1
|
Lee HS, Kim EN, Jeong GS. Lupenone preserves T cell activity by recovery of CD40L expression and protection from cytotoxicity due to methamphetamine exposure. PLoS One 2025; 20:e0314054. [PMID: 40111983 PMCID: PMC11925290 DOI: 10.1371/journal.pone.0314054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/04/2024] [Indexed: 03/22/2025] Open
Abstract
Methamphetamine (METH) is one of the most highly compulsive drugs in the world and has become a major public health problem over the last two decades. Exposure to METH has been investigated to cause neuronal toxicity but little is known about the effect of METH on the activity and toxicity of T lymphocytes. Lupenone has been reported to possess anti-diabetic, anti-inflammatory and anti-apoptotic effects but little is known about whether lupenone has a protective effect on T cell activation in METH-exposed cells. We evaluated the cytotoxicity and cytoprotective effects of lupenone in METH-stimulated Jurkat T cells. Results from the inhibitor assay using CD40L blocking antibodies revealed that this was due to enhanced CD40L expression on the T cells by pre-treatment with lupenone. Pre-treatment with lupenone significantly reduces METH-induced toxicity by restoring the expression of anti-apoptotic proteins in activated T cells. The protective effects of lupenone on activated T cells exposed to METH were associated with the prevention of MAPK and PI3K/Akt/mTOR pathways. These data suggest lupenone protected T cell activity by elevating CD40L expression and cell viability in cells exposed to methamphetamine. Our data showed that lupenone treatment recovered the expression of IL-2 and CD69 in METH-exposed cells.
Collapse
Affiliation(s)
- Hyun-Su Lee
- Department of Physiology, Daegu Catholic University School of Medicine, Daegu, Republic of Korea
| | - Eun-Nam Kim
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Gil-Saeng Jeong
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
2
|
Chen MY, Zhang F, Goedegebuure SP, Gillanders WE. Dendritic cell subsets and implications for cancer immunotherapy. Front Immunol 2024; 15:1393451. [PMID: 38903502 PMCID: PMC11188312 DOI: 10.3389/fimmu.2024.1393451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
Dendritic cells (DCs) play a central role in the orchestration of effective T cell responses against tumors. However, their functional behavior is context-dependent. DC type, transcriptional program, location, intratumoral factors, and inflammatory milieu all impact DCs with regard to promoting or inhibiting tumor immunity. The following review introduces important facets of DC function, and how subset and phenotype can affect the interplay of DCs with other factors in the tumor microenvironment. It will also discuss how current cancer treatment relies on DC function, and survey the myriad ways with which immune therapy can more directly harness DCs to enact antitumor cytotoxicity.
Collapse
Affiliation(s)
- Michael Y. Chen
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Felicia Zhang
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Simon Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, MO, United States
| | - William E. Gillanders
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
3
|
Naseri S, Cordova MM, Wenthe J, Lövgren T, Eriksson E, Loskog A, Ullenhag GJ. CD40 stimulation via CD40 ligand enhances adenovirus-mediated tumour immunogenicity including 'find-me', 'eat-me', and 'kill-me' signalling. J Cell Mol Med 2024; 28:e18162. [PMID: 38494863 PMCID: PMC10945091 DOI: 10.1111/jcmm.18162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/06/2023] [Accepted: 01/12/2024] [Indexed: 03/19/2024] Open
Abstract
Immunostimulatory gene therapy using oncolytic viruses is currently evaluated as a promising therapy for cancer aiming to induce anti-tumour immunity. Here, we investigate the capacity of oncolytic adenoviruses (LOAd) and their transgenes to induce immunogenicity in the infected tumour cells. Oncolysis and death-related markers were assessed after infection of eight human solid cancer cell lines with different LOAd viruses expressing a trimerized, membrane-bound (TMZ)-CD40L, TMZ-CD40L and 41BBL, or no transgenes. The viruses induced transgene expression post infection before they were killed by oncolysis. Death receptors TRAIL-R1, TRAIL-R2 and Fas as well as immunogenic cell death marker calreticulin were upregulated in cell lines post infection. Similarly, caspase 3/7 activity was increased in most cell lines. Interestingly, in CD40+ cell lines there was a significant effect of the TMZ-CD40L-encoding viruses indicating activation of the CD40-mediated apoptosis pathway. Further, these cell lines showed a significant increase of calreticulin, and TRAIL receptor 1 and 2 post infection. However, LOAd viruses induced PD-L1 upregulation which may hamper anti-tumour immune responses. In conclusion, LOAd infection increased the immunogenicity of infected tumour cells and this was potentiated by CD40 stimulation. Due to the simultaneous PD-L1 increase, LOAd viruses may benefit from combination with antibodies blocking PD1/PD-L1.
