10.18632/oncotarget.12022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/09/2016] [Indexed: 12/18/2022] Open
Abstract
We assessed the prognostic value of hypoxia (carbonic anhydrase 9; CA9), vessel density (CD31), with macrophages (CD68) and B cells (CD20) that can interact and lead to immune suppression and disease progression using scanning and histological mapping of whole-mount FFPE pancreatectomy tissue sections from 141 primarily resectable pancreatic ductal adenocarcinoma (PDAC) samples treated with surgery and adjuvant chemotherapy. Their expression was correlated with clinicopathological characteristics, and overall survival (OS), progression-free survival (PFS), local progression-free survival (LPFS) and distant metastases free-survival (DMFS), also in the context of stroma density (haematoxylin-eosin) and activity (alpha-smooth muscle actin). The median OS was 21 months after a mean follow-up of 20 months (range, 2-69 months). The median tumor surface area positive for CA9 and CD31 was 7.8% and 8.1%, respectively. Although total expression of these markers lacked prognostic value in the entire cohort, nevertheless, high tumor compartment CD68 expression correlated with worse PFS (p = 0.033) and DMFS (p = 0.047). Also, high CD31 expression predicted for worse OS (p = 0.004), PFS (p = 0.008), LPFS (p = 0.014) and DMFS (p = 0.004) in patients with moderate density stroma. High stromal and peripheral compartment CD68 expression predicted for significantly worse outcome in patients with loose and moderate stroma density, respectively. Altogether, in contrast to the current notion, hypoxia levels in PDAC appear to be comparable to other malignancies. CD31 and CD68 constitute prognostic markers in patient subgroups that vary according to tumor compartment and stromal density. Our study provides important insight on the pathophysiology of PDAC and should be exploited for future treatments.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antigens, CD/metabolism
- Antigens, CD20/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor
- Carbonic Anhydrase IX/metabolism
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/therapy
- Combined Modality Therapy
- Female
- Humans
- Hypoxia/metabolism
- Immunohistochemistry
- Macrophages/metabolism
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Staging
- Neovascularization, Pathologic/metabolism
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/therapy
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Prognosis
- Stromal Cells/metabolism
- Survival Analysis
Collapse
Affiliation(s)
- Angela Diana
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Lai Mun Wang
- Department of Pathology, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - Zenobia D'Costa
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Abul Azad
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Michael A. Silva
- Department of Surgery, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - Zahir Soonawalla
- Department of Surgery, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - Paul Allen
- Department of Pathology, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - Stanley Liu
- Department of Radiation Oncology, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - W. Gillies McKenna
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Ruth J. Muschel
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Emmanouil Fokas
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
- Current Address: Department of Radiotherapy and Oncology, Goethe University of Frankfurt, Frankfurt, Germany
| |
Collapse
|
176
|
Tsou P, Katayama H, Ostrin EJ, Hanash SM. The Emerging Role of B Cells in Tumor Immunity. Cancer Res 2016; 76:5597-5601. [PMID: 27634765 DOI: 10.1158/0008-5472.can-16-0431] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/21/2016] [Indexed: 12/24/2022]
Abstract
There is increasing evidence supporting a role for B cells in tumor immunology. Paraneoplastic syndromes occurring before a cancer diagnosis have pointed to the potential for harnessing the humoral immune response for early cancer detection. The presence of tumor-infiltrating B lymphocytes has been linked to a favorable clinical outcome in many types of cancers. However, B cells represent a heterogeneous population with functionally distinct subsets, and the balance among subtypes impacts tumor development. Here, we review recent findings related to B cells and to the humoral immune response in cancer and their translational significance. Cancer Res; 76(19); 5597-601. ©2016 AACR.
Collapse
Affiliation(s)
- Peiling Tsou
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Edwin J Ostrin
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Samir M Hanash
- The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
177
|
Abstract
PURPOSE OF REVIEW The review intends to describe recent studies on the development of pancreatic cancer from a genetic, molecular, and microenvironment perspective. RECENT FINDINGS Pancreatic cancer has been discovered to have distinct molecular subtypes based on transcriptome analyses that may have implications for treatment. Recent studies are also mapping the complex molecular biology of this cancer as it relates to the core signaling abnormalities inherent to this disease. There have been discoveries of novel modes of regulation of pancreatic cancer development, including alterations in key transcription factors, epigenetic modifiers, and metabolic pathways. Studies of the tumor-associated microenvironment continue to reveal its complex role in tumor development. SUMMARY Pancreatic cancer development appears to depend on a multifaceted network of signals that are dynamic, involve multiple cell types, and are linked to spatiotemporal factors in tumor evolution. Understanding the development of pancreatic cancer in this context is key to identifying novel and effective targets for treatment.
