101
|
Kang BW, Chau I. Emerging agents for metastatic pancreatic cancer: spotlight on early phase clinical trials. Expert Opin Investig Drugs 2021; 30:1089-1107. [PMID: 34727804 DOI: 10.1080/13543784.2021.1995354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Despite the recent development of new chemotherapeutic regimens and combination strategies, metastatic pancreatic cancer (mPC) still shows only a modest response to conventional cytotoxic agents. However, several novel therapeutic agents targeting the unique features of mPC are showing promise in clinical trials. AREA COVERED This article reviews the current state of development of new agents targeting various systems and molecular pathways. We searched PubMed and clinicaltrials.gov in September 2021 with a special focus on ongoing early phase clinical trials to identify the promising therapeutic strategies for mPC. EXPERT OPINION Extensive tumor heterogeneity, complex tumor microenvironment, genetic alterations of the oncogenic signaling pathways, metabolic dysregulation, and a low immunogenicity are hurdles for current treatment approaches. Ongoing research efforts strive to overcome these hurdles and are showing some promising early results.
Collapse
Affiliation(s)
- Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Kyungpook National University, Daegu, Republic of Korea
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London, Surrey, UK
| |
Collapse
|
102
|
Guiding immunotherapy combinations: Who gets what? Adv Drug Deliv Rev 2021; 178:113962. [PMID: 34481029 DOI: 10.1016/j.addr.2021.113962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 01/27/2023]
Abstract
Although PD-1 and CTLA-4 inhibitors have proven successful in a range of malignancies, there are subsets of patients that do not respond to these agents due to upregulation of adaptive and innate resistance mechanisms by the tumor and its surrounding microenvironment. As new immunotherapeutic strategies are developed, there is a need for rational implementation of novel immunotherapy combinations that target complementary mechanisms of immunotherapy resistance intrinsic to each patient and tumor type. In this short review, we cover mechanisms by which tumors evade the immune system, as well as summarize available clinical data on emerging therapeutic agents that target these defense mechanisms. Rational implementation of combination immunotherapy targeting patient- and malignancy-specific immune evasion mechanisms may thus lead to enhanced response rates and allow immunotherapy to be effective even in tumors that are historically considered poorly responsive to immunotherapy.
Collapse
|
103
|
Freed-Pastor WA, Lambert LJ, Ely ZA, Pattada NB, Bhutkar A, Eng G, Mercer KL, Garcia AP, Lin L, Rideout WM, Hwang WL, Schenkel JM, Jaeger AM, Bronson RT, Westcott PMK, Hether TD, Divakar P, Reeves JW, Deshpande V, Delorey T, Phillips D, Yilmaz OH, Regev A, Jacks T. The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer. Cancer Cell 2021; 39:1342-1360.e14. [PMID: 34358448 PMCID: PMC8511341 DOI: 10.1016/j.ccell.2021.07.007] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/26/2021] [Accepted: 07/12/2021] [Indexed: 02/08/2023]
Abstract
The CD155/TIGIT axis can be co-opted during immune evasion in chronic viral infections and cancer. Pancreatic adenocarcinoma (PDAC) is a highly lethal malignancy, and immune-based strategies to combat this disease have been largely unsuccessful to date. We corroborate prior reports that a substantial portion of PDAC harbors predicted high-affinity MHC class I-restricted neoepitopes and extend these findings to advanced/metastatic disease. Using multiple preclinical models of neoantigen-expressing PDAC, we demonstrate that intratumoral neoantigen-specific CD8+ T cells adopt multiple states of dysfunction, resembling those in tumor-infiltrating lymphocytes of PDAC patients. Mechanistically, genetic and/or pharmacologic modulation of the CD155/TIGIT axis was sufficient to promote immune evasion in autochthonous neoantigen-expressing PDAC. Finally, we demonstrate that the CD155/TIGIT axis is critical in maintaining immune evasion in PDAC and uncover a combination immunotherapy (TIGIT/PD-1 co-blockade plus CD40 agonism) that elicits profound anti-tumor responses in preclinical models, now poised for clinical evaluation.
Collapse
Affiliation(s)
- William A Freed-Pastor
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Laurens J Lambert
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zackery A Ely
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nimisha B Pattada
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arjun Bhutkar
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - George Eng
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kim L Mercer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ana P Garcia
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lin Lin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - William M Rideout
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - William L Hwang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jason M Schenkel
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alex M Jaeger
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Roderick T Bronson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter M K Westcott
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Toni Delorey
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Devan Phillips
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Omer H Yilmaz
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Aviv Regev
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
104
|
Schlick K, Kiem D, Greil R. Recent Advances in Pancreatic Cancer: Novel Prognostic Biomarkers and Targeted Therapy-A Review of the Literature. Biomolecules 2021; 11:1469. [PMID: 34680101 PMCID: PMC8533343 DOI: 10.3390/biom11101469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic adenocarcinoma carries a devastating prognosis. For locally advanced and metastatic disease, several chemotherapeutic regimens are currently being used. Over the past years, novel approaches have included targeting EGFR, NTRK, PARP, K-Ras as well as stroma and fibrosis, leading to approval of NTRK and PARP inhibitors. Moreover, immune check point inhibitors and different combinational approaches involving immunotherapeutic agents are being investigated in many clinical trials. MiRNAs represent a novel tool and are thought to greatly improve management by allowing for earlier diagnosis and for more precise guidance of treatment.
Collapse
Affiliation(s)
- Konstantin Schlick
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (K.S.); (D.K.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Dominik Kiem
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (K.S.); (D.K.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Richard Greil
- Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria; (K.S.); (D.K.)
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Salzburg Cancer Research Institute, 5020 Salzburg, Austria
| |
Collapse
|
105
|
Mucileanu A, Chira R, Mircea PA. PD-1/PD-L1 expression in pancreatic cancer and its implication in novel therapies. Med Pharm Rep 2021; 94:402-410. [PMID: 36105495 PMCID: PMC9389876 DOI: 10.15386/mpr-2116] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/11/2021] [Accepted: 07/14/2021] [Indexed: 07/26/2023] Open
Abstract
Pancreatic cancer is the seventh leading cause of death in developed countries and it still has a poor prognosis despite intense research in the last 20 years. Immunotherapy is a relatively new strategy in cancer treatment. The aim of immunotherapy is to block the immunosuppressive effect of tumoral cells. The PD1/PD-L1 axis has an important role in the inhibition of effector T cells and the development of regulatory T cells (Tregs). Blocking these checkpoints, and also inhibitory signals, leads to apoptosis of Tregs and increased immune response of effector T cells against tumoral antigens. Unfortunately, pancreatic cancer is generally considered to be a non-immunogenic tumor. Thus PD-1/PD-L1 inhibitors demonstrated poor results in pancreatic cancer, excepting some patients with MSI/dMMR (microsatellite instability/deficient mismatch repair). Furthermore, pancreatic cancer has a particular microenvironment with a strong desmoplastic reaction, increased interstitial fluid pressure, hypoxic conditions, and acidic extracellular pH, which promote tumorigenesis and progression of the tumor. Mismatch repair deficiency (dMMR) is correlated with a high level of mutation-associated neoantigens, most recognized by immune cells which could predict a favorable response to anti-PD-1/PD-L1 therapy. PD-1/PD-L1 molecules could be also found as soluble forms (sPD-1, sPD-L1). These molecules have a potential role in the prognosis and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Adrian Mucileanu
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Romeo Chira
- Gastroenterology Department, Medical Clinic No. 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Petru Adrian Mircea
- Gastroenterology Department, Medical Clinic No. 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
106
|
Botta GP, Kato S, Patel H, Fanta P, Lee S, Okamura R, Kurzrock R. SWI/SNF complex alterations as a biomarker of immunotherapy efficacy in pancreatic cancer. JCI Insight 2021; 6:e150453. [PMID: 34375311 PMCID: PMC8492298 DOI: 10.1172/jci.insight.150453] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/04/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUNDImmune checkpoint inhibitors (ICIs) fail to demonstrate efficacy in pancreatic cancer. Recently, genomic biomarkers have been associated with response to ICIs: microsatellite instability high (MSI-H) and tumor mutation burden (TMB) > 10 mutations/Mb. Alterations in Switch/Sucrose Nonfermentable (SWI/SNF) chromatin remodeling genes may predispose to improved outcomes with immunotherapy. The current study examined a possible role for SWI/SNF complex abnormalities in pancreatic cancer responsiveness to ICIs.METHODSA database of 6831 cancer patients that had undergone next-generation sequencing (NGS) was filtered for advanced pancreatic cancer, SWI/SNF alterations, and outcomes depending on immunotherapy treatment.RESULTSNine patients had metastatic pancreatic adenocarcinoma harboring SWI/SNF chromatin remodeling gene alterations and had received ICIs: 7 had an ARID1A alteration (77%); 2, ARID1B (22%); 3, SMARCA4 (33%); 1, SMARCB1 (11%); and 1, PBRM1 (11%). Three patients possessed more than 1 SWI/SNF complex alteration. Only 3 tumors were microsatellite unstable. Eight of 9 patients (89%) achieved an objective response, including a complete remission, with the 2 longest responses ongoing at 33+ and 36+ months. Median progression-free and overall survival was 9 and 15 months, respectively. Responses occurred even in the presence of microsatellite stability, low TMB, and/or low PD-L1 expression.CONCLUSIONA small subset of patients with pancreatic cancer have genomic alterations in SWI/SNF chromatin remodeling components and appear to be responsive to ICIs, suggesting the need for prospective trials.TRIAL REGISTRATIONClinicalTrials.gov, NCT02478931.FUNDINGJoan and Irwin Jacobs Fund, NIH P30 CA023100 (RK) and LRP KYGF9753 (GPB), the Gershenson, Duarte, and anonymous patient families (GPB).
Collapse
Affiliation(s)
- Gregory P. Botta
- Center for Personalized Cancer Therapy, Department of Medicine, and
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy, Department of Medicine, and
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Hitendra Patel
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Paul Fanta
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy, Department of Medicine, and
| | - Ryosuke Okamura
- Department of Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Department of Medicine, and
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
107
|
Gao Y, Lin H, Guo D, Cheng S, Zhou Y, Zhang L, Yao J, Farooq MA, Ajmal I, Duan Y, He C, Tao L, Wu S, Liu M, Jiang W. Suppression of 4.1R enhances the potency of NKG2D-CAR T cells against pancreatic carcinoma via activating ERK signaling pathway. Oncogenesis 2021; 10:62. [PMID: 34548478 PMCID: PMC8455638 DOI: 10.1038/s41389-021-00353-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/12/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic carcinoma (PC) is one of the most common malignancies. Chimeric antigen receptor (CAR)-modified T cells has achieved remarkable efficacy in the treatment of hematological malignancies. However, lack of tumor-specific targets and the existence of inhibitory factors limit the function of CAR T cells when treating solid tumors. 4.1R has been reported to suppress the anti-tumor activity of T cell responses. In this study, we investigated the anti-tumor activity of 4.1R deletion in natural killer group 2D (NKG2D)-CAR T cells against PC. The CAR T cells were obtained by transfecting T cells with lentiviral vector carrying NKG2D-CAR, NC-NKG2D-CAR, or KD2-NKG2D-CAR. In vitro, NKG2D-CAR T cells showed higher cytotoxicity than Mock T cells. However, compared to NKG2D-CAR T cells, furtherly higher cytotoxicity against PC cells in a dose-dependent manner was found in KD2-NKG2D-CAR T cells. In addition, the proliferation rate and cytotoxic activity of KD2-NKG2D-CAR T cells were significantly higher than those of NKG2D-CAR T cells. Besides, the inhibitory receptors PD-1 and TIM-3 were expressed in lower level on KD2-NKG2D-CAR T cells. In vivo, KD2-NKG2D-CAR T cells suppressed tumor growth more effectively in a xenograft model compared to NKG2D-CAR T cells. Mechanistically, 4.1R regulated CAR T cell function via activating ERK signaling pathway. Therefore, the study provides a new idea to enhance the anti-tumor efficiency of CAR T therapy.
