Immunotherapy Converts Nonimmunogenic Pancreatic Tumors into Immunogenic Foci of Immune Regulation

Cancer Immunology Research - Tập 2 Số 7 - Trang 616-631 - 2014
Eric R. Lutz1,2,3,4, Annie A. Wu5,1,4, Elaine Bigelow1,4, Rajni Sharma6, Guanglan Mo1,2,4, Kevin C. Soares5,1,2,3,4, Sara Solt1,2,4, Alvin Dorman1,2,4, Anthony Wamwea1,2,4, Allison Yager1,4, Daniel A. Laheru1,2,4, Christopher L. Wolfgang5,1,3,4, Jiang Wang7, Ralph H. Hruban6,1,3,4, Robert A. Anders6,1,3,4, Elizabeth M. Jaffee6,1,2,3,4, Lei Zheng5,1,2,3,4
14The Sidney Kimmel Cancer Center;
25The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care;
36The Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
4Authors' Affiliations: Departments of 1Oncology,
53Surgery;
62Pathology and
77Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio

Tóm tắt

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is considered a “nonimmunogenic” neoplasm. Single-agent immunotherapies have failed to demonstrate significant clinical activity in PDAC and other “nonimmunogenic” tumors, in part due to a complex tumor microenvironment (TME) that provides a formidable barrier to immune infiltration and function. We designed a neoadjuvant and adjuvant clinical trial comparing an irradiated, granulocyte-macrophage colony-stimulating factor (GM-CSF)–secreting, allogeneic PDAC vaccine (GVAX) given as a single agent or in combination with low-dose cyclophosphamide to deplete regulatory T cells (Treg) as a means to study how the TME is altered by immunotherapy. Examination of resected PDACs revealed the formation of vaccine-induced intratumoral tertiary lymphoid aggregates in 33 of 39 patients 2 weeks after vaccine treatment. Immunohistochemical analysis showed these aggregates to be regulatory structures of adaptive immunity. Microarray analysis of microdissected aggregates identified gene-expression signatures in five signaling pathways involved in regulating immune-cell activation and trafficking that were associated with improved postvaccination responses. A suppressed Treg pathway and an enhanced Th17 pathway within these aggregates were associated with improved survival, enhanced postvaccination mesothelin-specific T-cell responses, and increased intratumoral Teff:Treg ratios. This study provides the first example of immune-based therapy converting a “nonimmunogenic” neoplasm into an “immunogenic” neoplasm by inducing infiltration of T cells and development of tertiary lymphoid structures in the TME. Post-GVAX T-cell infiltration and aggregate formation resulted in the upregulation of immunosuppressive regulatory mechanisms, including the PD-1–PD-L1 pathway, suggesting that patients with vaccine-primed PDAC may be better candidates than vaccine-naïve patients for immune checkpoint and other immunomodulatory therapies. Cancer Immunol Res; 2(7); 616–31. ©2014 AACR.

Từ khóa


Tài liệu tham khảo

American Cancer Society, 2012, Cancer facts and figures

Brahmer, 2010, Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates, J Clin Oncol, 28, 3167, 10.1200/JCO.2009.26.7609

Hodi, 2008, Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients, Proc Natl Acad Sci U S A, 105, 3005, 10.1073/pnas.0712237105

Sharma, 2011, Novel cancer immunotherapy agents with survival benefit: recent successes and next steps, Nat Rev Cancer, 11, 805, 10.1038/nrc3153

Clark, 2009, Immunosurveillance of pancreatic adenocarcinoma: insights from genetically engineered mouse models of cancer, Cancer Lett, 279, 1, 10.1016/j.canlet.2008.09.037

Hiraoka, 2006, Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions, Clin Cancer Res, 12, 5423, 10.1158/1078-0432.CCR-06-0369

Melief, 1991, T-cell immunotherapy of cancer, Res Immunol, 142, 425, 10.1016/0923-2494(91)90042-H

von Bernstorff, 2001, Systemic and local immunosuppression in pancreatic cancer patients, Clin Cancer Res, 7, 925s

Koido, 2011, Current immunotherapeutic approaches in pancreatic cancer, Clin Dev Immunol, 2011, 267539, 10.1155/2011/267539

Brahmer, 2012, Safety and activity of anti-PD-L1 antibody in patients with advanced cancer, N Engl J Med, 366, 2455, 10.1056/NEJMoa1200694

Royal, 2010, Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma, J Immunother, 33, 828, 10.1097/CJI.0b013e3181eec14c

Le, 2013, Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer, J Immunother, 36, 382, 10.1097/CJI.0b013e31829fb7a2

Soares, 2012, Vaccines for pancreatic cancer, Cancer J, 18, 642, 10.1097/PPO.0b013e3182756903

Jaffee, 2001, Novel allogeneic granulocyte-macrophage colony-stimulating factor–secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation, J Clin Oncol, 19, 145, 10.1200/JCO.2001.19.1.145