Collapse
|
178
|
Abstract
Three recent studies, approaching the question from different angles and using different and/or overlapping models, provide compelling evidence for the involvement of tumor-infiltrating B cells in the initiation and progression of pancreatic ductal adenocarcinoma. These studies highlight the need for a better understanding of pancreatic tumor-immune system interactions and the immunologic mechanisms that promote or inhibit tumorigenesis, paving the way for better treatment strategies.
Collapse
Affiliation(s)
- Ali Roghanian
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts. Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Christopher Fraser
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Marianna Kleyman
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
179
|
Triner D, Shah YM. Hypoxia-inducible factors: a central link between inflammation and cancer. J Clin Invest 2016; 126:3689-3698. [PMID: 27525434 DOI: 10.1172/jci84430] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The tumor immune response is in a dynamic balance between antitumor mechanisms, which serve to decrease cancer growth, and the protumor inflammatory response, which increases immune tolerance, cell survival, and proliferation. Hypoxia and expression of HIF-1α and HIF-2α are characteristic features of all solid tumors. HIF signaling serves as a major adaptive mechanism in tumor growth in a hypoxic microenvironment. HIFs represent a critical signaling node in the switch to protumorigenic inflammatory responses through recruitment of protumor immune cells and altered immune cell effector functions to suppress antitumor immune responses and promote tumor growth through direct growth-promoting cytokine production, angiogenesis, and ROS production. Modulating HIF function will be an important mechanism to dampen the tumor-promoting inflammatory response and inhibit cancer growth.
Collapse
|
180
|
Schwartz M, Zhang Y, Rosenblatt JD. B cell regulation of the anti-tumor response and role in carcinogenesis. J Immunother Cancer 2016; 4:40. [PMID: 27437104 PMCID: PMC4950763 DOI: 10.1186/s40425-016-0145-x] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 06/30/2016] [Indexed: 02/06/2023] Open
Abstract
The balance between immune effector cells such as T cells and natural killer cells, and immunosuppressive Treg cells, dendritic, myeloid and monocytic sub-populations in the tumor microenvironment acts to calibrate the immune response to malignant cells. Accumulating evidence is pointing to a role for B cells in modulating the immune response to both solid tumors and hematologic cancer. Evidence from murine autoimmune models has defined B regulatory cell (Breg) subsets that express cytokines such as IL-10, TGF-β, and/or express immune regulatory ligands such as PD-L1, which can suppress T cell and/or natural killer cell responses. Multiple murine tumor models exhibit decreased tumor growth in B cell deficient or B cell depleted mice. In several of these models, B cells inhibit T cell mediated tumor immunity and/or facilitate conversion of T cells to CD4+CD25+FoxP3+ T regs, which act to attenuate the innate and/or adaptive antitumor immune response. Mechanisms of suppression include the acquisition of inhibitory ligand expression, and phosphorylation of Stat3, and induction of IL-10 and TGF-β, resulting in a Breg phenotype. Breg suppressive activity may affect diverse cell subtypes, including T effector cells, NK cells, myeloid derived suppressor cells (MDSC) and/or tumor associated macrophages. B cells may also directly promote tumorigenesis through recruitment of inflammatory cells, and upregulation of pro-angiogenic genes and pro-metastatic collagenases. Breg infiltration has now been identified in a variety of solid tumor malignancies including but not limited to ovarian, gastric, non-small cell lung cancer, pancreatic, esophageal, head and neck, and hepatocellular carcinomas. Increasing evidence suggests that recruitment of B cells and acquisition of suppressive activity within the tumor bed may be an important mechanism through which B cells may modulate innate and/or adaptive anti-tumor immunity. B cell depletion in the clinic using anti-CD20 antibodies and/or inhibitors of BTK and/or other signaling pathways, may be a useful strategy for augmenting the anti-tumor immune response.