Collapse
Affiliation(s)
- Sedigheh Naseri
- Department of Immunology, Genetics and Pathology (IGP), Science for Life LaboratoriesUppsala UniversityUppsalaSweden
| | - Mariela Mejia Cordova
- Department of Immunology, Genetics and Pathology (IGP), Science for Life LaboratoriesUppsala UniversityUppsalaSweden
| | - Jessica Wenthe
- Department of Immunology, Genetics and Pathology (IGP), Science for Life LaboratoriesUppsala UniversityUppsalaSweden
| | - Tanja Lövgren
- Department of Immunology, Genetics and Pathology (IGP), Science for Life LaboratoriesUppsala UniversityUppsalaSweden
| | - Emma Eriksson
- Department of Immunology, Genetics and Pathology (IGP), Science for Life LaboratoriesUppsala UniversityUppsalaSweden
- Lokon Pharma ABUppsalaSweden
| | - Angelica Loskog
- Department of Immunology, Genetics and Pathology (IGP), Science for Life LaboratoriesUppsala UniversityUppsalaSweden
- Lokon Pharma ABUppsalaSweden
| | - Gustav J. Ullenhag
- Department of Immunology, Genetics and Pathology (IGP), Science for Life LaboratoriesUppsala UniversityUppsalaSweden
- Department of OncologyUppsala University HospitalUppsalaSweden
| |
Collapse
|
4
|
Grazia G, Bastos D, Villa L. CD40/CD40L expression and its prognostic value in cervical cancer. Braz J Med Biol Res 2023; 56:e13047. [PMID: 37970926 PMCID: PMC10644966 DOI: 10.1590/1414-431x2023e13047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023] Open
Abstract
CD40, a member of the tumor necrosis factor receptor (TNFR) family, is known to be involved in immune system regulation, acting as a costimulatory molecule, and in antitumor responses against cancer cells. It is a protein that is expressed in different types of cells, including immune cells and cancer cells (e.g., cervical cancer, breast cancer, melanoma). In this study, we investigated CD40/CD40L transcriptional and protein levels in cervical cancer cell lines and tumors. Higher CD40 expression was observed in cervical cancer cell lines derived from squamous cell carcinomas than from adenocarcinomas. Search of CD40/CD40L expression in cervical cancer tissues in public data sets revealed that about 83% of squamous cell carcinomas express CD40 compared to other cervical tumor subtypes. Moreover, expression of CD40 and CD40L in squamous cervical carcinomas is associated with better overall survival. Therefore, these proteins could be explored as prognostic markers in cervical cancers.
Collapse
Affiliation(s)
- G.A. Grazia
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brasil
| | - D.R. Bastos
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brasil
| | - L.L. Villa
- Departamento de Radiologia e Oncologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
- Centro de Investigação Translacional em Oncologia, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
5
|
Huang N, Peng L, Yang J, Li J, Zhang S, Sun M. FAM111B Acts as an Oncogene in Bladder Cancer. Cancers (Basel) 2023; 15:5122. [PMID: 37958297 PMCID: PMC10648174 DOI: 10.3390/cancers15215122] [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: 09/19/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Bladder cancer (BLCA) is a prevalent malignancy of the urinary system, associated with a high recurrence rate and poor prognosis. FAM111B, which encodes a protein containing a trypsin-like cysteine/serine peptidase domain, has been implicated in the progression of various human cancers; however, its involvement in BLCA remains unclear. In this study, we investigated the expression of FAM111B gene in tumor tissues compared to para-tumor tissues using immunohistochemistry and observed a significantly higher FAM111B gene expression in tumor tissues. Furthermore, analysis of clinical characteristics indicated that the increased FAM111B gene expression correlated with lymphatic metastasis and reduced overall survival. To investigate its functional role, we employed FAM111B-knockdown BLCA cell models and performed cell proliferation, wound-healing, transwell, and flow cytometry assays. The results showed that decreased FAM111B gene expression inhibited proliferation and migration but induced apoptosis in BLCA cells. In vivo experiments further validated that FAM111B knockdown suppressed tumor growth. Overall, our findings suggest that FAM111B acts as an oncogene in BLCA, playing a critical role in tumorigenesis, progression, and metastasis of BLCA. In conclusion, we have demonstrated a strong correlation between the expression of FAM111B gene and the development, progression, and metastasis of bladder cancer (BLCA). Thus, FAM111B is an oncogene associated with BLCA and holds promise as a molecular target for future treatment of this cancer.
Collapse
Affiliation(s)
- Ning Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai 200433, China; (N.H.); (L.P.); (J.Y.); (J.L.)
| | - Lei Peng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai 200433, China; (N.H.); (L.P.); (J.Y.); (J.L.)
| | - Jiaping Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai 200433, China; (N.H.); (L.P.); (J.Y.); (J.L.)
| | - Jinqian Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai 200433, China; (N.H.); (L.P.); (J.Y.); (J.L.)
| | - Sheng Zhang
- Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Mingjuan Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai 200433, China; (N.H.); (L.P.); (J.Y.); (J.L.)