Jaffee, 1998, Development and characterization of a cytokine-secreting pancreatic adenocarcinoma vaccine from primary tumors for use in clinical trials, Cancer J Sci Am, 4, 194

Laheru, 2008, Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation, Clin Cancer Res, 14, 1455, 10.1158/1078-0432.CCR-07-0371

Lutz, 2011, A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A Phase II trial of safety, efficacy, and immune activation, Ann Surg, 253, 328, 10.1097/SLA.0b013e3181fd271c

Thomas, 2004, Mesothelin-specific CD8(+) T cell responses provide evidence of in vivo cross-priming by antigen-presenting cells in vaccinated pancreatic cancer patients, J Exp Med, 200, 297, 10.1084/jem.20031435

Gajewski, 2006, Immune resistance orchestrated by the tumor microenvironment, Immunol Rev, 213, 131, 10.1111/j.1600-065X.2006.00442.x

Curiel, 2004, Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival, Nat Med, 10, 942, 10.1038/nm1093

Linehan, 2005, CD25+ CD4+ regulatory T-cells in cancer, Immunol Res, 32, 155, 10.1385/IR:32:1-3:155

Liyanage, 2002, Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma, J Immunol, 169, 2756, 10.4049/jimmunol.169.5.2756

Mougiakakos, 2010, Regulatory T cells in cancer, Adv Cancer Res, 107, 57, 10.1016/S0065-230X(10)07003-X

Ercolini, 2005, Recruitment of latent pools of high-avidity CD8(+) T cells to the antitumor immune response, J Exp Med, 201, 1591, 10.1084/jem.20042167

Ghiringhelli, 2004, CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative, Eur J Immunol, 34, 336, 10.1002/eji.200324181

Hermans, 2003, Synergistic effect of metronomic dosing of cyclophosphamide combined with specific antitumor immunotherapy in a murine melanoma model, Cancer Res, 63, 8408

Leao, 2008, Effective depletion of regulatory T cells allows the recruitment of mesothelin-specific CD8 T cells to the antitumor immune response against a mesothelin-expressing mouse pancreatic adenocarcinoma, Clin Transl Sci, 1, 228, 10.1111/j.1752-8062.2008.00070.x

Weiss, 2012, Targeting the right regulatory T-cell population for tumor immunotherapy, Oncoimmunology, 1, 1191, 10.4161/onci.20664

Weiss, 2012, Trafficking of high avidity HER-2/neu–specific T cells into HER-2/neu–expressing tumors after depletion of effector/memory-like regulatory T cells, PLoS ONE, 7, e31962, 10.1371/journal.pone.0031962

Cipponi, 2012, Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases, Cancer Res, 72, 3997, 10.1158/0008-5472.CAN-12-1377

Messina, 2012, 12-Chemokine gene signature identifies lymph node–like structures in melanoma: potential for patient selection for immunotherapy?, Sci Rep, 2, 765, 10.1038/srep00765

Coppola, 2011, Unique ectopic lymph node–like structures present in human primary colorectal carcinoma are identified by immune gene array profiling, Am J Pathol, 179, 37, 10.1016/j.ajpath.2011.03.007

Dieu-Nosjean, 2008, Long-term survival for patients with non–small-cell lung cancer with intratumoral lymphoid structures, J Clin Oncol, 26, 4410, 10.1200/JCO.2007.15.0284

Spranger, 2013, Up-regulation of PD-L1, IDO, and Tregs in the melanoma tumor microenvironment is driven by CD8+ T cells, Sci Transl Med, 5, 200ra116, 10.1126/scitranslmed.3006504

Taube, 2012, Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape, Sci Transl Med, 4, 127ra37, 10.1126/scitranslmed.3003689

Ghiringhelli, 2007, Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients, Cancer Immunol immunother, 56, 641, 10.1007/s00262-006-0225-8

Bigelow, 2013, Immunohistochemical staining of B7-H1 (PD-L1) on paraffin-embedded slides of pancreatic adenocarcinoma tissue, J Vis Exp, 4059

Lubbers, 2013, The type I IFN signature as a biomarker of preclinical rheumatoid arthritis, Ann Rheum Dis, 72, 776, 10.1136/annrheumdis-2012-202753

Subramanian, 2005, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles, Proc Natl Acad Sci U S A, 102, 15545, 10.1073/pnas.0506580102

van Nierop, 2002, Human follicular dendritic cells: function, origin and development, Sem Immunol, 14, 251, 10.1016/S1044-5323(02)00057-X

Fan, 2000, Cutting edge: ectopic expression of the chemokine TCA4/SLC is sufficient to trigger lymphoid neogenesis, J Immunol, 164, 3955, 10.4049/jimmunol.164.8.3955

Kline, 2010, Clinical development of mAbs to block the PD1 pathway as an immunotherapy for cancer, Curr Opin Investig Drugs, 11, 1354

Ino, 2013, Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer, Br J Cancer, 108, 914, 10.1038/bjc.2013.32