Collapse
Affiliation(s)
- Yaoxin Gao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Haizhen Lin
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Dandan Guo
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Sijia Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Ying Zhou
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Li Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Jie Yao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Muhammad Asad Farooq
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Iqra Ajmal
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Yixin Duan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Cong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Lei Tao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Shijia Wu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| |
Collapse
|
108
|
Leblond MM, Zdimerova H, Desponds E, Verdeil G. Tumor-Associated Macrophages in Bladder Cancer: Biological Role, Impact on Therapeutic Response and Perspectives for Immunotherapy. Cancers (Basel) 2021; 13:cancers13184712. [PMID: 34572939 PMCID: PMC8467100 DOI: 10.3390/cancers13184712] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are one of the most abundant infiltrating immune cells of solid tumors. Despite their possible dual role, i.e., pro- or anti-tumoral, there is considerable evidence showing that the accumulation of TAMs promotes tumor progression rather than slowing it. Several strategies are being developed and clinically tested to target these cells. Bladder cancer (BCa) is one of the most common cancers, and despite heavy treatments, including immune checkpoint inhibitors (ICIs), the overall patient survival for advanced BCa is still poor. TAMs are present in bladder tumors and play a significant role in BCa development. However, few investigations have analyzed the effect of targeting TAMs in BCa. In this review, we focus on the importance of TAMs in a cancerous bladder, their association with patient outcome and treatment efficiency as well as on how current BCa treatments impact these cells. We also report different strategies used in other cancer types to develop new immunotherapeutic strategies with the aim of improving BCa management through TAMs targeting.
Collapse
Affiliation(s)
- Marine M. Leblond
- UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, Normandie University, 14000 Caen, France;
| | - Hana Zdimerova
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (H.Z.); (E.D.)
| | - Emma Desponds
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (H.Z.); (E.D.)
| | - Grégory Verdeil
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (H.Z.); (E.D.)
- Correspondence:
| |
Collapse
|
109
|
Wang R, Guo H, Tang X, Zhang T, Liu Y, Zhang C, Yu H, Li Y. Interferon Gamma-Induced Interferon Regulatory Factor 1 Activates Transcription of HHLA2 and Induces Immune Escape of Hepatocellular Carcinoma Cells. Inflammation 2021; 45:308-330. [PMID: 34536158 DOI: 10.1007/s10753-021-01547-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/23/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Immunosuppression developed by cancer cells remains a leading cause of treating failure of immunotherapies. This study aimed to explore the function of human endogenous retrovirus-H long terminal repeat-associating 2 (HHLA2), an immune checkpoint molecule from the B7 family, in the immune escape in hepatocellular carcinoma (HCC). Mouse models with primary HCC or with xenograft tumors were established. The portion of tumor-associated macrophages (TAMs) and the level of PD-L1 in the tumor tissues were examined. THP-1 cells were treated with PMA to obtain a macrophage-like phenotype. The PMA-treated THP-1 cells were co-cultured with the HCC cells in Transwell chambers to examine the function of HHLA2 in chemotactic migration and polarization of macrophages. HHLA2 expression was correlated with infiltration of immune cells, especially macrophages, and was linked to poor prognosis of patients with HCC. HHLA2 knockdown reduced incidence rate of primary HCC in mice. It also reduced tumor metastasis, the portion of M2 macrophages, and the expression of PD-L1 in primary and xenograft tumors. In vitro, HHLA2 upregulation increased expression of PD-L1 in HCC cells indirectly by inducing M2 polarization and chemotactic migration of macrophages. Interferon gamma (IFNG) enhanced expression of interferon regulatory factor 1 (IFR1) in HCC cells, and IFR1 bound to the promoter region of HHLA2 to activate HHLA2 expression. This study suggested that the IFNG/IFR1/HHLA2 axis in HCC induces M2 polarization and chemotactic migration of macrophages, which leads to immune escape and development of HCC.
Collapse
Affiliation(s)
- Rui Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical University, Bengbu, 233004, Anhui, People's Republic of China
| | - Hui Guo
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical University, Bengbu, 233004, Anhui, People's Republic of China
| | - Xiaotong Tang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Tiantian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Yang Liu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Cheng Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical University, Bengbu, 233004, Anhui, People's Republic of China
| | - Hanbing Yu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Yumei Li
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.
| |
Collapse
|
110
|
Reis-Sobreiro M, Teixeira da Mota A, Jardim C, Serre K. Bringing Macrophages to the Frontline against Cancer: Current Immunotherapies Targeting Macrophages. Cells 2021; 10:2364. [PMID: 34572013 PMCID: PMC8464913 DOI: 10.3390/cells10092364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/07/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022] Open
Abstract
Macrophages are found in all tissues and display outstanding functional diversity. From embryo to birth and throughout adult life, they play critical roles in development, homeostasis, tissue repair, immunity, and, importantly, in the control of cancer growth. In this review, we will briefly detail the multi-functional, protumoral, and antitumoral roles of macrophages in the tumor microenvironment. Our objective is to focus on the ever-growing therapeutic opportunities, with promising preclinical and clinical results developed in recent years, to modulate the contribution of macrophages in oncologic diseases. While the majority of cancer immunotherapies target T cells, we believe that macrophages have a promising therapeutic potential as tumoricidal effectors and in mobilizing their surroundings towards antitumor immunity to efficiently limit cancer progression.
Collapse
Affiliation(s)
| | | | | | - Karine Serre
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; (M.R.-S.); (A.T.d.M.); (C.J.)
| |
Collapse
|
111
|
Jung BK, Ko HY, Kang H, Hong J, Ahn HM, Na Y, Kim H, Kim JS, Yun CO. Relaxin-expressing oncolytic adenovirus induces remodeling of physical and immunological aspects of cold tumor to potentiate PD-1 blockade. J Immunother Cancer 2021; 8:jitc-2020-000763. [PMID: 32753544 PMCID: PMC7406118 DOI: 10.1136/jitc-2020-000763] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 02/07/2023] Open
Abstract
Background Currently, several antibody (Ab)-based therapies have shown excellent therapeutic effects in the clinic. Nonetheless, Ab penetration into tumor tissues is limited due to abnormal vasculature, tumor interstitial pressure, and excessive extracellular matrix (ECM) accumulation, thus demanding novel strategies to overcome these barriers. Methods The intratumoral distribution of therapeutic Abs were detected by fluorescence microscopy or positron emission tomography in both human gastric xenograft and syngeneic pancreatic hamster tumor models. The antitumor efficacy by combination of oncolytic adenovirus (Ad), which coexpresses relaxin (RLX), interleukin (IL)-12, and granulocyte macrophage colony-stimulating factor (GM-CSF) (oAd/IL12/GM-RLX) and antibody against the programmed cell death protein 1 (αPD-1) was examined in hamster subcutaneous and orthotopic pancreatic tumor models. The immunological aspects of these combination therapy regimen were assessed by flow cytometry or immunohistochemistry in subcutaneous hamster tumor models. Results Relaxin-expressing oncolytic Ad effectively degraded tumor ECM and enhanced the tumor penetration of trastuzumab in comparison with trastuzumab monotherapy. Based on these results, an oAd/IL12/GM-RLX was used to enhance the potency of immune checkpoint blockade. The combination of the oAd/IL12/GM-RLX and αPD-1 promoted a concomitant degradation of the tumor ECM and amelioration of the immunosuppressive tumor niches, ultimately enhanced intratumoral infiltration of both αPD-1 and activated T cells. Of note, the combination therapy was able to elicit a potent and durable antitumor immune response against cold tumors that were refractory to immune checkpoint inhibitor monotherapy. Conclusions Our findings are the first to demonstrate that expression of four genes (IL-12p35, IL-12p40, GM-CSF, and RLX) mediated by a single oncolytic Ad vector can promote remodeling of both physical and immunological aspects of the tumor niches to overcome the major limitations of Ab-based therapies that have emerged in recent clinical trials.
Collapse
Affiliation(s)
- Bo-Kyeong Jung
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of)
| | - Hae Young Ko
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of)
| | - Hyunji Kang
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of)
| | - JinWoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of).,Department of Research and Development, GeneMedicine Co., Ltd, Seoul, Korea (the Republic of)
| | - Hyo Min Ahn
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of).,Department of Research and Development, GeneMedicine Co., Ltd, Seoul, Korea (the Republic of)
| | - Youjin Na
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of)
| | - Hyeongi Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of)
| | - Jin Su Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, Korea (the Republic of) .,Radiological and Medico-Oncological Sciences, University of science and technology (UST), Seoul, Korea (the Republic of)
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea (the Republic of) .,Department of Research and Development, GeneMedicine Co., Ltd, Seoul, Korea (the Republic of).,Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Korea (the Republic of)
| |
Collapse
|
112
|
Stifter K, Krieger J, Ruths L, Gout J, Mulaw M, Lechel A, Kleger A, Seufferlein T, Wagner M, Schirmbeck R. IFN-γ treatment protocol for MHC-I lo/PD-L1 + pancreatic tumor cells selectively restores their TAP-mediated presentation competence and CD8 T-cell priming potential. J Immunother Cancer 2021; 8:jitc-2020-000692. [PMID: 32868392 PMCID: PMC7462314 DOI: 10.1136/jitc-2020-000692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
Background Many cancer cells express a major histocompatibility complex class I low/ programmed cell death 1 ligand 1 positive (MHC-Ilo/PD-L1+) cell surface profile. For immunotherapy, there is, thus, an urgent need to restore presentation competence of cancer cells with defects in MHC-I processing/presentation combined with immune interventions that tackle the tumor-initiated PD-L1/PD-1 signaling axis. Using pancreatic ductal adenocarcinoma cells (PDACCs) as a model, we here explored if (and how) expression/processing of tumor antigens via transporters associated with antigen processing (TAP) affects priming of CD8 T cells in PD-1/PD-L1-competent/-deficient mice. Methods We generated tumor antigen-expressing vectors, immunized TAP-competent/-deficient mice and determined de novo primed CD8 T-cell frequencies by flow cytometry. Similarly, we explored the antigenicity and PD-L1/PD-1 sensitivity of PDACCs versus interferon-γ (IFN-γ)-treated PDACCs in PD-1/PD-L1-competent/deficient mice. The IFN-γ-induced effects on gene and cell surface expression profiles were determined by microarrays and flow cytometry. Results We identified two antigens (cripto-1 and an endogenous leukemia virus-derived gp70) that were expressed in the Endoplasmic Reticulum (ER) of PDACCs and induced CD8 T-cell responses either independent (Cripto-1:Kb/Cr16-24) or dependent (gp70:Kb/p15E) on TAP by DNA immunization. IFN-γ-treatment of PDACCs in vitro upregulated MHC-I- and TAP- but also PD-L1-expression. Mechanistically, PD-L1/PD-1 signaling was superior to the reconstitution of MHC-I presentation competence, as subcutaneously transplanted IFN-γ-treated PDACCs developed tumors in C57BL/6J and PD-L1-/- but not in PD-1-/- mice. Using PDACCs, irradiated at day 3 post-IFN-γ-treatment or PD-L1 knockout PDACCs as vaccines, we could selectively bypass upregulation of PD-L1, preferentially induce TAP-dependent gp70:Kb/p15E-specific CD8 T cells associated with a weakened PD-1+ exhaustion phenotype and reject consecutively injected tumor transplants in C57BL/6J mice. Conclusions The IFN-γ-treatment protocol is attractive for cell-based immunotherapies, because it restores TAP-dependent antigen processing in cancer cells, facilitates priming of TAP-dependent effector CD8 T-cell responses without additional check point inhibitors and could be combined with genetic vaccines that complement priming of TAP-independent CD8 T cells.
Collapse
Affiliation(s)
- Katja Stifter
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Jana Krieger
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Leonie Ruths
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Johann Gout
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Medhanie Mulaw
- Institute of Experimental Cancer Research, University Hospital Ulm, Ulm, Germany
| | - Andre Lechel
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | | | - Martin Wagner
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | |
Collapse
|
113
|
Li E, Huang X, Zhang G, Liang T. Combinational blockade of MET and PD-L1 improves pancreatic cancer immunotherapeutic efficacy. J Exp Clin Cancer Res 2021; 40:279. [PMID: 34479614 PMCID: PMC8414725 DOI: 10.1186/s13046-021-02055-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/31/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Dysregulated expression and activation of receptor tyrosine kinases (RTKs) are associated with a range of human cancers. However, current RTK-targeting strategies exert little effect on pancreatic cancer, a highly malignant tumor with complex immune microenvironment. Given that immunotherapy for pancreatic cancer still remains challenging, this study aimed to elucidate the prognostic role of RTKs in pancreatic tumors with different immunological backgrounds and investigate their targeting potential in pancreatic cancer immunotherapy. METHODS Kaplan-Meier plotter was used to analyze the prognostic significance of each of the all-known RTKs to date in immune "hot" and "cold" pancreatic cancers. Gene Expression Profiling Interactive Analysis-2 was applied to assess the differential expression of RTKs between pancreatic tumors and normal pancreatic tissues, as well as its correlation with immune checkpoints (ICPs). One hundred and fifty in-house clinical tissue specimens of pancreatic cancer were collected for expression and correlation validation via immunohistochemical analysis. Two pancreatic cancer cell lines were used to demonstrate the regulatory effects of RTKs on ICPs by biochemistry and flow cytometry. Two in vivo models bearing pancreatic tumors were jointly applied to investigate the combinational regimen of RTK inhibition and immune checkpoint blockade for pancreatic cancer immunotherapy. RESULTS MET was identified as a pancreatic cancer-specific RTK, which is significantly associated with prognosis in both immune "hot" and "cold" pancreatic cancers. MET was observed to be highly upregulated in pancreatic cancer tissues, and positively correlated with PD-L1 levels. Elevated MET and PD-L1 expressions were closely associated with lymph node metastasis, tumor TNM stage, and overall survival in pancreatic cancer. Mechanistically, MET could interact with PD-L1, and maintain its expression level in multiple ways. MET deficiency was found to facilitate lymphocyte infiltration into pancreatic tumors. Finally, significant benefits of combining MET inhibition with PD-1/PD-L1 blockage were verified in both orthotopic and subcutaneous mouse models of pancreatic cancer. CONCLUSIONS This study systematically investigated the potential effectiveness of a novel pancreatic cancer immunotherapy targeting RTKs, and revealed the function of MET in PD-L1 regulation as well as the combined therapeutic efficacy of MET and PD-L1 in pancreatic cancer.