| |
Collapse
|
6
|
Alanazi J, Bender O, Dogan R, Malik JA, Atalay A, Ali TFS, Beshr EAM, Shawky AM, Aly OM, Alqahtani YNH, Anwar S. Combination of an Oxindole Derivative with (-)-β-Elemene Alters Cell Death Pathways in FLT3/ITD + Acute Myeloid Leukemia Cells. Molecules 2023; 28:5253. [PMID: 37446914 DOI: 10.3390/molecules28135253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Acute myeloid leukemia (AML) is one of the cancers that grow most aggressively. The challenges in AML management are huge, despite many treatment options. Mutations in FLT3 tyrosine kinase receptors make the currently available therapies less responsive. Therefore, there is a need to find new lead molecules that can specifically target mutated FLT3 to block growth factor signaling and inhibit AML cell proliferation. Our previous studies on FLT3-mutated AML cells demonstrated that β-elemene and compound 5a showed strong inhibition of proliferation by blocking the mutated FLT3 receptor and altering the key apoptotic genes responsible for apoptosis. Furthermore, we hypothesized that both β-elemene and compound 5a could be therapeutically effective. Therefore, combining these drugs against mutated FLT3 cells could be promising. In this context, dose-matrix combination-based cellular inhibition analyses, cell morphology studies and profiling of 43 different apoptotic protein targets via combinatorial treatment were performed. Our studies provide strong evidence for the hypothesis that β-elemene and compound 5a combination considerably increased the therapeutic potential of both compounds by enhancing the activation of several key targets implicated in AML cell death.
Collapse
Affiliation(s)
- Jowaher Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 55476, Saudi Arabia
| | - Onur Bender
- Biotechnology Institute, Ankara University, Ankara 06135, Turkey
| | - Rumeysa Dogan
- Biotechnology Institute, Ankara University, Ankara 06135, Turkey
| | - Jonaid Ahmad Malik
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar 140001, India
| | - Arzu Atalay
- Biotechnology Institute, Ankara University, Ankara 06135, Turkey
| | - Taha F S Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Eman A M Beshr
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Ahmed M Shawky
- Science and Technology Unit (STU), Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Omar M Aly
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University, Port Said 42511, Egypt
| | | | - Sirajudheen Anwar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 55476, Saudi Arabia
| |
Collapse
|
7
|
Urbaniak A, Jablonska K, Suchanski J, Partynska A, Szymczak-Kulus K, Matkowski R, Maciejczyk A, Ugorski M, Dziegiel P. Prolactin-induced protein (PIP) increases the sensitivity of breast cancer cells to drug-induced apoptosis. Sci Rep 2023; 13:6574. [PMID: 37085653 PMCID: PMC10121699 DOI: 10.1038/s41598-023-33707-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/18/2023] [Indexed: 04/23/2023] Open
Abstract
We have previously shown that high expression of prolactin-induced protein (PIP) correlates with the response of breast cancer (BC) patients to standard adjuvant chemotherapy (doxorubicin and cyclophosphamide), which suggests that the absence of this glycoprotein is associated with resistance of tumor cells to chemotherapy. Therefore, in the present study, we analyzed the impact of PIP expression on resistance of BC cells to anti-cancer drugs and its biological role in BC progression. Expression of PIP and apoptotic genes in BC cell lines was analyzed using real-time PCR and Western blotting. PIP was detected in BC tissue specimens using immunohistochemistry. The tumorigenicity of cancer cells was analyzed by the in vivo tumor growth assay. Apoptotic cells were detected based on caspase-3 activation, Annexin V binding and TUNEL assay. The interaction of PIP with BC cells was analyzed using flow cytometry. Using two cellular models of BC (i.e. T47D cells with the knockdown of the PIP gene and MDA-MB-231 cells overexpressing PIP), we found that high expression of PIP resulted in (1) increased sensitivity of BC cells to apoptosis induced by doxorubicin (DOX), 4-hydroperoxycyclophosphamide (4-HC), and paclitaxel (PAX), and (2) improved efficacy of anti-cancer therapy with DOX in the xenograft mice model. Accordingly, a clinical study revealed that BC patients with higher PIP expression were characterized by longer 5-year overall survival and disease-free survival. Subsequent studies showed that PIP up-regulated the expression of the following pro-apoptotic genes: CRADD, DAPK1, FASLG, CD40 and BNIP2. This pro-apoptotic activity is mediated by secreted PIP and most probably involves the specific surface receptor. This study demonstrates that a high expression level of PIP sensitizes BC cells to anti-cancer drugs. Increased sensitivity to chemotherapy is the result of pro-apoptotic activity of PIP, which is evidenced by up-regulation of specific pro-apoptotic genes. As high expression of PIP significantly correlated with a better response of patients to anti-cancer drugs, this glycoprotein can be a marker for the prognostic evaluation of adjuvant chemotherapy.
Collapse
Affiliation(s)
- Anna Urbaniak
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 31, 50-375, Wroclaw, Poland
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland
- Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, 53-114, Wroclaw, Poland
| | - Karolina Jablonska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland
| | - Jaroslaw Suchanski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 31, 50-375, Wroclaw, Poland
| | - Aleksandra Partynska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland
| | - Katarzyna Szymczak-Kulus
- Laboratory of Glycobiology, Hirszfeld Institute of Immunology and Experimental Therapy, 53-114, Wroclaw, Poland
| | - Rafal Matkowski
- Department of Oncology, Wroclaw Medical University, 50-368, Wroclaw, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413, Wroclaw, Poland
| | - Adam Maciejczyk
- Department of Oncology, Wroclaw Medical University, 50-368, Wroclaw, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, 53-413, Wroclaw, Poland
| | - Maciej Ugorski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 31, 50-375, Wroclaw, Poland.
| | - Piotr Dziegiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chalubinskiego 6a, 50-368, Wroclaw, Poland.