Collapse
Affiliation(s)
- Marc Schwartz
- Department of Medicine, University of Miami Miller School of Medicine, 1120 NW 14th St., CRB 610, Miami, FL 33136 USA
| | - Yu Zhang
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, 1120 NW 14th St., CRB 610, Miami, FL 33136 USA ; Department of Medicine, University of Miami Miller School of Medicine, 1120 NW 14th St., CRB 610, Miami, FL 33136 USA ; UM Sylvester Comprehensive Cancer Center, 1120 NW 14th St., CRB 610, Miami, FL 33136 USA
| | - Joseph D Rosenblatt
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, 1120 NW 14th St., CRB 610, Miami, FL 33136 USA ; Department of Medicine, University of Miami Miller School of Medicine, 1120 NW 14th St., CRB 610, Miami, FL 33136 USA ; UM Sylvester Comprehensive Cancer Center, 1120 NW 14th St., CRB 610, Miami, FL 33136 USA
| |
Collapse
|
181
|
Hutcheson J, Balaji U, Porembka MR, Wachsmann MB, McCue PA, Knudsen ES, Witkiewicz AK. Immunologic and Metabolic Features of Pancreatic Ductal Adenocarcinoma Define Prognostic Subtypes of Disease. Clin Cancer Res 2016; 22:3606-17. [PMID: 26858311 PMCID: PMC4947442 DOI: 10.1158/1078-0432.ccr-15-1883] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/25/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDA) is associated with an immunosuppressive microenvironment that supports the growth of the malignancy as well as immune system evasion. Here we examine markers of immunosuppression in PDA within the context of the glycolytic tumor microenvironment, their interrelationship with tumor biology and association with overall survival. EXPERIMENTAL DESIGN We utilized tissue microarrays consisting of 223 PDA patients annotated for clinical stage, tumor size, lymph node involvement, and survival. Expression of CD163, FoxP3, PD-L1, and MCT4 was assessed by IHC and statistical associations were evaluated by univariate and multivariate analysis. Multimarker subtypes were defined by random forest analysis. Mechanistic interactions were evaluated using PDA cell lines and models for myeloid differentiation. RESULTS PDA exhibits discrete expression of CD163, FoxP3, and PD-L1 with modest individual significance. However, combined low expression of these markers was associated with improved prognosis (P = 0.02). PDA tumor cells altered macrophage phenotype and function, which supported enhanced invasiveness in cell-based models. Lactate efflux mediated by MCT4 was associated with, and required for, the selective conversion of myeloid cells. Correspondingly, MCT4 expression correlated with immune markers in PDA cases, and increased the significance of prognostic subtypes (P = 0.002). CONCLUSIONS There exists a complex interplay between PDA tumor cells and the host immune system wherein immunosuppression is associated with negative outcome. MCT4 expression, representative of the glycolytic state of PDA, contributes to the phenotypic conversion of myeloid cells. Thus, metabolic status of PDA tumors is an important determinant of the immunosuppressive environment. Clin Cancer Res; 22(14); 3606-17. ©2016 AACR.
Collapse
Affiliation(s)
- Jack Hutcheson
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas
| | - Uthra Balaji
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas
| | - Matthew R Porembka
- Simmons Cancer Center, UT Southwestern Medical Center, Dallas, Texas. Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Megan B Wachsmann
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Erik S Knudsen
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Agnieszka K Witkiewicz
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, Texas. Department of Pathology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
182
|
Ying H, Dey P, Yao W, Kimmelman AC, Draetta GF, Maitra A, DePinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 2016; 30:355-85. [PMID: 26883357 PMCID: PMC4762423 DOI: 10.1101/gad.275776.115] [Citation(s) in RCA: 391] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ying et al. review pancreatic ductal adenocarcinoma (PDAC) genetics and biology, particularly altered cancer cell metabolism, the complexity of immune regulation in the tumor microenvironment, and impaired DNA repair processes. With 5-year survival rates remaining constant at 6% and rising incidences associated with an epidemic in obesity and metabolic syndrome, pancreatic ductal adenocarcinoma (PDAC) is on track to become the second most common cause of cancer-related deaths by 2030. The high mortality rate of PDAC stems primarily from the lack of early diagnosis and ineffective treatment for advanced tumors. During the past decade, the comprehensive atlas of genomic alterations, the prominence of specific pathways, the preclinical validation of such emerging targets, sophisticated preclinical model systems, and the molecular classification of PDAC into specific disease subtypes have all converged to illuminate drug discovery programs with clearer clinical path hypotheses. A deeper understanding of cancer cell biology, particularly altered cancer cell metabolism and impaired DNA repair processes, is providing novel therapeutic strategies that show strong preclinical activity. Elucidation of tumor biology principles, most notably a deeper understanding of the complexity of immune regulation in the tumor microenvironment, has provided an exciting framework to reawaken the immune system to attack PDAC cancer cells. While the long road of translation lies ahead, the path to meaningful clinical progress has never been clearer to improve PDAC patient survival.