Collapse
Affiliation(s)
- Enliang Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310009, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, 310009, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Disease, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310058, Hangzhou, Zhejiang, China
- Research Center for Healthcare Data Science, Zhejiang Lab, 310003, Hangzhou, Zhejiang, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310009, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, 310009, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Disease, 310003, Hangzhou, Zhejiang, China.
- Zhejiang University Cancer Center, 310058, Hangzhou, Zhejiang, China.
- Research Center for Healthcare Data Science, Zhejiang Lab, 310003, Hangzhou, Zhejiang, China.
| | - Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310009, Hangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Innovation Center for the Study of Pancreatic Diseases, 310009, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Disease, 310003, Hangzhou, Zhejiang, China
- Zhejiang University Cancer Center, 310058, Hangzhou, Zhejiang, China
- Research Center for Healthcare Data Science, Zhejiang Lab, 310003, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310009, Hangzhou, Zhejiang, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Innovation Center for the Study of Pancreatic Diseases, 310009, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Disease, 310003, Hangzhou, Zhejiang, China.
- Zhejiang University Cancer Center, 310058, Hangzhou, Zhejiang, China.
- Research Center for Healthcare Data Science, Zhejiang Lab, 310003, Hangzhou, Zhejiang, China.
| |
Collapse
|
114
|
Targeting PI3K Pathway in Pancreatic Ductal Adenocarcinoma: Rationale and Progress. Cancers (Basel) 2021; 13:cancers13174434. [PMID: 34503244 PMCID: PMC8430624 DOI: 10.3390/cancers13174434] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains among the deadliest solid tumors that remain treatment-refractory and show a dismal prognosis. More than 90% of PDAC tumors harbor mutations in the K-Ras that exert a strong pro-tumorigenic effect by activating several downstream effector pathways, including phosphatidylinositol-3-kinase (PI3K)-Akt. The role of frequently activated PI3K/Akt pathway in promoting PDAC aggressiveness is well established. Therapeutic approaches targeting PI3K and downstream signaling components in different cellular compartments, including tumor, stromal and immune cells, have directly impacted the tumor burden in this cancer type. Our previous work has demonstrated that targeting the PI3K/Akt/mTOR pathway reduced tumor growth and improved survival in the genetic mouse model of PDAC. Here, we discuss the significance of targeting PI3K signaling and the biological impact of PI3K inhibition in modulating the tumor-stromal immune crosstalk within the microenvironment of pancreatic cancer. Furthermore, this review updates on the current challenges involving the therapeutic implications of targeting this pathway in PDAC.
Collapse
|
115
|
Gu Z, Du Y, Zhao X, Wang C. Tumor microenvironment and metabolic remodeling in gemcitabine-based chemoresistance of pancreatic cancer. Cancer Lett 2021; 521:98-108. [PMID: 34461181 DOI: 10.1016/j.canlet.2021.08.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid malignant tumor with a very low operative rate and a poor patient prognosis. Therefore, gemcitabine (GEM)-based chemotherapy remains one of the most important treatment choices for PDAC. However, the efficacy of GEM monotherapy or GEM combination chemotherapy in improving the survival of patients with advanced PDAC is very limited, primarily due to GEM resistance. The mechanism of GEM resistance is complex and unclear. An extensive and dense fibrous matrix in the tumor microenvironment (TME) is an important feature of PDAC. Increasing evidence indicates that this fibrotic TME not only actively participates in the growth and spread of PDAC but also contributes to the induction of GEM resistance. Metabolic remodeling reduces GEM transport and synthesis in PDAC. This review focuses on the main cellular and molecular mechanisms underlying the involvement of the extracellular matrix (ECM), immune cells, and metabolic remodeling in the induction of GEM resistance; highlights the prospect of targeting the TME as an essential strategy to overcome GEM resistance; and provides new precise interventions for chemotherapy sensitization and improving the overall prognosis of patients with PDAC.
Collapse
Affiliation(s)
- Zongting Gu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yongxing Du
- Department of Pancreatic and Gastric Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xueping Zhao
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| | - Chengfeng Wang
- Department of Pancreatic and Gastric Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
116
|
Im E, Sim DY, Lee HJ, Park JE, Park WY, Ko S, Kim B, Shim BS, Kim SH. Immune functions as a ligand or a receptor, cancer prognosis potential, clinical implication of VISTA in cancer immunotherapy. Semin Cancer Biol 2021; 86:1066-1075. [PMID: 34428551 DOI: 10.1016/j.semcancer.2021.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 01/15/2023]
Abstract
Since cancer immunotherapy with immune checkpoint inhibitors of PD/PDL-1 and CTLA-4 limited efficacy to the patients due to resistance during the current decade, novel target is required for customized treatment due to tumor heterogeneity. V-domain Ig-containing suppressor of T cell activation (VISTA), a programmed death protein-1(PD-1) homolog expressed on T cells and on antigen presenting cells(APC), has emerged as a new target in several cancers. Though VISTA inhibitors including CA-170 are considered attractive in cancer immunotherapy to date, the information on VISTA as a potent biomarker of cancer prognosis and its combination therapy is still lacking to date. Thus, in this review, we discussed extracellular domain, ligands, expression, immune functions and clinical implications of VISTA and finally suggested conclusion and perspectives.
Collapse
Affiliation(s)
- Eunji Im
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Deok Yong Sim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyo-Jung Lee
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ji Eon Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woon Yi Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - SeongGyu Ko
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bum Sang Shim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
117
|
Genoud V, Espinoza FI, Marinari E, Rochemont V, Dietrich PY, McSheehy P, Bachmann F, Lane HA, Walker PR. Treating ICB-resistant glioma with anti-CD40 and mitotic spindle checkpoint controller BAL101553 (lisavanbulin). JCI Insight 2021; 6:e142980. [PMID: 34403371 PMCID: PMC8492343 DOI: 10.1172/jci.insight.142980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/12/2021] [Indexed: 11/23/2022] Open
Abstract
Glioblastoma is a highly malignant brain tumor with no curative treatment options, and immune checkpoint blockade has not yet shown major impact. We hypothesized that drugs targeting mitosis might affect the tumor microenvironment and sensitize cancer cells to immunotherapy. We used 2 glioblastoma mouse models with different immunogenicity profiles, GL261 and SB28, to test the efficacy of antineoplastic and immunotherapy combinations. The spindle assembly checkpoint activator BAL101553 (lisavanbulin), agonistic anti-CD40 antibody, and double immune checkpoint blockade (anti–programmed cell death 1 and anti–cytotoxic T lymphocyte–associated protein 4; anti–PD-1 and anti–CTLA-4) were evaluated individually or in combination for treating orthotopic GL261 and SB28 tumors. Genomic and immunological analyses were used to predict and interpret therapy responsiveness. BAL101553 monotherapy increased survival in immune checkpoint blockade–resistant SB28 glioblastoma tumors and synergized with anti-CD40 antibody, in a T cell–independent manner. In contrast, the more immunogenic and highly mutated GL261 model responded best to anti–PD-1 and anti–CTLA-4 therapy and more modestly to BAL101553 and anti-CD40 combination. Our results show that BAL101553 is a promising therapeutic agent for glioblastoma and could synergize with innate immune stimulation. Overall, these data strongly support immune profiling of glioblastoma patients and preclinical testing of combination therapies with appropriate models for particular patient groups.
Collapse
Affiliation(s)
- Vassilis Genoud
- Translational Research Center for Hemato-Oncology, University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Felipe I Espinoza
- Translational Research Center for Hemato-Oncology, University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Eliana Marinari
- Translational Research Center for Hemato-Oncology, University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Viviane Rochemont
- Translational Research Center for Hemato-Oncology, University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | | | - Paul McSheehy
- Department of Oncology, Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Felix Bachmann
- Department of Oncology, Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Heidi A Lane
- Department of Oncology, Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Paul R Walker
- Translational Research Center for Hemato-Oncology, University of Geneva, Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
118
|
Byrne KT, Betts CB, Mick R, Sivagnanam S, Bajor DL, Laheru DA, Chiorean EG, O'Hara MH, Liudahl SM, Newcomb C, Alanio C, Ferreira AP, Park BS, Ohtani T, Huffman AP, Väyrynen SA, Dias Costa A, Kaiser JC, Lacroix AM, Redlinger C, Stern M, Nowak JA, Wherry EJ, Cheever MA, Wolpin BM, Furth EE, Jaffee EM, Coussens LM, Vonderheide RH. Neoadjuvant Selicrelumab, an Agonist CD40 Antibody, Induces Changes in the Tumor Microenvironment in Patients with Resectable Pancreatic Cancer. Clin Cancer Res 2021; 27:4574-4586. [PMID: 34112709 PMCID: PMC8667686 DOI: 10.1158/1078-0432.ccr-21-1047] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/29/2021] [Accepted: 05/28/2021] [Indexed: 01/09/2023]
Abstract
PURPOSE CD40 activation is a novel clinical opportunity for cancer immunotherapy. Despite numerous active clinical trials with agonistic CD40 monoclonal antibodies (mAb), biological effects and treatment-related modulation of the tumor microenvironment (TME) remain poorly understood. PATIENTS AND METHODS Here, we performed a neoadjuvant clinical trial of agonistic CD40 mAb (selicrelumab) administered intravenously with or without chemotherapy to 16 patients with resectable pancreatic ductal adenocarcinoma (PDAC) before surgery followed by adjuvant chemotherapy and CD40 mAb. RESULTS The toxicity profile was acceptable, and overall survival was 23.4 months (95% confidence interval, 18.0-28.8 months). Based on a novel multiplexed immunohistochemistry platform, we report evidence that neoadjuvant selicrelumab leads to major differences in the TME compared with resection specimens from treatment-naïve PDAC patients or patients given neoadjuvant chemotherapy/chemoradiotherapy only. For selicrelumab-treated tumors, 82% were T-cell enriched, compared with 37% of untreated tumors (P = 0.004) and 23% of chemotherapy/chemoradiation-treated tumors (P = 0.012). T cells in both the TME and circulation were more active and proliferative after selicrelumab. Tumor fibrosis was reduced, M2-like tumor-associated macrophages were fewer, and intratumoral dendritic cells were more mature. Inflammatory cytokines/sec CXCL10 and CCL22 increased systemically after selicrelumab. CONCLUSIONS This unparalleled examination of CD40 mAb therapeutic mechanisms in patients provides insights for design of subsequent clinical trials targeting CD40 in cancer.
Collapse
Affiliation(s)
- Katelyn T Byrne
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Courtney B Betts
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute, Oregon Health and Science University-Portland State University School of Public Health, Portland, Oregon
| | - Rosemarie Mick
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shamilene Sivagnanam
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | | | - Daniel A Laheru
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - E Gabriela Chiorean
- University of Washington School of Medicine, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark H O'Hara
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shannon M Liudahl
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Craig Newcomb
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cécile Alanio
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ana P Ferreira
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Byung S Park
- Knight Cancer Institute, Oregon Health and Science University-Portland State University School of Public Health, Portland, Oregon
| | - Takuya Ohtani
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Austin P Huffman
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara A Väyrynen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | - Colleen Redlinger
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Martin Stern
- Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Jonathan A Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - E John Wherry
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Emma E Furth
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - 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 State University School of Public Health, Portland, Oregon
| | - Robert H Vonderheide
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
119
|
Kandikattu HK, Venkateshaiah SU, Mishra A. Chronic Pancreatitis and the Development of Pancreatic Cancer. Endocr Metab Immune Disord Drug Targets 2021; 20:1182-1210. [PMID: 32324526 DOI: 10.2174/1871530320666200423095700] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/31/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Pancreatitis is a fibro-inflammatory disorder of the pancreas that can occur acutely or chronically as a result of the activation of digestive enzymes that damage pancreatic cells, which promotes inflammation. Chronic pancreatitis with persistent fibro-inflammation of the pancreas progresses to pancreatic cancer, which is the fourth leading cause of cancer deaths across the globe. Pancreatic cancer involves cross-talk of inflammatory, proliferative, migratory, and fibrotic mechanisms. In this review, we discuss the role of cytokines in the inflammatory cell storm in pancreatitis and pancreatic cancer and their role in the activation of SDF1α/CXCR4, SOCS3, inflammasome, and NF-κB signaling. The aberrant immune reactions contribute to pathological damage of acinar and ductal cells, and the activation of pancreatic stellate cells to a myofibroblast-like phenotype. We summarize several aspects involved in the promotion of pancreatic cancer by inflammation and include a number of regulatory molecules that inhibit that process.