- Department of Human Biology, Faculty of Physiotherapy, Wroclaw University of Health and Sport Sciences, 51-612, Wroclaw, Poland.
| |
Collapse
|
8
|
Bates KM, Vathiotis I, MacNeil T, Ahmed FS, Aung TN, Katlinskaya Y, Bhattacharya S, Psyrri A, Yea S, Parkes A, Sadraei NH, Roychoudhury S, Rimm DL, Gavrielatou N. Spatial characterization and quantification of CD40 expression across cancer types. BMC Cancer 2023; 23:220. [PMID: 36894898 PMCID: PMC9996913 DOI: 10.1186/s12885-023-10650-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND CD40, a TNF receptor family member, is expressed by a variety of immune cells and is involved in the activation of both adaptive and innate immune responses. Here, we used quantitative immunofluorescence (QIF) to evaluate CD40 expression on the tumor epithelium of solid tumors in large patient cohorts of lung, ovarian, and pancreatic cancers. METHODS Tissue samples from nine different solid tumors (bladder, breast, colon, gastric, head and neck, non-small cell lung cancer (NSCLC), ovarian, pancreatic and renal cell carcinoma), constructed in tissue microarray format, were initially assessed for CD40 expression by QIF. CD40 expression was then evaluated on the large available patient cohorts for three of the tumor types demonstrating high CD40 positivity rate; NSCLC, ovarian and pancreatic cancer. The prognostic impact of CD40 expression on tumor cells was also investigated. RESULTS CD40 expression on tumor cells was found to be common, with 80% of the NSCLC population, 40% of the ovarian cancer population, and 68% of the pancreatic adenocarcinoma population displaying some degree of CD40 expression on cancer cells. All of three of these cancer types displayed considerable intra-tumoral heterogeneity of CD40 expression, as well as partial correlation between expression of CD40 on tumor cells and on surrounding stromal cells. CD40 was not found to be prognostic for overall survival in NSCLC, ovarian cancer, or pancreatic adenocarcinoma. CONCLUSIONS The high percentage of tumor cells expressing CD40 in each of these solid tumors should be considered in the development of therapeutic agents designed to target CD40.
Collapse
Affiliation(s)
- Katherine M Bates
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, BML 112, New Haven, CT, 06510-8023, USA
| | - Ioannis Vathiotis
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, BML 112, New Haven, CT, 06510-8023, USA
| | - Tyler MacNeil
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, BML 112, New Haven, CT, 06510-8023, USA
| | | | - Thazin Nwe Aung
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, BML 112, New Haven, CT, 06510-8023, USA
| | | | | | - Amanda Psyrri
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, Athens, Greece
| | | | | | | | | | - David L Rimm
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, BML 112, New Haven, CT, 06510-8023, USA
| | - Niki Gavrielatou
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, BML 112, New Haven, CT, 06510-8023, USA.
| |
Collapse
|
9
|
Kim JH, Yang HJ, Choi SS, Lee HJ, Song YS. Changes of proapoptotic and antiapoptotic genes affect sensitivity to apoptotic stimuli in impaired contractility due to long term bladder outlet obstruction. PLoS One 2022; 17:e0279503. [PMID: 36574386 PMCID: PMC9794071 DOI: 10.1371/journal.pone.0279503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/08/2022] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The normal biological process that necessitates cell removal greatly depends on apoptosis. Long term bladder outlet obstruction (BOO) causes damaged smooth muscle cells to undergo apoptosis. However, smooth muscle cell apoptosis that BOO causes is not well known in impaired bladder contractility. Therefore, we designed this study to investigate whether long-term BOO could induce apoptosis activities and to obtain an expression profile of apoptosis related genes. MATERIALS AND METHODS We used 10 Sprague-Dawley six-week-old female rats. We separated them equally into two groups: a sham intervention group (group 1) and an eight-week BOO group (group 2). We conducted cystometric evaluation eight weeks following BOO onset, with processing of bladder tissue for PCR array. Every array comprised 84 genes, which were established to contribute to an apoptosis response, cell differentiation and metabolism, and 12 sequences were established for the regulation of loading and the quality of cDNA. We performed real-time PCR. Changes in gene expression presented as a fold increase/decrease. Alterations of more than two-fold constituted the cut-off determining expression. RESULTS Group 2 had a greater bladder weight and Impaired bladder contractility. Immunofluorescent staining with CAS3, TUNEL showed increased in the BOO group. In comparison to group 1, group 2 exhibited an at least two-fold upregulation in five genes, the Bcl-2 (15.1), Birc5 (5.8), Cd40lg (7.5), Il10 (16.2), and Naip2 (13.2). They also demonstrated at least a two-fold downregulation in the PRLR (-18.1) gene. Genes Bcl2ald, Circ5, Cd40lg, Il10, Naip2, and PRLR were among the genes with activity against apoptosis. TNF, STAT3 and TP53 mediated the effect that genes had on one another. CONCLUSION This study demonstrated that the relative ratios of pro- and antiapoptotic genes determine bladder cell sensitivity cells to apoptotic stimuli in impaired contractility caused by long term BOO. Although we cannot confirm whether this finding is the result of the decompensated phase of the bladder or the process, the gene expression profiles could explain molecular mechanisms of apoptosis in impaired bladder contractility caused by long-term BOO with further studies.