Shen, 2010, Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer, J Cancer Res Clin Oncol, 136, 1585, 10.1007/s00432-010-0816-9

Sinicrope, 2009, Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma, Gastroenterology, 137, 1270, 10.1053/j.gastro.2009.06.053

Chauhan, 2014, CCL-21 conditioned regulatory T cells induce allotolerance through enhanced homing to lymphoid tissue, J Immunol, 192, 817, 10.4049/jimmunol.1203469

Chen, 2013, High CCR6/CCR7 expression and Foxp3+ Treg cell number are positively related to the progression of laryngeal squamous cell carcinoma, Oncol Rep, 30, 1380, 10.3892/or.2013.2603

Chenivesse, 2012, Pulmonary CCL18 recruits human regulatory T cells, J Immunol, 189, 128, 10.4049/jimmunol.1003616

Porta, 2007, Tumor promotion by tumor-associated macrophages, Adv Exp Med Biol, 604, 67, 10.1007/978-0-387-69116-9_5

Sawanobori, 2008, Chemokine-mediated rapid turnover of myeloid-derived suppressor cells in tumor-bearing mice, Blood, 111, 5457, 10.1182/blood-2008-01-136895

Haile, 2010, CD49d is a new marker for distinct myeloid-derived suppressor cell subpopulations in mice, J Immunol, 185, 203, 10.4049/jimmunol.0903573

Jin, 2009, Regulation of Th17 cell differentiation and EAE induction by MAP3K NIK, Blood, 113, 6603, 10.1182/blood-2008-12-192914

Ruan, 2012, Nuclear factor-kappaB in immunity and inflammation: the Treg and Th17 connection, Adv Exp Med Biol, 946, 207, 10.1007/978-1-4614-0106-3_12

Chang, 2012, Ubc13 maintains the suppressive function of regulatory T cells and prevents their conversion into effector-like T cells, Nat Immunol, 13, 481, 10.1038/ni.2267

Ciofani, 2012, A validated regulatory network for Th17 cell specification, Cell, 151, 289, 10.1016/j.cell.2012.09.016

Lee, 2009, Late developmental plasticity in the T helper 17 lineage, Immunity, 30, 92, 10.1016/j.immuni.2008.11.005

Aloisi, 2006, Lymphoid neogenesis in chronic inflammatory diseases, Nat Rev Immunol, 6, 205, 10.1038/nri1786

Hjelmstrom, 2001, Lymphoid neogenesis: de novo formation of lymphoid tissue in chronic inflammation through expression of homing chemokines, J Leuk Biol, 69, 331, 10.1189/jlb.69.3.331

Quezada, 2006, CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells, J Clin Invest, 116, 1935, 10.1172/JCI27745

Weih, 2003, Regulation of secondary lymphoid organ development by the nuclear factor-kappaB signal transduction pathway, Immunol Rev, 195, 91, 10.1034/j.1600-065X.2003.00064.x

Foo, 2010, Regulation of inducible BALT formation and contribution to immunity and pathology, Mucosal immunol, 3, 537, 10.1038/mi.2010.52

Kocks, 2007, Regulatory T cells interfere with the development of bronchus-associated lymphoid tissue, J Exp Med, 204, 723, 10.1084/jem.20061424

Grogan, 2012, A role for Th17 cells in the regulation of tertiary lymphoid follicles, Eur J Immunol, 42, 2255, 10.1002/eji.201242656

Peters, 2011, Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation, Immunity, 35, 986, 10.1016/j.immuni.2011.10.015

Wang, 2009, IL-17 can promote tumor growth through an IL-6-Stat3 signaling pathway, J Exp Med, 206, 1457, 10.1084/jem.20090207

Acosta-Rodriguez, 2007, Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17–producing human T helper cells, Nat Immunol, 8, 942, 10.1038/ni1496

Hirota, 2011, Fate mapping of IL-17–producing T cells in inflammatory responses, Nat Immunol, 12, 255, 10.1038/ni.1993

Muranski, 2013, Essentials of Th17 cell commitment and plasticity, Blood, 121, 2402, 10.1182/blood-2012-09-378653

Sallusto, 2012, Human Th17 subsets, Eur J immunol, 42, 2215, 10.1002/eji.201242741

Zielinski, 2012, Pathogen-induced human TH17 cells produce IFN-gamma or IL-10 and are regulated by IL-1beta, Nature, 484, 514, 10.1038/nature10957

Martin, 2012, Controversies on the role of Th17 in cancer: a TGF-beta–dependent immunosuppressive activity?, Trends in Mol Med, 18, 742, 10.1016/j.molmed.2012.09.007

Wilke, 2011, Th17 cells in cancer: help or hindrance?, Carcinogenesis, 32, 643, 10.1093/carcin/bgr019

Ye, 2013, The role and regulation of human Th17 cells in tumor immunity, Am J Pathol, 182, 10, 10.1016/j.ajpath.2012.08.041