Collapse
Affiliation(s)
- Hemanth K Kandikattu
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Sathisha U Venkateshaiah
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Anil Mishra
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
120
|
Del Re M, Vivaldi C, Rofi E, Salani F, Crucitta S, Catanese S, Fontanelli L, Massa V, Cucchiara F, Fornaro L, Capuano A, Fogli S, Vasile E, Danesi R. Gemcitabine Plus Nab-Paclitaxel Induces PD-L1 mRNA Expression in Plasma-Derived Microvesicles in Pancreatic Cancer. Cancers (Basel) 2021; 13:3738. [PMID: 34359638 PMCID: PMC8345069 DOI: 10.3390/cancers13153738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/04/2021] [Accepted: 07/12/2021] [Indexed: 01/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a non-immunogenic tumor poorly responsive to immune checkpoint inhibitors. This study investigates the effect of 5-fluorouracil (5-FU), irinotecan, and oxaliplatin (FOLFIRINOX), and gemcitabine plus nab-paclitaxel (GEMnPAC) regimens on PD-L1 mRNA expression in plasma-derived microvesicles (MVs) in 50 PDAC patients. Plasma was collected before starting chemotherapy and after 3 months of treatment. mRNA was extracted from MVs, and PD-L1 expression was measured by digital droplet PCR. Twenty-eight patients were PD-L1 positive in MVs at baseline, of which 18 were in the GEMnPAC cohort and 10 in the FOLFIRINOX one. The amount of PD-L1 expression in MVs increased from baseline to 3 months of treatment in patients receiving GEMnPAC (median value 0.002 vs. 0.005; p = 0.01) compared to those treated with FOLFIRINOX (median 0.003 vs. 0.004; p = 0.97). The increase in PD-L1 mRNA expression in MVs was not related to tumor response (PR + SD: p = 0.08; PD: p = 0.28). Our findings demonstrate that GEMnPAC can increase PD-L1 mRNA expression in patient-derived circulating MVs, providing a rationale for testing the efficacy of this regimen in sequential or simultaneous combinations with immunotherapy in PDAC patients.
Collapse
Affiliation(s)
- Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.R.); (E.R.); (S.C.); (L.F.); (F.C.); (S.F.)
| | - Caterina Vivaldi
- Department of Translational Research and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy; (C.V.); (F.S.); (S.C.); (V.M.)
| | - Eleonora Rofi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.R.); (E.R.); (S.C.); (L.F.); (F.C.); (S.F.)
| | - Francesca Salani
- Department of Translational Research and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy; (C.V.); (F.S.); (S.C.); (V.M.)
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.R.); (E.R.); (S.C.); (L.F.); (F.C.); (S.F.)
| | - Silvia Catanese
- Department of Translational Research and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy; (C.V.); (F.S.); (S.C.); (V.M.)
| | - Lorenzo Fontanelli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.R.); (E.R.); (S.C.); (L.F.); (F.C.); (S.F.)
| | - Valentina Massa
- Department of Translational Research and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy; (C.V.); (F.S.); (S.C.); (V.M.)
| | - Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.R.); (E.R.); (S.C.); (L.F.); (F.C.); (S.F.)
| | - Lorenzo Fornaro
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (L.F.); (E.V.)
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80100 Naples, Italy;
| | - Stefano Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.R.); (E.R.); (S.C.); (L.F.); (F.C.); (S.F.)
| | - Enrico Vasile
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (L.F.); (E.V.)
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.R.); (E.R.); (S.C.); (L.F.); (F.C.); (S.F.)
| |
Collapse
|
121
|
Myeloid Cell Mediated Immune Suppression in Pancreatic Cancer. Cell Mol Gastroenterol Hepatol 2021; 12:1531-1542. [PMID: 34303882 PMCID: PMC8529393 DOI: 10.1016/j.jcmgh.2021.07.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA), the most common pancreatic cancer, is a nearly universally lethal malignancy. PDA is characterized by extensive infiltration of immunosuppressive myeloid cells, including tumor-associated macrophages and myeloid-derived suppressor cells. Myeloid cells in the tumor microenvironment inhibit cytotoxic T-cell responses promoting carcinogenesis. Immune checkpoint therapy has not been effective in PDA, most likely because of this robust immune suppression, making it critical to elucidate mechanisms behind this phenomenon. Here, we review myeloid cell infiltration and cellular crosstalk in PDA progression and highlight current therapeutic approaches to target myeloid cell-driven immune suppression.
Collapse
Key Words
- adm, acinar to ductal metaplasia
- csf1r, colony-stimulating factor 1 receptor
- ctla-4, cytotoxic t lymphocyte antigen 4
- egfr, epidermal growth factor receptor
- gm-csf, granulocyte-macrophage colony-stimulating factor
- hb-egf, heparin-binding egf-like growth factor
- ikk, inhibitory κb kinase
- il, interleukin
- mapk, mitogen-activated protein kinase
- mdsc, myeloid-derived suppressor cell
- m-mdsc, mononuclear myeloid-derived suppressor cell
- nf-κb, nuclear factor kappa b
- panin, pancreatic intraepithelial neoplasia
- pda, pancreatic ductal adenocarcinoma
- pd-1, programmed cell death
- pmn, polymorphonuclear
- tam, tumor-associated macrophage
- tme, tumor microenvironment
- tnf, tumor necrosis factor
Collapse
|
122
|
Gunaydin G. CAFs Interacting With TAMs in Tumor Microenvironment to Enhance Tumorigenesis and Immune Evasion. Front Oncol 2021; 11:668349. [PMID: 34336660 PMCID: PMC8317617 DOI: 10.3389/fonc.2021.668349] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer associated fibroblasts (CAFs) and tumor associated macrophages (TAMs) are among the most important and abundant players of the tumor microenvironment. CAFs as well as TAMs are known to play pivotal supportive roles in tumor growth and progression. The number of CAF or TAM cells is mostly correlated with poor prognosis. Both CAFs and TAMs are in a reciprocal communication with the tumor cells in the tumor milieu. In addition to such interactions, CAFs and TAMs are also involved in a dynamic and reciprocal interrelationship with each other. Both CAFs and TAMs are capable of altering each other's functions. Here, the current understanding of the distinct mechanisms about the complex interplay between CAFs and TAMs are summarized. In addition, the consequences of such a mutual relationship especially for tumor progression and tumor immune evasion are highlighted, focusing on the synergistic pleiotropic effects. CAFs and TAMs are crucial components of the tumor microenvironment; thus, they may prove to be potential therapeutic targets. A better understanding of the tri-directional interactions of CAFs, TAMs and cancer cells in terms of tumor progression will pave the way for the identification of novel theranostic cues in order to better target the crucial mechanisms of carcinogenesis.
Collapse
Affiliation(s)
- Gurcan Gunaydin
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
123
|
Awadasseid A, Wu Y, Zhang W. Advance investigation on synthetic small-molecule inhibitors targeting PD-1/PD-L1 signaling pathway. Life Sci 2021; 282:119813. [PMID: 34256042 DOI: 10.1016/j.lfs.2021.119813] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 06/27/2021] [Accepted: 07/06/2021] [Indexed: 01/01/2023]
Abstract
Immune checkpoint blockade has displayed substantial anti-tumor resistance in a variety of forms of cancer, but the fundamental regulation role remains unclear, and several questions continue to be addressed. PD-1/PD-L1 has been recognized as an anti-cancer drug target for several years, and through targeting the PD-1/PD-L1 signaling pathway, many monoclonal antibodies have thus far produced promising results in cancer therapy. The discovery of small-molecule inhibitors focused on the PD-1/PD-L1 signaling pathway is steadily reviving over decades, owing to the intrinsic shortcomings of the antibodies. PD-1 function and its PD-L1 or PD-L2 ligands are essential for the activation, proliferation, and cytotoxic secretion of T-cells in cancer to degenerate anti-tumor immune response. The axis PD-1/PD-L1 is important for the immune escape of cancer which has an immense impact on cancer treatment. In this review, we summarize the function of PD-1 and PD-L1 in cancer and aiming to enhance cancer therapy.
Collapse
Affiliation(s)
- Annoor Awadasseid
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China; Department of Biochemistry & Food Sciences, University of Kordofan, El-Obeid 51111, Sudan
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China.
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
124
|
Filbert EL, Björck PK, Srivastava MK, Bahjat FR, Yang X. APX005M, a CD40 agonist antibody with unique epitope specificity and Fc receptor binding profile for optimal therapeutic application. Cancer Immunol Immunother 2021; 70:1853-1865. [PMID: 33392713 PMCID: PMC8195934 DOI: 10.1007/s00262-020-02814-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022]
Abstract
Targeting CD40 with agonist antibodies is a promising approach to cancer immunotherapy. CD40 acts as a master regulator of immunity by mobilizing multiple arms of the immune system to initiate highly effective CD8 + T-cell-mediated responses against foreign pathogens and tumors. The clinical development of CD40 agonist antibodies requires careful optimization of the antibody to maximize therapeutic efficacy while minimizing adverse effects. Both epitope specificity and isotype are critical for CD40 agonist antibody mechanism of action and potency. We developed a novel antibody, APX005M, which binds with high affinity to the CD40 ligand-binding site on CD40 and is optimized for selective interaction with Fcγ receptors to enhance agonistic potency while limiting less desirable Fc-effector functions like antibody-dependent cellular cytotoxicity of CD40-expressing immune cells. APX005M is a highly potent inducer of innate and adaptive immune effector responses and represents a promising CD40 agonist antibody for induction of an effective anti-tumor immune response with a favorable safety profile.
Collapse
Affiliation(s)
- Erin L Filbert
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Pia K Björck
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Minu K Srivastava
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Frances R Bahjat
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA
| | - Xiaodong Yang
- Apexigen, Inc, 75 Shoreway Road, Suite C, San Carlos, CA, 94070, USA.
| |
Collapse
|
125
|
Sasaki K, Takano S, Tomizawa S, Miyahara Y, Furukawa K, Takayashiki T, Kuboki S, Takada M, Ohtsuka M. C4b-binding protein α-chain enhances antitumor immunity by facilitating the accumulation of tumor-infiltrating lymphocytes in the tumor microenvironment in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:212. [PMID: 34167573 PMCID: PMC8228942 DOI: 10.1186/s13046-021-02019-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023]
Abstract
Background Recent studies indicate that complement plays pivotal roles in promoting or suppressing cancer progression. We have previously identified C4b-binding protein α-chain (C4BPA) as a serum biomarker for the early detection of pancreatic ductal adenocarcinoma (PDAC). However, its mechanism of action remains unclear. Here, we elucidated the functional roles of C4BPA in PDAC cells and the tumor microenvironment. Methods We assessed stromal C4BPA, the C4BPA binding partner CD40, and the number of CD8+ tumor-infiltrating lymphocytes in resected human PDAC tissues via immunohistochemical staining. The biological functions of C4BPA were investigated in peripheral blood mononuclear cells (PBMCs) and human PDAC cell lines. Mouse C4BPA (mC4BPA) peptide, which is composed of 30 amino acids from the C-terminus and binds to CD40, was designed for further in vitro and in vivo experiments. In a preclinical experiment, we assessed the efficacy of gemcitabine plus nab-paclitaxel (GnP), dual immune checkpoint blockades (ICBs), and mC4BPA peptide in a mouse orthotopic transplantation model. Results Immunohistochemical analysis revealed that high stromal C4BPA and CD40 was associated with favorable PDAC prognosis (P=0.0005). Stromal C4BPA strongly correlated with the number of CD8+ tumor-infiltrating lymphocytes (P=0.001). In in vitro experiments, flow cytometry revealed that recombinant human C4BPA (rhC4BPA) stimulation increased CD4+ and CD8+ T cell numbers in PBMCs. rhC4BPA also promoted the proliferation of CD40-expressing PDAC cells. By contrast, combined treatment with gemcitabine and rhC4BPA increased PDAC cell apoptosis rate. mC4BPA peptide increased the number of murine T lymphocytes in vitro and the number of CD8+ tumor-infiltrating lymphocytes surrounding PDAC tumors in vivo. In a preclinical study, GnP/ICBs/mC4BPA peptide treatment, but not GnP treatment, led to the accumulation of a greater number of CD8+ T cells in the periphery of PDAC tumors and to greater tumor regression than did control treatment. Conclusions These findings demonstrate that the combination of GnP therapy with C4BPA inhibits PDAC progression by promoting antitumor T cell accumulation in the tumor microenvironment. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02019-0.