Collapse
Affiliation(s)
- Jae Heon Kim
- Department of Urology, Soonchunhyang University School of Medicine, Seoul, Republic of Korea
| | - Hee Jo Yang
- Department of Urology, Soonchunhyang University School of Medicine, Cheonan, Republic of Korea
| | - Sung Sik Choi
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea,Research Institute, e-biogen Inc., Seoul, Republic of Korea
| | - Hong J. Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea,Research Institute, e-biogen Inc., Seoul, Republic of Korea,* E-mail: (HJL); (YSS)
| | - Yun Seob Song
- Department of Urology, Soonchunhyang University School of Medicine, Seoul, Republic of Korea,* E-mail: (HJL); (YSS)
| |
Collapse
|
10
|
Lang I, Zaitseva O, Wajant H. FcγRs and Their Relevance for the Activity of Anti-CD40 Antibodies. Int J Mol Sci 2022; 23:12869. [PMID: 36361658 PMCID: PMC9655775 DOI: 10.3390/ijms232112869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 03/14/2024] Open
Abstract
Inhibitory targeting of the CD40L-CD40 system is a promising therapeutic option in the field of organ transplantation and is also attractive in the treatment of autoimmune diseases. After early complex results with neutralizing CD40L antibodies, it turned out that lack of Fcγ receptor (FcγR)-binding is the crucial factor for the development of safe inhibitory antibodies targeting CD40L or CD40. Indeed, in recent years, blocking CD40 antibodies not interacting with FcγRs, has proven to be well tolerated in clinical studies and has shown initial clinical efficacy. Stimulation of CD40 is also of considerable therapeutic interest, especially in cancer immunotherapy. CD40 can be robustly activated by genetically engineered variants of soluble CD40L but also by anti-CD40 antibodies. However, the development of CD40L-based agonists is biotechnologically and pharmacokinetically challenging, and anti-CD40 antibodies typically display only strong agonism in complex with FcγRs or upon secondary crosslinking. The latter, however, typically results in poorly developable mixtures of molecule species of varying stoichiometry and FcγR-binding by anti-CD40 antibodies can elicit unwanted side effects such as antibody-dependent cellular cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP) of CD40 expressing immune cells. Here, we summarize and compare strategies to overcome the unwanted target cell-destroying activity of anti-CD40-FcγR complexes, especially the use of FcγR type-specific mutants and the FcγR-independent cell surface anchoring of bispecific anti-CD40 fusion proteins. Especially, we discuss the therapeutic potential of these strategies in view of the emerging evidence for the dose-limiting activities of systemic CD40 engagement.
Collapse
Affiliation(s)
| | | | - Harald Wajant
- Department of Internal Medicine II, Division of Molecular Internal Medicine, University Hospital Würzburg, Auvera Haus, Grombühlstrasse 12, 97080 Würzburg, Germany
| |
Collapse
|
11
|
Kerneur C, Cano CE, Olive D. Major pathways involved in macrophage polarization in cancer. Front Immunol 2022; 13:1026954. [PMID: 36325334 PMCID: PMC9618889 DOI: 10.3389/fimmu.2022.1026954] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages play an important role in tissue homeostasis, tissue remodeling, immune response, and progression of cancer. Consequently, macrophages exhibit significant plasticity and change their transcriptional profile and function in response to environmental, tissue, and inflammatory stimuli resulting in pro- and anti-tumor effects. Furthermore, the categorization of tissue macrophages in inflammatory situations remains difficult; however, there is an agreement that macrophages are predominantly polarized into two different subtypes with pro- and anti-inflammatory properties, the so-called M1-like and M2-like macrophages, respectively. These two macrophage classes can be considered as the extreme borders of a continuum of many intermediate subsets. On one end, M1 are pro-inflammatory macrophages that initiate an immunological response, damage tissue integrity, and dampen tumor progression by fostering robust T and natural killer (NK) cell anti-tumoral responses. On the other end, M2 are anti-inflammatory macrophages involved in tissue remodeling and tumor growth, that promote cancer cell proliferation, invasion, tumor metastasis, angiogenesis and that participate to immune suppression. These decisive roles in tumor progression occur through the secretion of cytokines, chemokines, growth factors, and matrix metalloproteases, as well as by the expression of immune checkpoint receptors in the case of M2 macrophages. Moreover, macrophage plasticity is supported by stimuli from the Tumor Microenvironment (TME) that are relayed to the nucleus through membrane receptors and signaling pathways that result in gene expression reprogramming in macrophages, thus giving rise to different macrophage polarization outcomes. In this review, we will focus on the main signaling pathways involved in macrophage polarization that are activated upon ligand-receptor recognition and in the presence of other immunomodulatory molecules in cancer.