Collapse
Affiliation(s)
- Haoqiang Ying
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Prasenjit Dey
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wantong Yao
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Alec C Kimmelman
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Giulio F Draetta
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA; Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Anirban Maitra
- Department of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA; Sheikh Ahmed Pancreatic Cancer Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
183
|
Gunderson AJ, Kaneda MM, Tsujikawa T, Nguyen AV, Affara NI, Ruffell B, Gorjestani S, Liudahl SM, Truitt M, Olson P, Kim G, Hanahan D, Tempero MA, Sheppard B, Irving B, Chang BY, Varner JA, Coussens LM. Bruton Tyrosine Kinase-Dependent Immune Cell Cross-talk Drives Pancreas Cancer. Cancer Discov 2016; 6:270-85. [PMID: 26715645 PMCID: PMC4783268 DOI: 10.1158/2159-8290.cd-15-0827] [Citation(s) in RCA: 392] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Pancreas ductal adenocarcinoma (PDAC) has one of the worst 5-year survival rates of all solid tumors, and thus new treatment strategies are urgently needed. Here, we report that targeting Bruton tyrosine kinase (BTK), a key B-cell and macrophage kinase, restores T cell-dependent antitumor immune responses, thereby inhibiting PDAC growth and improving responsiveness to standard-of-care chemotherapy. We report that PDAC tumor growth depends on cross-talk between B cells and FcRγ(+) tumor-associated macrophages, resulting in T(H)2-type macrophage programming via BTK activation in a PI3Kγ-dependent manner. Treatment of PDAC-bearing mice with the BTK inhibitor PCI32765 (ibrutinib) or by PI3Kγ inhibition reprogrammed macrophages toward a T(H)1 phenotype that fostered CD8(+) T-cell cytotoxicity, and suppressed PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. These data indicate that pharmacologic inhibition of BTK in PDAC can reactivate adaptive immune responses, presenting a new therapeutic modality for this devastating tumor type. SIGNIFICANCE We report that BTK regulates B-cell and macrophage-mediated T-cell suppression in pancreas adenocarcinomas. Inhibition of BTK with the FDA-approved inhibitor ibrutinib restores T cell-dependent antitumor immune responses to inhibit PDAC growth and improves responsiveness to chemotherapy, presenting a new therapeutic modality for pancreas cancer.
Collapse
Affiliation(s)
- Andrew J Gunderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Megan M Kaneda
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Takahiro Tsujikawa
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon. Department of Otolaryngology-Head and Neck Surgery, Oregon Health and Science University, Portland, Oregon
| | - Abraham V Nguyen
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Nesrine I Affara
- Department of Pathology, University of California, San Francisco, California
| | - Brian Ruffell
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Sara Gorjestani
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Shannon M Liudahl
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Morgan Truitt
- Department of Biochemistry and Biophysics, University of California, San Francisco, California
| | - Peter Olson
- Department of Biochemistry and Biophysics, University of California, San Francisco, California
| | - Grace Kim
- Department of Pathology, University of California, San Francisco, California. Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Margaret A Tempero
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California. Department of Medicine, University of California, San Francisco, California
| | - Brett Sheppard
- Department of Surgery, Oregon Health and Science University, Portland, Oregon. Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | | | | | - Judith A Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, California. Department of Pathology, University of California, San Diego, La Jolla, California.
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon. Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
184
|
Pylayeva-Gupta Y, Das S, Handler JS, Hajdu CH, Coffre M, Koralov SB, Bar-Sagi D. IL35-Producing B Cells Promote the Development of Pancreatic Neoplasia. Cancer Discov 2016; 6:247-55. [PMID: 26715643 PMCID: PMC5709038 DOI: 10.1158/2159-8290.cd-15-0843] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022]
Abstract
UNLABELLED A salient feature of pancreatic ductal adenocarcinoma (PDAC) is an abundant fibroinflammatory response characterized by the recruitment of immune and mesenchymal cells and the consequent establishment of a protumorigenic microenvironment. Here, we report the prominent presence of B cells in human pancreatic intraepithelial neoplasia and PDAC lesions as well as in oncogenic Kras-driven pancreatic neoplasms in the mouse. The growth of orthotopic pancreatic neoplasms harboring oncogenic Kras was significantly compromised in B-cell-deficient mice (μMT), and this growth deficiency could be rescued by the reconstitution of a CD1d(hi)CD5(+) B-cell subset. The protumorigenic effect of B cells was mediated by their expression of IL35 through a mechanism involving IL35-mediated stimulation of tumor cell proliferation. Our results identify a previously unrecognized role for IL35-producing CD1d(hi)CD5(+) B cells in the pathogenesis of pancreatic cancer and underscore the potential significance of a B-cell/IL35 axis as a therapeutic target. SIGNIFICANCE This study identifies a B-cell subpopulation that accumulates in the pancreatic parenchyma during early neoplasia and is required to support tumor cell growth. Our findings provide a rationale for exploring B-cell-based targeting approaches for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yuliya Pylayeva-Gupta
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York
| | - Shipra Das
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York
| | - Jesse S Handler
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York
| | - Cristina H Hajdu
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Maryaline Coffre
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York.
| |
Collapse
|
185
|
|