Collapse
Affiliation(s)
- Kosuke Sasaki
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| | - Shigetsugu Takano
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan.
| | - Satoshi Tomizawa
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| | - Yoji Miyahara
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| | - Katsunori Furukawa
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| | - Tsukasa Takayashiki
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| | - Satoshi Kuboki
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| | - Mamoru Takada
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba City, Chiba, 260-8677, Japan
| |
Collapse
|
126
|
Combination chemotherapeutic and immune-therapeutic anticancer approach via anti-PD-L1 antibody conjugated albumin nanoparticles. Int J Pharm 2021; 605:120816. [PMID: 34161810 DOI: 10.1016/j.ijpharm.2021.120816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/28/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023]
Abstract
Anticancer regimens have been substantially enriched through monoclonal antibodies targeting immune checkpoints, programmed cell death-1/programmed cell death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen-4. Inconsistent clinical efficacy after solo immunotherapy may be compensated by nanotechnology-driven combination therapy. We loaded human serum albumin (HSA) nanoparticles with paclitaxel (PTX) via nanoparticle albumin-bound technology and pooled them with anti-PD-L1 monoclonal antibody through a pH-sensitive linker for targeting and immune response activation. Our tests demonstrated satisfactory preparation of paclitaxel-loaded, PD-L1-targeted albumin nanoparticles (PD-L1/PTX@HSA). They had small particle size (~200 nm) and polydispersity index (~0.12) and successfully incorporated each constituent. Relative to normal physiological pH, the formulation exhibited higher drug-release profiles favoring cancer cell-targeted release at low pH. Modifying nanoparticles with programmed cell death-ligand 1 increased cancer cell internalization in vitro and tumor accumulation in vivo in comparison with non-PD-L1-modified nanoparticles. PD-L1/PTX@HSA constructed by nanoparticle albumin-bound technology displayed successful tumor inhibition efficacy both in vitro and in vivo. There was successful effector T-cell infiltration, immunosuppressive programmed cell death-ligand 1, and regulatory T-cell suppression because of cytotoxic T-lymphocyte antigen-4 synergy. Moreover, PD-L1/PTX@HSA had low organ toxicity. Hence, the anti-tumor immune responses of PD-L1/PTX@HSA combined with chemotherapy and cytotoxic T-lymphocyte antigen-4 is a potential anti-tumor strategy for improving quantitative and qualitative clinical efficacy.
Collapse
|
127
|
Abstract
SummaryAt the ESMO (European Society for Medical Oncology) 2020 several interesting albeit not practice-changing studies in the field of pancreatic cancer were presented. The Canadian phase II randomized PA.7 trial investigated the additional benefit of dual checkpoint inhibition with durvalumab and tremelimumab to a standard chemotherapy regimen as first-line treatment in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC). Unfortunately, no significant improvement of responses or outcome could be achieved rendering this study a negative trial. Within the German platform-based QoliXane trial, quality of life was shown to be an essential prognosticator of survival with fatigue and nausea being independently associated with outcome of patients. Moreover, promising results could be observed with new targeted therapy approaches, which may lead to its investigation in larger randomized clinical trials.
Collapse
|
128
|
Hani U, Osmani RAM, Siddiqua A, Wahab S, Batool S, Ather H, Sheraba N, Alqahtani A. A systematic study of novel drug delivery mechanisms and treatment strategies for pancreatic cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
129
|
Huang J, Chen P, Liu K, Liu J, Zhou B, Wu R, Peng Q, Liu ZX, Li C, Kroemer G, Lotze M, Zeh H, Kang R, Tang D. CDK1/2/5 inhibition overcomes IFNG-mediated adaptive immune resistance in pancreatic cancer. Gut 2021; 70:890-899. [PMID: 32816920 DOI: 10.1136/gutjnl-2019-320441] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 07/03/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Adaptive immune resistance mediated by the cytokine interferon gamma (IFNG) still constitutes a major problem in cancer immunotherapy. We develop strategies for overcoming IFNG-mediated adaptive immune resistance in pancreatic ductal adenocarcinoma cancer (PDAC). DESIGN We screened 429 kinase inhibitors for blocking IFNG-induced immune checkpoint (indoleamine 2,3-dioxygenase 1 (IDO1) and CD274) expression in a human PDAC cell line. We evaluated the ability of the cyclin-dependent kinase (CDK) inhibitor dinaciclib to block IFNG-induced IDO1 and CD274 expression in 24 human and mouse cancer cell lines as well as in primary cancer cells from patients with PDAC or ovarian carcinoma. We tested the effects of dinaciclib on IFNG-induced signal transducer and activator of transcription 1 activation and immunological cell death, and investigated the potential utility of dinaciclib in combination with IFNG for pancreatic cancer therapy in vivo, and compared gene expression levels between human cancer tissues with patient survival times using the Cancer Genome Atlas datasets. RESULTS Pharmacological (using dinaciclib) or genetic (using shRNA or siRNA) inactivation of CDK1/2/5 not only blocks JUN-dependent immune checkpoint expression, but also triggers histone-dependent immunogenic cell death in immortalised or primary cancer cells in response to IFNG. This dual mechanism turns an immunologically 'cold' tumour microenvironment into a 'hot' one, dramatically improving overall survival rates in mouse pancreatic tumour models (subcutaneous, orthotopic and transgenic models). The abnormal expression of CDK1/2/5 and IDO1 was associated with poor patient survival in several cancer types, including PDAC. CONCLUSION CDK1/2/5 kinase activity is essential for IFNG-mediated cancer immunoevasion. CDK1/2/5 inhibition by dinaciclib provides a novel strategy to overcome IFNG-triggered acquired resistance in pancreatic tumour immunity.
Collapse
Affiliation(s)
- Jin Huang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pan Chen
- Department of Hepatobiliary Surgery, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Ke Liu
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jiao Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Borong Zhou
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Runliu Wu
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Qiu Peng
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ze-Xian Liu
- Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Changfeng Li
- Endoscopy Center, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Guido Kroemer
- Equipe Labellisée Par la Ligue Contre le Cancer, Université Paris Descartes, Paris, Île-de-France, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Biology, Chinese Academy of Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Herbert Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daolin Tang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China .,Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
130
|
Winograd R, Simeone DM, Bar-Sagi D. A novel target for combination immunotherapy in pancreatic cancer: IL-1β mediates immunosuppression in the tumour microenvironment. Br J Cancer 2021; 124:1754-1756. [PMID: 33758330 PMCID: PMC8144204 DOI: 10.1038/s41416-021-01303-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/13/2021] [Accepted: 02/02/2021] [Indexed: 01/07/2023] Open
Abstract
Immune checkpoint blockade (ICB) has demonstrated efficacy in multiple cancers, offering the potential of long-term disease control not achievable with cytotoxic or targeted therapies. However, the field has not yet achieved the crucial next steps - the expansion of the response rate and achievement of clinical efficacy in so-called "cold tumours". Mechanistic studies of tumour-type specific immunosuppressive pathways can reveal underlying biological hurdles to immunotherapy and offer new therapeutic insights. Our finding that tumour-derived IL-1β mediates immunosuppression in pancreatic cancer has precipitated a new clinical trial.
Collapse
Affiliation(s)
- Rafael Winograd
- Department of Hematology and Oncology, NYU Langone Health, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Diane M Simeone
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Langone Health, New York, NY, USA
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
131
|
Gu ZT, Li ZZ, Wang CF. Advances in research of extracellular mechanisms underlying gemcitabine resistance in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2021; 29:421-434. [DOI: 10.11569/wcjd.v29.i8.421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a solid malignant tumor with the worst prognosis worldwide, and about 90% of cases are pancreatic ductal adenocarcinoma (PDAC). Although surgical resection is the only potential way to cure PDAC, the overall survival rate after surgery is still not optimistic. Consequently, gemcitabine (GEM)-based chemotherapy is still one of the most important treatment options for PDAC. However, the survival improvement by GEM monotherapy for advanced PDAC is very limited, and GEM resistance is the key reason. The mechanism underlying gemcitabine resistance is complex and still unclear in PDAC. The extensive and dense fibrous mesenchyme in the tumor microenvironment (TME) is an important feature of PDAC. More and more evidence has shown that TME is not only an active participant in tumor growth and spread, but also a contributor to the induction of GEM resistance. This article will review the recent advances in the understanding of the cellular and molecular mechanisms underlying GEM resistance in PDAC, and discuss potential GEM chemosensitization strategies, in order to improve the effective rate of chemotherapy and the outcome.
Collapse
Affiliation(s)
- Zong-Ting Gu
- Cheng-Feng Wang, State Key Laboratory of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zong-Ze Li
- Cheng-Feng Wang, State Key Laboratory of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | | |
Collapse
|
132
|
Labiano S, Roh V, Godfroid C, Hiou-Feige A, Romero J, Sum E, Rapp M, Boivin G, Wyss T, Simon C, Bourhis J, Umaña P, Trumpfheller C, Tolstonog GV, Vozenin MC, Romero P. CD40 Agonist Targeted to Fibroblast Activation Protein α Synergizes with Radiotherapy in Murine HPV-Positive Head and Neck Tumors. Clin Cancer Res 2021; 27:4054-4065. [PMID: 33903200 DOI: 10.1158/1078-0432.ccr-20-4717] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/13/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE The incidence of human papillomavirus-associated head and neck squamous cell carcinoma (HPV+-HNSCC) is rising worldwide and although current therapeutic modalities are efficient in the majority of patients, there is a high rate of treatment failures. Thus, novel combination approaches are urgently needed to achieve better disease control in patients with HPV+-HNSCC. We investigated the safety and therapeutic efficacy of a novel fibroblast activation protein (FAP)-targeted CD40 agonist (FAP-CD40) in combination with local hypofractionated radiation in a syngeneic HPV+-HNSCC model. EXPERIMENTAL DESIGN Using an established orthotopic model, we treated tumor-bearing mice with local hypofractionated radiotherapy (2 × 6 Gy) alone or in combination with a systemic administration of the FAP-CD40 antibody. Following up the mice, we evaluated the changes in the tumor microenvironment (TME) by immunofluorescence, FACS, and NanoString RNA analysis. RESULTS The suboptimal radiotherapy regimen chosen failed to control tumors in the treated mice. The FAP-CD40 administered in monotherapy transiently controlled tumor growth, whereas the combined therapy induced durable complete responses in more than 80% of the tumor-bearing mice. This notable efficacy relied on the radiotherapy-induced remodeling of the TME and activation of the CD8+ T-cell-cDC1 axis and was devoid of the systemic toxicity frequently associated with CD40-targeted therapy. Moreover, the robust immunologic memory developed effectively prevented tumor relapses, a common feature in patients with HNSCC. CONCLUSIONS Our study provides proof of concept, as well as mechanistic insights of the therapeutic efficacy of a bispecific FAP-CD40 combined with local radiotherapy in a FAP+-HNSCC model increasing overall survival and inducing long-term antitumor immunity.