Collapse
Affiliation(s)
- Clément Kerneur
- ImCheck Therapeutics, Marseille, France
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| | - Carla E. Cano
- ImCheck Therapeutics, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
- *Correspondence: Clément Kerneur, ; Carla E. Cano, ; Daniel Olive,
| |
Collapse
|
12
|
Human Cytomegalovirus and Human Herpesvirus 6 Coinfection of Dermal Fibroblasts Enhances the Pro-Inflammatory Pathway Predisposing to Fibrosis: The Possible Impact on Systemic Sclerosis. Microorganisms 2022; 10:microorganisms10081600. [PMID: 36014018 PMCID: PMC9415275 DOI: 10.3390/microorganisms10081600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Systemic sclerosis (SSc) is a severe autoimmune disease likely triggered by genetic and environmental factors, including viral infections. Human cytomegalovirus (HCMV) and human herpesvirus 6A species (HHV-6A) have been associated with SSc, based on in vivo and in vitro evidence, but the data are still inconclusive. Furthermore, despite both viruses being highly prevalent in humans and able to exacerbate each other’s effects, no data are available on their joint effects. Hence, we aimed to study their simultaneous impact on the expression of cell factors correlated with fibrosis and apoptosis in in vitro coinfected fibroblasts, representing the main target cell type in SSc. The results, obtained by a microarray detecting 84 fibrosis/apoptosis-associated factors, indicated that coinfected cells underwent higher and more sustained expression of fibrosis-associated parameters compared with single-infected cells. Thus, the data, for the first time, suggest that HCMV and HHV-6A may cooperate in inducing alterations potentially leading to cell fibrosis, thus further supporting their joint role in SSc. However, further work is required to definitively answer whether β-herpesviruses are causally linked to the disease and to enable the possible use of targeted antiviral treatments to improve clinical outcomes.
Collapse
|
13
|
Novel Functions of Integrins as Receptors of CD154: Their Role in Inflammation and Apoptosis. Cells 2022; 11:cells11111747. [PMID: 35681441 PMCID: PMC9179867 DOI: 10.3390/cells11111747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022] Open
Abstract
CD154, an inflammatory mediator also known as CD40 ligand, has been identified as a novel binding partner for some members of the integrin family. The αIIbβ3, specifically expressed on platelets, was the first integrin to be described as a receptor for CD154 after CD40. Its interaction with soluble CD154 (sCD154) highly contributes to thrombus formation and stability. Identifying αIIbβ3 opened the door for investigating other integrins as partners of CD154. The αMβ2 expressed on myeloid cells was shown capable of binding CD154 and contributing as such to cell activation, adhesion, and release of proinflammatory mediators. In parallel, α5β1 communicates with sCD154, inducing pro-inflammatory responses. Additional pathogenic effects involving apoptosis-preventing functions were exhibited by the CD154–α5β1 dyad in T cells, conferring a role for such interaction in the survival of malignant cells, as well as the persistence of autoreactive T cells. More recently, CD154 receptors integrated two new integrin members, αvβ3 and α4β1, with little known as to their biological significance in this context. This article provides an overview of the novel role of integrins as receptors of CD154 and as critical players in pro-inflammatory and apoptotic responses.
Collapse
|
14
|
Chamseddine AN, Assi T, Mir O, Chouaib S. Modulating tumor-associated macrophages to enhance the efficacy of immune checkpoint inhibitors: A TAM-pting approach. Pharmacol Ther 2021; 231:107986. [PMID: 34481812 DOI: 10.1016/j.pharmthera.2021.107986] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) plasticity and diversity are both essential hallmarks of the monocyte-macrophage lineage and the tumor-derived inflammation. TAM exemplify the perfect adaptable cell with dynamic phenotypic modifications that reflect changes in their functional polarization status. Under several tumor microenvironment (TME)-related cues, TAM shift their polarization, hence promoting or halting cancer progression. Immune checkpoint inhibitors (ICI) displayed unprecedented clinical responses in various refractory cancers; but only approximately a third of patients experienced durable responses. It is, therefore, crucial to enhance the response rate of immunotherapy. Several mechanisms of resistance to ICI have been elucidated including TAM role with its essential immunosuppressive functions that reduce both anti-tumor immunity and the subsequent ICI efficacy. In the past few years, thorough research has led to a better understanding of TAM biology and innovative approaches can now be adapted through targeting macrophages' recruitment axis as well as TAM activation and polarization status within the TME. Some of these therapeutic strategies are currently being evaluated in several clinical trials in association with ICI agents. This combination between TAM modulation and ICI allows targeting TAM intrinsic immunosuppressive functions and tumor-promoting factors as well as overcoming ICI resistance. Hence, such strategies, with a better understanding of the mechanisms driving TAM modulation, may have the potential to optimize ICI efficacy.
Collapse
Affiliation(s)
- Ali N Chamseddine
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Biostatistics and Epidemiology, CESP INSERM U1018, OncoStat, Gustave Roussy, F-94805, Villejuif, France.
| | - Tarek Assi
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Pharmacology, Gustave Roussy, F-94805, Villejuif, France; Department of Ambulatory Care, Gustave Roussy, F-94805, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, F-94805, Villejuif, France
| |
Collapse
|
15
|
Yan C, Saleh N, Yang J, Nebhan CA, Vilgelm AE, Reddy EP, Roland JT, Johnson DB, Chen SC, Shattuck-Brandt RL, Ayers GD, Richmond A. Novel induction of CD40 expression by tumor cells with RAS/RAF/PI3K pathway inhibition augments response to checkpoint blockade. Mol Cancer 2021; 20:85. [PMID: 34092233 PMCID: PMC8182921 DOI: 10.1186/s12943-021-01366-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND While immune checkpoint blockade (ICB) is the current first-line treatment for metastatic melanoma, it is effective for ~ 52% of patients and has dangerous side effects. The objective here was to identify the feasibility and mechanism of RAS/RAF/PI3K pathway inhibition in melanoma to sensitize tumors to ICB therapy. METHODS Rigosertib (RGS) is a non-ATP-competitive small molecule RAS mimetic. RGS monotherapy or in combination therapy with ICB were investigated using immunocompetent mouse models of BRAFwt and BRAFmut melanoma and analyzed in reference to patient data. RESULTS RGS treatment (300 mg/kg) was well tolerated in mice and resulted in ~ 50% inhibition of tumor growth as monotherapy and ~ 70% inhibition in combination with αPD1 + αCTLA4. RGS-induced tumor growth inhibition depends on CD40 upregulation in melanoma cells followed by immunogenic cell death, leading to enriched dendritic cells and activated T cells in the tumor microenvironment. The RGS-initiated tumor suppression was partially reversed by either knockdown of CD40 expression in melanoma cells or depletion of CD8+ cytotoxic T cells. Treatment with either dabrafenib and trametinib or with RGS, increased CD40+SOX10+ melanoma cells in the tumors of melanoma patients and patient-derived xenografts. High CD40 expression level correlates with beneficial T-cell responses and better survival in a TCGA dataset from melanoma patients. Expression of CD40 by melanoma cells is associated with therapeutic response to RAF/MEK inhibition and ICB. CONCLUSIONS Our data support the therapeutic use of RGS + αPD1 + αCTLA4 in RAS/RAF/PI3K pathway-activated melanomas and point to the need for clinical trials of RGS + ICB for melanoma patients who do not respond to ICB alone. TRIAL REGISTRATION NCT01205815 (Sept 17, 2010).
Collapse
Affiliation(s)
- Chi Yan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nabil Saleh
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jinming Yang
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Caroline A Nebhan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anna E Vilgelm
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - E Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph T Roland
- Departments of Surgery and Pediatrics and the Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sheau-Chiann Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca L Shattuck-Brandt
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA. .,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
16
|
Kaur J, Singh P, Enzler T, Sahai V. Emerging antibody therapies for pancreatic adenocarcinoma: a review of recent phase 2 trials. Expert Opin Emerg Drugs 2021; 26:103-129. [PMID: 33734833 DOI: 10.1080/14728214.2021.1905795] [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] [Indexed: 02/07/2023]
Abstract
Introduction: Pancreatic adenocarcinoma is now the third-leading cause of cancer-related deaths in the US which can be attributed to rising incidence, diagnosis at advanced stages and early development of metastasis. Systemic therapy remains palliative with early development of resistance possibly related to the constitutive activation of 'undruggable' KRAS, immunosuppressive microenvironment, and intense desmoplasia. The advancements in molecular biology has led to the development and investigation of targeted and immune therapeutics.Areas covered: This study provides a comprehensive review of the literature to further the understanding of molecular targets with their respective antibody-based therapies in clinical development in pancreatic cancer. PubMed was systematically searched for English-language articles discussing antibody-based therapies under phase 2 clinical trial investigation in pancreatic adenocarcinoma.Expert opinion: PDAC remains highly resistant to chemotherapy with no significant improvement in survival for patients with advanced or metastatic cancer. Unfortunately, the majority of the antibody-based targeted and immune therapeutics have failed to meet their primary efficacy endpoints in early phase trials. However, there are a few promising antibody-based drugs with intriguing preliminary data that merit further investigation, while many more continue to be developed and investigated preclinically, and in early phase trials.
Collapse
Affiliation(s)
- Jasmeet Kaur
- Department of Internal Medicine, Saint Joseph Mercy Oakland Hospital, Pontiac, MI, USA
| | - Paramveer Singh
- Division of Hematology and Oncology, Department of Internal Medicine, Wayne State University, Detroit, MI, USA
| | - Thomas Enzler
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Vaibhav Sahai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
17
|
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
|
18
|
Wong W, Alouani E, Wei A, Ryu YK, Chabot JA, Manji GA. Future of immunotherapy in pancreas cancer and the trials, tribulations and successes thus far. Semin Oncol 2021; 48:57-68. [PMID: 33965249 DOI: 10.1053/j.seminoncol.2021.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 11/11/2022]
Abstract
Pancreas ductal adenocarcinoma (PDAC) has a dismal prognosis with a 5-year survival rate of 10%. Currently, chemotherapy remains the standard of care for systemic treatment. Immunotherapy with checkpoint inhibitors unfortunately has not been found to be effective in the treatment of PDAC to date, likely due to the highly desmoplastic and immunosuppressive tumor microenvironment (TME). Treatment targeting pathways against the immunosuppressive mechanisms of PDAC are of mounting interest to improve outcomes in PDAC. In this review, we discuss prior efforts and the current state of immunotherapy in PDAC. We will also review the emerging targets and treatments with significant clinical potential for the treatment of PDAC such as: CD40 pathway, the adenosine pathway, the CXCR4/CXCL12 axis, the CCR2/CCL2 axis, IDO pathway, and others.