Collapse
Affiliation(s)
- Sara Labiano
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
| | - Vincent Roh
- Department of Otolaryngology - Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Céline Godfroid
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
| | - Agnès Hiou-Feige
- Department of Otolaryngology - Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jackeline Romero
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eva Sum
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Schlieren, Switzerland
| | - Moritz Rapp
- Roche Innovation Center Munich (RICM), pRED, Penzberg, Germany
| | - Gael Boivin
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Tania Wyss
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Christian Simon
- Department of Otolaryngology - Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jean Bourhis
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Schlieren, Switzerland
| | - Christine Trumpfheller
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Schlieren, Switzerland
| | - Genrich V Tolstonog
- Department of Otolaryngology - Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology. Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pedro Romero
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
133
|
Isvoranu G, Surcel M, Munteanu AN, Bratu OG, Ionita-Radu F, Neagu MT, Chiritoiu-Butnaru M. Therapeutic potential of interleukin-15 in cancer (Review). Exp Ther Med 2021; 22:675. [PMID: 33986840 PMCID: PMC8112152 DOI: 10.3892/etm.2021.10107] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Abstract
The immune system is dysfunctional in cancer, and therapeutic approaches designated to restore immunity and increase long-term overall survival are desirable. The role of immunotherapy is to trigger the immune system to recognize and destroy tumor cells. Interleukin-15 (IL-15) is a member of the common gamma-chain (γc) cytokines that promote the differentiation and expansion of T cells, B cells and natural killer (NK) cells, leading to enhanced antitumor responses. This suggests that IL-15 is a promising candidate for anticancer therapy. Renewed interest in cancer immunotherapy has led to an increased number of preclinical studies and clinical trials that have investigated the reliability and potency of IL-15-based agents, not only as single therapy, but also in combination with others. This review provides a description of these studies which show the advantages and disadvantages of IL-15 as an immunotherapeutic agent. We present here the role of IL-15 and pharmacologically improved IL-15 superagonists as a single treatment or in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Gheorghita Isvoranu
- Department of Animal Husbandry, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Surcel
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Adriana Narcisa Munteanu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Ovidiu Gabriel Bratu
- Department of Clinical Department III, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Clinic of Urology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania.,Academy of Romanian Scientists, 050094 Bucharest, Romania
| | - Florentina Ionita-Radu
- Department of Gastroenterology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania
| | - Monica Teodora Neagu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Marioara Chiritoiu-Butnaru
- Department of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy (IBAR), 060031 Bucharest, Romania
| |
Collapse
|
134
|
Singh K, Lin J, Lecomte N, Mohan P, Gokce A, Sanghvi VR, Jiang M, Grbovic-Huezo O, Burčul A, Stark SG, Romesser PB, Chang Q, Melchor JP, Beyer RK, Duggan M, Fukase Y, Yang G, Ouerfelli O, Viale A, de Stanchina E, Stamford AW, Meinke PT, Rätsch G, Leach SD, Ouyang Z, Wendel HG. Targeting eIF4A-Dependent Translation of KRAS Signaling Molecules. Cancer Res 2021; 81:2002-2014. [PMID: 33632898 PMCID: PMC8137674 DOI: 10.1158/0008-5472.can-20-2929] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/01/2020] [Accepted: 02/22/2021] [Indexed: 11/16/2022]
Abstract
Pancreatic adenocarcinoma (PDAC) epitomizes a deadly cancer driven by abnormal KRAS signaling. Here, we show that the eIF4A RNA helicase is required for translation of key KRAS signaling molecules and that pharmacological inhibition of eIF4A has single-agent activity against murine and human PDAC models at safe dose levels. EIF4A was uniquely required for the translation of mRNAs with long and highly structured 5' untranslated regions, including those with multiple G-quadruplex elements. Computational analyses identified these features in mRNAs encoding KRAS and key downstream molecules. Transcriptome-scale ribosome footprinting accurately identified eIF4A-dependent mRNAs in PDAC, including critical KRAS signaling molecules such as PI3K, RALA, RAC2, MET, MYC, and YAP1. These findings contrast with a recent study that relied on an older method, polysome fractionation, and implicated redox-related genes as eIF4A clients. Together, our findings highlight the power of ribosome footprinting in conjunction with deep RNA sequencing in accurately decoding translational control mechanisms and define the therapeutic mechanism of eIF4A inhibitors in PDAC. SIGNIFICANCE: These findings document the coordinate, eIF4A-dependent translation of RAS-related oncogenic signaling molecules and demonstrate therapeutic efficacy of eIF4A blockade in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Kamini Singh
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jianan Lin
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| | - Nicolas Lecomte
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Prathibha Mohan
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Askan Gokce
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Viraj R Sanghvi
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Man Jiang
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Olivera Grbovic-Huezo
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Antonija Burčul
- Computational Biology Department, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Stefan G Stark
- Computational Biology Department, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Computer Science, Biomedical Informatics, ETH, Zürich, Zürich, Switzerland
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Qing Chang
- Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jerry P Melchor
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Rachel K Beyer
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Mark Duggan
- Tri-Institutional Drug Development Initiative, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Yoshiyuki Fukase
- Tri-Institutional Drug Development Initiative, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Guangli Yang
- The Organic Synthesis Core Facility, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ouathek Ouerfelli
- The Organic Synthesis Core Facility, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Agnes Viale
- Integrated Genomics Operation, Center for Molecular Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Andrew W Stamford
- Tri-Institutional Drug Development Initiative, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Peter T Meinke
- Tri-Institutional Drug Development Initiative, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Gunnar Rätsch
- Computational Biology Department, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Computer Science, Biomedical Informatics, ETH, Zürich, Zürich, Switzerland
| | - Steven D Leach
- Molecular Systems Biology and Surgery, Geisel School of Medicine, Dartmouth, Norris Cotton Cancer Center at Dartmouth-Hitchcock, Lebanon, New Hampshire
| | - Zhengqing Ouyang
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts
| | - Hans-Guido Wendel
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
135
|
Rai ZL, Feakins R, Pallett LJ, Manas D, Davidson BR. Irreversible Electroporation (IRE) in Locally Advanced Pancreatic Cancer: A Review of Current Clinical Outcomes, Mechanism of Action and Opportunities for Synergistic Therapy. J Clin Med 2021; 10:1609. [PMID: 33920118 PMCID: PMC8068938 DOI: 10.3390/jcm10081609] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Locally advanced pancreatic cancer (LAPC) accounts for 30% of patients with pancreatic cancer. Irreversible electroporation (IRE) is a novel cancer treatment that may improve survival and quality of life in LAPC. This narrative review will provide a perspective on the clinical experience of pancreas IRE therapy, explore the evidence for the mode of action, assess treatment complications, and propose strategies for augmenting IRE response. A systematic search was performed using PubMed regarding the clinical use and safety profile of IRE on pancreatic cancer, post-IRE sequential histological changes, associated immune response, and synergistic therapies. Animal data demonstrate that IRE induces both apoptosis and necrosis followed by fibrosis. Major complications may result from IRE; procedure related mortality is up to 2%, with an average morbidity as high as 36%. Nevertheless, prospective and retrospective studies suggest that IRE treatment may increase median overall survival of LAPC to as much as 30 months and provide preliminary data justifying the well-designed trials currently underway, comparing IRE to the standard of care treatment. The mechanism of action of IRE remains unknown, and there is a lack of data on treatment variables and efficiency in humans. There is emerging data suggesting that IRE can be augmented with synergistic therapies such as immunotherapy.
Collapse
Affiliation(s)
- Zainab L. Rai
- Centre of Surgical Innovation, Organ Regeneration and Transplantation, University College London (UCL), London NW3 2QG, UK;
- Wellcome/EPSRC Center for Interventional and Surgical Sciences (WEISS), London W1W 7TY, UK
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| | - Roger Feakins
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| | - Laura J. Pallett
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London WC1E 6BT, UK;
| | - Derek Manas
- Newcastle Upon Tyne NHS Foundation Trust, Newcastle-Upon-Tyne NE7 7DN, UK;
| | - Brian R. Davidson
- Centre of Surgical Innovation, Organ Regeneration and Transplantation, University College London (UCL), London NW3 2QG, UK;
- Royal Free NHS Foundation Trust, London NW3 2QG, UK;
| |
Collapse
|
136
|
Charmsaz S, Gross N, Jaffee E, Ho WJ. A global live cell barcoding approach for multiplexed mass cytometry profiling of mouse tumors. JCI Insight 2021; 6:143283. [PMID: 33690223 PMCID: PMC8119183 DOI: 10.1172/jci.insight.143283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
With the advent of cancer immunology, mass cytometry has been increasingly employed to characterize the responses to cancer therapies and the tumor microenvironment (TME). One of its most notable applications is efficient multiplexing of samples into batches by dedicating a number of metal isotope channels to barcodes, enabling robust data acquisition and analysis. Barcoding is most effective when markers are present in all cells of interest. While CD45 has been shown to be a reliable marker for barcoding all immune cells in a given sample, a strategy to reliably barcode mouse cancer cells has not been demonstrated. To this end, we identified CD29 and CD98 as markers widely expressed by commonly used mouse cancer cell lines. We conjugated anti-CD29 and anti-CD98 antibodies to cadmium or indium metals and validated their utility in 10-plex barcoding of live cells. Finally, we established a potentially novel barcoding system incorporating the combination of CD29, CD98, and CD45 to multiplex 10 tumors from s.c. MC38 and KPC tumor models, while successfully recapitulating the known contrast in the PD1-PDL1 axis between the 2 models. The ability to barcode tumor cells along with immune cells empowers the interrogation of the tumor-immune interactions in mouse TME studies.
Collapse
|
137
|
Yap TA, Parkes EE, Peng W, Moyers JT, Curran MA, Tawbi HA. Development of Immunotherapy Combination Strategies in Cancer. Cancer Discov 2021; 11:1368-1397. [PMID: 33811048 DOI: 10.1158/2159-8290.cd-20-1209] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/03/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
Harnessing the immune system to treat cancer through inhibitors of CTLA4 and PD-L1 has revolutionized the landscape of cancer. Rational combination strategies aim to enhance the antitumor effects of immunotherapies, but require a deep understanding of the mechanistic underpinnings of the immune system and robust preclinical and clinical drug development strategies. We review the current approved immunotherapy combinations, before discussing promising combinatorial approaches in clinical trials and detailing innovative preclinical model systems being used to develop rational combinations. We also discuss the promise of high-order immunotherapy combinations, as well as novel biomarker and combinatorial trial strategies. SIGNIFICANCE: Although immune-checkpoint inhibitors are approved as dual checkpoint strategies, and in combination with cytotoxic chemotherapy and angiogenesis inhibitors for multiple cancers, patient benefit remains limited. Innovative approaches are required to guide the development of novel immunotherapy combinations, ranging from improvements in preclinical tumor model systems to biomarker-driven trial strategies.
Collapse
Affiliation(s)
- Timothy A Yap
- Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eileen E Parkes
- Oxford Institute of Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Justin T Moyers
- Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
138
|
Liu X, Jiang J, Chang CH, Liao YP, Lodico JJ, Tang I, Zheng E, Qiu W, Lin M, Wang X, Ji Y, Mei KC, Nel AE, Meng H. Development of Facile and Versatile Platinum Drug Delivering Silicasome Nanocarriers for Efficient Pancreatic Cancer Chemo-Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005993. [PMID: 33682329 PMCID: PMC8035264 DOI: 10.1002/smll.202005993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/07/2020] [Indexed: 05/09/2023]
Abstract
In this study a mesoporous silica nanoparticle (MSNP) based platform is developed for high-dose loading of a range of activated platinum (Pt) chemo agents that can be attached to the porous interior through the use of electrostatic and coordination chemistry under weak-basic pH conditions. In addition to the design feature for improving drug delivery, the MSNP can also be encapsulated in a coated lipid bilayer (silicasome), to improve the colloidal stability after intravenous (IV) injection. Improved pharmacokinetics and intratumor delivery of encapsulated activated oxaliplatin (1,2-diamminocyclohexane platinum(II) (DACHPt)) over free drug in an orthotopic Kras-derived pancreatic cancer (PDAC) model is demonstrated. Not only does IV injection of the DACHPt silicasome provide more efficacious cytotoxic tumor cell killing, but can also demonstrate that chemotherapy-induced cell death is accompanied by the features of immunogenic cell death (ICD) as well as a dramatic reduction in bone marrow toxicity. The added ICD features are reflected by calreticulin and high-mobility group box 1 expression, along with increased CD8+ /FoxP3+ T-cell ratios and evidence of perforin and granzyme B release at the tumor site. Subsequent performance of a survival experiment, demonstrates that the DACHPt silicasome generates a significant improvement in survival outcome, which can be extended by delayed administration of the anti-PD-1 antibody.
Collapse
Affiliation(s)
- Xiangsheng Liu
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Current affiliation: The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jinhong Jiang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Chong Hyun Chang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Jared J. Lodico
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Department of Physics and Astronomy, University of California, Los Angeles, California 90095, United States
| | - Ivanna Tang
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Emily Zheng
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Waveley Qiu
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Matthew Lin
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Xiang Wang
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Ying Ji
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Kuo-Ching Mei
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Andre E. Nel
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Huan Meng
- Division of Nanomedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
139
|
Ajina R, Malchiodi ZX, Fitzgerald AA, Zuo A, Wang S, Moussa M, Cooper CJ, Shen Y, Johnson QR, Parks JM, Smith JC, Catalfamo M, Fertig EJ, Jablonski SA, Weiner LM. Antitumor T-cell Immunity Contributes to Pancreatic Cancer Immune Resistance. Cancer Immunol Res 2021; 9:386-400. [PMID: 33509790 PMCID: PMC8283778 DOI: 10.1158/2326-6066.cir-20-0272] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/27/2020] [Accepted: 01/26/2021] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the United States. Pancreatic tumors are minimally infiltrated by T cells and are largely refractory to immunotherapy. Accordingly, the role of T-cell immunity in pancreatic cancer has been somewhat overlooked. Here, we hypothesized that immune resistance in pancreatic cancer was induced in response to antitumor T-cell immune responses and that understanding how pancreatic tumors respond to immune attack may facilitate the development of more effective therapeutic strategies. We now provide evidence that T-cell-dependent host immune responses induce a PDAC-derived myeloid mimicry phenomenon and stimulate immune resistance. Three KPC mouse models of pancreatic cancer were used: the mT3-2D (Kras+/LSL-G12D; Trp53+/LSL-R172H; Pdx1-Cre) subcutaneous and orthotopic models, as well as the KP1 (p48-CRE/LSL-Kras/Trp53 flox/flox ) subcutaneous model. KPC cancer cells were grown in immunocompetent and immunodeficient C57BL/6 mice and analyzed to determine the impact of adaptive immunity on malignant epithelial cells, as well as on whole tumors. We found that induced T-cell antitumor immunity, via signal transducer and activator of transcription 1 (STAT1), stimulated malignant epithelial pancreatic cells to induce the expression of genes typically expressed by myeloid cells and altered intratumoral immunosuppressive myeloid cell profiles. Targeting the Janus Kinase (JAK)/STAT signaling pathway using the FDA-approved drug ruxolitinib overcame these tumor-protective responses and improved anti-PD-1 therapeutic efficacy. These findings provide future directions for treatments that specifically disable this mechanism of resistance in PDAC.
Collapse
Affiliation(s)
- Reham Ajina
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Zoe X Malchiodi
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Allison A Fitzgerald
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Annie Zuo
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Shangzi Wang
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Maha Moussa
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, District of Columbia
| | - Connor J Cooper
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee
| | - Yue Shen
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee
| | - Quentin R Johnson
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia
| | - Jerry M Parks
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia
| | - Jeremy C Smith
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, District of Columbia
| | - Elana J Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sandra A Jablonski
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Louis M Weiner
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia.
| |
Collapse
|
140
|
Genoud V, Migliorini D. Challenging Hurdles of Current Targeting in Glioblastoma: A Focus on Immunotherapeutic Strategies. Int J Mol Sci 2021; 22:3493. [PMID: 33800593 PMCID: PMC8036548 DOI: 10.3390/ijms22073493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/23/2023] Open
Abstract
Glioblastoma is the most frequent primary neoplasm of the central nervous system and still suffers from very poor therapeutic impact. No clear improvements over current standard of care have been made in the last decade. For other cancers, but also for brain metastasis, which harbors a very distinct biology from glioblastoma, immunotherapy has already proven its efficacy. Efforts have been pursued to allow glioblastoma patients to benefit from these new approaches, but the road is still long for broad application. Here, we aim to review key glioblastoma immune related characteristics, current immunotherapeutic strategies being explored, their potential caveats, and future directions.
Collapse
Affiliation(s)
- Vassilis Genoud
- Department of Oncology, University Hospital of Geneva, 1205 Geneva, Switzerland;
- Center for Translational Research in Onco-Haematology, University of Geneva, 1205 Geneva, Switzerland
| | - Denis Migliorini
- Department of Oncology, University Hospital of Geneva, 1205 Geneva, Switzerland;
- Center for Translational Research in Onco-Haematology, University of Geneva, 1205 Geneva, Switzerland
- Brain Tumor and Immune Cell Engineering Laboratory, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, 1205 Geneva, Switzerland
| |
Collapse
|
141
|
Xiong J, Wang H, Wang Q. Suppressive Myeloid Cells Shape the Tumor Immune Microenvironment. Adv Biol (Weinh) 2021; 5:e1900311. [PMID: 33729699 DOI: 10.1002/adbi.201900311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/09/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the outcome of the conflict between the host immune system and cancer cells. The crosstalk between immune cells and tumor cells within the tumor microenvironment (TME) influences tumor progression and metastasis. Many studies have clarified the cellular and molecular events that can induce cancer cells to escape immune surveillance, including those involving tumor-induced myeloid cell-mediated immunosuppression. Emerging evidence indicates that tumor-infiltrating myeloid cells (TIMs) accelerate tumor growth and induce angiogenesis, metastasis, and therapy resistance once converted into potent immunosuppressive cells. Here, how tumor infiltrating myeloid cells participate in tumor immune evasion and the prospects of these cells in cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Jia Xiong
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, 310058, China
| | - Hui Wang
- China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, 310058, China
| |
Collapse
|
142
|
Agonistic CD40 Antibodies in Cancer Treatment. Cancers (Basel) 2021; 13:cancers13061302. [PMID: 33804039 PMCID: PMC8000216 DOI: 10.3390/cancers13061302] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/16/2021] [Accepted: 03/12/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary CD40 is a costimulatory molecule that is key for the activation of antigen-presenting cells and other innate immune cells. It plays an important role in anti-tumor immunity, and agonists of CD40 have been shown to eliminate tumors in both pre-clinical and clinical settings, alone and in combination with other treatment modalities. Here we assess the expression of CD40 and associations with other mediators of immunity in a variety of tumor types and review the potential of CD40 agonists for cancer treatment, given the promise of enhancing the interplay between innate and adaptive immunity. Abstract CD40 is expressed on a variety of antigen-presenting cells. Stimulation of CD40 results in inflammation by upregulation of other costimulatory molecules, increased antigen presentation, maturation (licensing) of dendritic cells, and activation of CD8+ T cells. Here we analyzed gene expression data from The Cancer Genome Atlas in melanoma, renal cell carcinoma, and pancreatic adenocarcinoma and found correlations between CD40 and several genes involved in antigen presentation and T cell function, supporting further exploration of CD40 agonists to treat cancer. Agonist CD40 antibodies have induced anti-tumor effects in several tumor models and the effect has been more pronounced when used in combination with other treatments (immune checkpoint inhibition, chemotherapy, and colony-stimulating factor 1 receptor inhibition). The reduction in tumor growth and ability to reprogram the tumor microenvironment in preclinical models lays the foundation for clinical development of agonistic CD40 antibodies (APX005M, ChiLob7/4, ADC-1013, SEA-CD40, selicrelumab, and CDX-1140) that are currently being evaluated in early phase clinical trials. In this article, we focus on CD40 expression and immunity in cancer, agonistic human CD40 antibodies, and their pre-clinical and clinical development. With the broad pro-inflammatory effects of CD40 and its ligand on dendritic cells and macrophages, and downstream B and T cell activation, agonists of this pathway may enhance the anti-tumor activity of other systemic therapies.
Collapse
|
143
|
Zhang Y, Chandra V, Riquelme Sanchez E, Dutta P, Quesada PR, Rakoski A, Zoltan M, Arora N, Baydogan S, Horne W, Burks J, Xu H, Hussain P, Wang H, Gupta S, Maitra A, Bailey JM, Moghaddam SJ, Banerjee S, Sahin I, Bhattacharya P, McAllister F. Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J Exp Med 2021; 217:152058. [PMID: 32860704 PMCID: PMC7953739 DOI: 10.1084/jem.20190354] [Citation(s) in RCA: 279] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/25/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a lethal malignancy with an immunosuppressive microenvironment that is resistant to most therapies. IL17 is involved in pancreatic tumorigenesis, but its role in invasive PDAC is undetermined. We hypothesized that IL17 triggers and sustains PDAC immunosuppression. We inhibited IL17/IL17RA signaling using pharmacological and genetic strategies alongside mass cytometry and multiplex immunofluorescence techniques. We uncovered that IL17 recruits neutrophils, triggers neutrophil extracellular traps (NETs), and excludes cytotoxic CD8 T cells from tumors. Additionally, IL17 blockade increases immune checkpoint blockade (PD-1, CTLA4) sensitivity. Inhibition of neutrophils or Padi4-dependent NETosis phenocopies IL17 neutralization. NMR spectroscopy revealed changes in tumor lactate as a potential early biomarker for IL17/PD-1 combination efficacy. Higher expression of IL17 and PADI4 in human PDAC corresponds with poorer prognosis, and the serum of patients with PDAC has higher potential for NETosis. Clinical studies with IL17 and checkpoint blockade represent a novel combinatorial therapy with potential efficacy for this lethal disease.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vidhi Chandra
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Erick Riquelme Sanchez
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX.,Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile
| | - Prasanta Dutta
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pompeyo R Quesada
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amanda Rakoski
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michelle Zoltan
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Seyda Baydogan
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - William Horne
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Jared Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hanwen Xu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Perwez Hussain
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, MD
| | - Huamin Wang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sonal Gupta
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anirban Maitra
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jennifer M Bailey
- Department of Gastroenterology, University of Texas Health Sciences Center, Houston, TX
| | - Seyed J Moghaddam
- Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sulagna Banerjee
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL
| | - Ismet Sahin
- Department of Engineering, Texas Southern University, Houston, TX
| | - Pratip Bhattacharya
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX.,Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
144
|
Fein MR, He XY, Almeida AS, Bružas E, Pommier A, Yan R, Eberhardt A, Fearon DT, Van Aelst L, Wilkinson JE, Dos Santos CO, Egeblad M. Cancer cell CCR2 orchestrates suppression of the adaptive immune response. J Exp Med 2021; 217:151949. [PMID: 32667673 PMCID: PMC7537399 DOI: 10.1084/jem.20181551] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/27/2019] [Accepted: 05/26/2020] [Indexed: 01/22/2023] Open
Abstract
C-C chemokine receptor type 2 (CCR2) is expressed on monocytes and facilitates their recruitment to tumors. Though breast cancer cells also express CCR2, its functions in these cells are unclear. We found that Ccr2 deletion in cancer cells led to reduced tumor growth and approximately twofold longer survival in an orthotopic, isograft breast cancer mouse model. Deletion of Ccr2 in cancer cells resulted in multiple alterations associated with better immune control: increased infiltration and activation of cytotoxic T lymphocytes (CTLs) and CD103+ cross-presenting dendritic cells (DCs), as well as up-regulation of MHC class I and down-regulation of checkpoint regulator PD-L1 on the cancer cells. Pharmacological or genetic targeting of CCR2 increased cancer cell sensitivity to CTLs and enabled the cancer cells to induce DC maturation toward the CD103+ subtype. Consistently, Ccr2−/− cancer cells did not induce immune suppression in Batf3−/− mice lacking CD103+ DCs. Our results establish that CCR2 signaling in cancer cells can orchestrate suppression of the immune response.
Collapse
Affiliation(s)
- Miriam R Fein
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Graduate Program in Genetics, Stony Brook University, Stony Brook, NY
| | - Xue-Yan He
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Ana S Almeida
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Emilis Bružas
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Watson School of Biological Sciences, Cold Spring Harbor, NY
| | | | - Ran Yan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Watson School of Biological Sciences, Cold Spring Harbor, NY
| | - Anaïs Eberhardt
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Département de Biologie, École Normale Supérieure de Lyon, Lyon, France
| | - Douglas T Fearon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK.,Weill Cornell Medical College, New York, NY
| | | | - John Erby Wilkinson
- Departments of Molecular and Integrative Physiology and Internal Medicine, University of Michigan, Ann Arbor, MI
| | | | | |
Collapse
|
145
|
Wang H, Yuan Y, Lu C, Zhou S, Zhang Y, Zhao J, Xu C, Yang J, Su H, Li B, Li X, Wang P, Xu G, Wang L, Zou X, Bao S, Zhang S, Lv Y. Analysis of T-cell receptor repertoire in peripheral blood of patients with pancreatic cancer and other pancreatic diseases. J Cell Mol Med 2021; 25:3991-4000. [PMID: 33682267 PMCID: PMC8051704 DOI: 10.1111/jcmm.16358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) has been the fourth cancer‐related death worldwide, diagnosed at an unresectable stage due to its rapid progression and few symptoms of this disease at early stages. The aim of this study was to determine the association between the diversity of T‐cell receptor (TCR) repertoire and clinicopathological characteristics of patients with PC and other benign pancreatic diseases. In order to make a comprehensive analysis the TCR repertoire, high‐throughput sequencing was used to differentiate complementarity determining region 3 (CDR3) of the TCR β chain in peripheral blood samples from 3 PC, 3 chronic pancreatitis, 3 pancreatic cystic lesions and 3 pancreatic neuroendocrine tumour patients. We found that there were significant differences related to TCR repertoire between PC and other pancreatic diseases, and PC is a relatively immunosuppressive tumour. Changes of peripheral TCR repertoire may be used to predict the progression of PC and the response to immunotherapy. And there may exist novel‐specific antigens in PC patients which could be used to design targeting immunotherapy in the nearly future.
Collapse
Affiliation(s)
- Hui Wang
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Yue Yuan
- Department of Gastroenterology, Nanjing Medical University Affiliated Drum Tower Clinical Medical College, Nanjing, China
| | - Chenglin Lu
- Department of General Surgery, The Afflicted Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Siqi Zhou
- Department of Gastroenterology, Jiangsu University Affiliated Drum Tower Hospital, Nanjing, China
| | - Yixuan Zhang
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Jing Zhao
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Chenghu Xu
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Jie Yang
- Department of Gastroenterology, Nanjing Medical University Affiliated Drum Tower Clinical Medical College, Nanjing, China
| | - Haochen Su
- Department of Gastroenterology, Nanjing Medical University Affiliated Drum Tower Clinical Medical College, Nanjing, China
| | - Borui Li
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Xihan Li
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Pin Wang
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Guifang Xu
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Shanhua Bao
- Department of General Surgery, The Afflicted Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Shu Zhang
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Ying Lv
- Department of Gastroenterology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China.,Department of Gastroenterology, Nanjing Medical University Affiliated Drum Tower Clinical Medical College, Nanjing, China
| |
Collapse
|
146
|
Manzo T, Prentice BM, Anderson KG, Raman A, Schalck A, Codreanu GS, Nava Lauson CB, Tiberti S, Raimondi A, Jones MA, Reyzer M, Bates BM, Spraggins JM, Patterson NH, McLean JA, Rai K, Tacchetti C, Tucci S, Wargo JA, Rodighiero S, Clise-Dwyer K, Sherrod SD, Kim M, Navin NE, Caprioli RM, Greenberg PD, Draetta G, Nezi L. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J Exp Med 2021; 217:151833. [PMID: 32491160 PMCID: PMC7398173 DOI: 10.1084/jem.20191920] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/14/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
CD8+ T cells are master effectors of antitumor immunity, and their presence at tumor sites correlates with favorable outcomes. However, metabolic constraints imposed by the tumor microenvironment (TME) can dampen their ability to control tumor progression. We describe lipid accumulation in the TME areas of pancreatic ductal adenocarcinoma (PDA) populated by CD8+ T cells infiltrating both murine and human tumors. In this lipid-rich but otherwise nutrient-poor TME, access to using lipid metabolism becomes particularly valuable for sustaining cell functions. Here, we found that intrapancreatic CD8+ T cells progressively accumulate specific long-chain fatty acids (LCFAs), which, rather than provide a fuel source, impair their mitochondrial function and trigger major transcriptional reprogramming of pathways involved in lipid metabolism, with the subsequent reduction of fatty acid catabolism. In particular, intrapancreatic CD8+ T cells specifically exhibit down-regulation of the very-long-chain acyl-CoA dehydrogenase (VLCAD) enzyme, which exacerbates accumulation of LCFAs and very-long-chain fatty acids (VLCFAs) that mediate lipotoxicity. Metabolic reprogramming of tumor-specific T cells through enforced expression of ACADVL enabled enhanced intratumoral T cell survival and persistence in an engineered mouse model of PDA, overcoming one of the major hurdles to immunotherapy for PDA.
Collapse
Affiliation(s)
- Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Boone M Prentice
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Kristin G Anderson
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Ayush Raman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aislyn Schalck
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Carina B Nava Lauson
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Silvia Tiberti
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Marissa A Jones
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Michelle Reyzer
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Breanna M Bates
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Jeffrey M Spraggins
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Nathan H Patterson
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carlo Tacchetti
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Sara Tucci
- Laboratory of Clinical Biochemistry and Metabolism Center for Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Simona Rodighiero
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Michael Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nicholas E Navin
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard M Caprioli
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Philip D Greenberg
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Giulio Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luigi Nezi
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
147
|
Lin JH, Huffman AP, Wattenberg MM, Walter DM, Carpenter EL, Feldser DM, Beatty GL, Furth EE, Vonderheide RH. Type 1 conventional dendritic cells are systemically dysregulated early in pancreatic carcinogenesis. J Exp Med 2021; 217:151817. [PMID: 32453421 PMCID: PMC7398166 DOI: 10.1084/jem.20190673] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 02/12/2020] [Accepted: 04/21/2020] [Indexed: 01/05/2023] Open
Abstract
Type 1 conventional dendritic cells (cDC1s) are typically thought to be dysregulated secondarily to invasive cancer. Here, we report that cDC1 dysfunction instead develops in the earliest stages of preinvasive pancreatic intraepithelial neoplasia (PanIN) in the KrasLSL-G12D/+ Trp53LSL-R172H/+ Pdx1-Cre–driven (KPC) mouse model of pancreatic cancer. cDC1 dysfunction is systemic and progressive, driven by increased apoptosis, and results in suboptimal up-regulation of T cell–polarizing cytokines during cDC1 maturation. The underlying mechanism is linked to elevated IL-6 concomitant with neoplasia. Neutralization of IL-6 in vivo ameliorates cDC1 apoptosis, rescuing cDC1 abundance in tumor-bearing mice. CD8+ T cell response to vaccination is impaired as a result of cDC1 dysregulation. Yet, combination therapy with CD40 agonist and Flt3 ligand restores cDC1 abundance to normal levels, decreases cDC1 apoptosis, and repairs cDC1 maturation to drive superior control of tumor outgrowth. Our study therefore reveals the unexpectedly early and systemic onset of cDC1 dysregulation during pancreatic carcinogenesis and suggests therapeutically tractable strategies toward cDC1 repair.
Collapse
Affiliation(s)
- Jeffrey H Lin
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Austin P Huffman
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Max M Wattenberg
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David M Walter
- Cell and Molecular Biology Graduate Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Erica L Carpenter
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David M Feldser
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gregory L Beatty
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emma E Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Robert H Vonderheide
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
148
|
Xia X, Li R, Zhou P, Xing Z, Lu C, Long Z, Wang F, Wang R. Decreased NSG3 enhances PD-L1 expression by Erk1/2 pathway to promote pancreatic cancer progress. Am J Cancer Res 2021; 11:916-929. [PMID: 33791163 PMCID: PMC7994162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/03/2020] [Indexed: 06/12/2023] Open
Abstract
Inhibiting the functioning of PD-1/PD-L1 to activate human immune system and improve the prognosis of pancreatic cancer (PC) would provide a significant boost to handling the disease. One research found the expression level of NSG3 was reduced in pediatric pilocytic astrocytoma, so is PC and we found NSG3 could regulate the expression of PD-L1. So NSG3 could become a new target for enhancing the immune response to PC. The GEPIA website was employed to analyze the prognoses in PC patients with different NSG3 levels. Immunohistochemistry (IHC) analysis was applied to detect different levels of NSG3 in para-PC and PC tissues. Cell biological function tests (in vitro) were performed and a subcutaneous nude mice tumor model (in vivo) was established to verify the effect of NSG3 on PC. Immunoblotting and RT-qPCR were utilized to demonstrate the inhibiting effect of NSG3 on PD-L1 through regulating Erk1/2 phosphorylation. A subcutaneous C57BL/6 tumor mice model was established to assess the possibility of a synergistic effect of NSG3 expression and the use of an anti-PD-L1 antibody on PC. PC tissues had decreased NSG3 expression levels, which led to poor prognosis. Overexpressing NSG3 suppressed proliferation, invasion and migration capacities of PC cells. On the contrary, knocking-down NSG3 prompted PC malignancy whether in vivo or in vitro. Importantly, NSG3 prevented Erk1/2 phosphorylation to inhibit PD-L1 expression. Additionally, NSG3 and an immune checkpoint inhibitor anti-PD-1 antibody acted synergistically, which enhanced the efficacy of the inhibitor. NSG3 inhibited PD-L1 expression by suppressing Erk1/2 phosphorylation to improve the immune response to PC. NSG3 is, therefore, a potential new diagnostic and prognostic marker, particularly useful in immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Xigang Xia
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430022, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Ran Li
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
- Guizhou Medical UniversityUniversity Town, Guian New District, Guizhou 550025, China
| | - Peng Zhou
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Zhixiang Xing
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Chao Lu
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Zhida Long
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Feiyang Wang
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Rui Wang
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| |
Collapse
|
149
|
Liang X, Peng J, Chen D, Tang L, Liu A, Fu Z, Shi L, Wang K, Shao C. Identification of novel hub genes and lncRNAs related to the prognosis and progression of pancreatic cancer by microarray and integrated bioinformatics analysis. Gland Surg 2021; 10:1104-1117. [PMID: 33842254 PMCID: PMC8033078 DOI: 10.21037/gs-21-151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/22/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is one of the most invasive and metastatic neoplasms among the fatal malignancies of the digestive system. Abnormal expression of genes and long non-coding RNAs (lncRNAs) are reportedly linked to multiple cancers. However, the lncRNA-mRNA expression profiles and their molecular mechanisms in PC progression are poorly known. This study aimed to map the hub genes and lncRNAs which might play core roles in the development of PC. METHODS This study used microarray expression analysis to screen for both differentially expressed genes (DEGs) and differentially expressed lncRNAs (DElncRNAs) between PC and matched adjacent non-tumor (AN) tissues. In order to clarify the functional classification of DEGs, we conducted GO and KEGG pathway enrichment analyses via the Enrichr database. LncRNA-mRNA co-expressed networks were also constructed to explore the probable core regulating DEGs and DElncRNAs. Subsequently, the hub genes and lncRNAs were validated via the ONCOMINE and GEPIA databases and the co-expressed networks. RESULTS By analyzing an mRNA-lncRNA microarray, we identified 943 mRNAs and 1,138 lncRNAs differentially expressed in PC tumors compared with the matched AN tissues. GO analysis confirmed that both up-regulated and down-regulated DEGs were enriched in multiple terms. The KEGG pathways enrichment analyses revealed that DEGs were mostly enriched in the focal adhesion and glutathione metabolism pathways, amongst others. Co-expressed networks were established to reveal the differential interactions between DEGs and DElncRNAs, and to indicate the core regulatory factors located at the core nodes of the co-expressed networks. The expression levels of potential core-regulating DEGs were validated by the GEPIA and ONCOMINE databases, and the relationship between overall survival and tumor stage and the potential core-regulating DEGs was analyzed using the GEPIA database. As a result, five genes and sixteen lncRNAs were finally considered as the hub transcripts in PC. CONCLUSIONS This study identified DEGs and DElncRNAs between PC tumors and matched AN tissues, and these transcripts were connected with malignant phenotypes in PC through different BPs and signaling pathways. Furthermore, five hub genes and sixteen lncRNAs were identified, which are expected to represent candidate diagnostic biomarkers or potential therapeutic targets for PC.
Collapse
Affiliation(s)
- Xing Liang
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Junfeng Peng
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Danlei Chen
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Liang Tang
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Anan Liu
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhiping Fu
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ligang Shi
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Keqi Wang
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Chenghao Shao
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
150
|
Javadrashid D, Baghbanzadeh A, Hemmat N, Hajiasgharzadeh K, Nourbakhsh NS, Lotfi Z, Baradaran B. Envisioning the immune system to determine its role in pancreatic ductal adenocarcinoma: Culprit or victim? Immunol Lett 2021; 232:48-59. [PMID: 33647329 DOI: 10.1016/j.imlet.2021.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma has a poor 5-year survival rate that makes it one of the most fatal human malignancies. Unfortunately, despite the serious improvement in the survival of most cancers, there has been a minor advance in pancreatic cancer (PC). Major advances in PC treatment have been assessed over the bygone twenty-year time span, yet some complications make the survival of the patients shorter. Getting to know the PC tumor microenvironment (TME) and the immunosuppression that happens during the pathogenesis of this malignancy could be a great help to understand the nature of the immune system and find better treatment modalities based on it. Although many immune cells are present in PC, immunosuppression of the TME leads to severe immune dysfunction in the patients, therefore immune effectors fail to do their functions. Lately, immunotherapy has been presented as one of the promising treatment strategies for different malignancies including hepatocellular carcinoma, melanoma, non-small cell lung cancer, and kidney cancer. In PC, there has been shown promising results centered around the TME, immune checkpoint inhibitors, cancer vaccines, and other approaches especially when used as combinational therapy. Here we dig a little deeper into the role of the immune system and possible therapeutic options in the treatment of PC.
Collapse
Affiliation(s)
- Darya Javadrashid
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Ziba Lotfi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|