Collapse
Affiliation(s)
- Winston Wong
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Emily Alouani
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Alexander Wei
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Yun Kyoung Ryu
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - John A Chabot
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY
| | - Gulam A Manji
- Division of Hematology and Oncology, Columbia University Irving Medical Center, and New York Presbyterian Hospital, New York, NY.
| |
Collapse
|
19
|
Li DK, Wang W. Characteristics and clinical trial results of agonistic anti-CD40 antibodies in the treatment of malignancies. Oncol Lett 2020; 20:176. [PMID: 32934743 PMCID: PMC7471753 DOI: 10.3892/ol.2020.12037] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/19/2020] [Indexed: 12/20/2022] Open
Abstract
Cluster of differentiation 40 (CD40) mediates many immune activities. Preclinical studies have shown that activation of CD40 can evoke massive antineoplastic effects in several tumour models in vivo, providing a rationale for using CD40 agonists in cancer immunotherapy. To date, several potential agonistic antibodies that target CD40 have been investigated in clinical trials. Early clinical trials have shown that the adverse events associated with agonists of CD40 thus far have been largely transient and clinically controllable, including storms of cytokine release, hepatotoxicity and thromboembolic events. An antitumour effect of targeting CD40 for monotherapy or combination therapy has been observed in some tumours. However, these antitumour effects have been moderate. The present review aimed to provide updated details of the clinical results of these agonists, and offer information to further investigate the strategies of combining CD40 activation with chemotherapy, radiotherapy, targeted therapy and immunomodulators. Furthermore, biomarkers should be identified for monitoring and predicting responses and informing resistance mechanisms.
Collapse
Affiliation(s)
- Da-Ke Li
- Department of Clinical Science, Shanghai R&D Center, State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd., Shanghai 201318, P.R. China
| | - Wen Wang
- Department of Clinical Science, Shanghai R&D Center, State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd., Shanghai 201318, P.R. China
| |
Collapse
|
20
|
Hong M, Clubb JD, Chen YY. Engineering CAR-T Cells for Next-Generation Cancer Therapy. Cancer Cell 2020; 38:473-488. [PMID: 32735779 DOI: 10.1016/j.ccell.2020.07.005] [Citation(s) in RCA: 412] [Impact Index Per Article: 82.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
Abstract
T cells engineered to express chimeric antigen receptors (CARs) with tumor specificity have shown remarkable success in treating patients with hematologic malignancies and revitalized the field of adoptive cell therapy. However, realizing broader therapeutic applications of CAR-T cells necessitates engineering approaches on multiple levels to enhance efficacy and safety. Particularly, solid tumors present unique challenges due to the biological complexity of the solid-tumor microenvironment (TME). In this review, we highlight recent strategies to improve CAR-T cell therapy by engineering (1) the CAR protein, (2) T cells, and (3) the interaction between T cells and other components in the TME.
Collapse
Affiliation(s)
- Mihe Hong
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Justin D Clubb
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
21
|
Wenthe J, Naseri S, Hellström AC, Wiklund HJ, Eriksson E, Loskog A. Immunostimulatory oncolytic virotherapy for multiple myeloma targeting 4-1BB and/or CD40. Cancer Gene Ther 2020; 27:948-959. [PMID: 32355275 PMCID: PMC7725669 DOI: 10.1038/s41417-020-0176-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/02/2020] [Accepted: 04/15/2020] [Indexed: 12/20/2022]
Abstract
Multiple myeloma (MM) is a plasma cell malignancy that is characterized by immune dysregulation. MM is commonly treated with immunomodulating agents, but still remains incurable. Herein, we proposed and evaluated immunostimulatory Lokon oncolytic adenoviruses (LOAd) for MM treatment. LOAd viruses are serotype 5/35 chimera, which enables infection of hematopoietic cells. Oncolysis is restricted to cells with a dysregulated retinoblastoma protein pathway, which is frequently observed in MM. Further, LOAd viruses are armed with human immunostimulatory transgenes: trimerized membrane-bound CD40L (LOAd700, LOAd703) and 4-1BBL (LOAd703). LOAd viruses were assessed in a panel of MM cell lines (ANBL-6, L363, LP-1, OPM-2, RPMI-8226, and U266-84). All cells were sensitive to infection, leading to viral replication and cell killing as analyzed by quantitative PCR and viability assay. Transgene expression was verified post infection with flow cytometry. Cell phenotypes were further altered with a downregulation of markers connected to MM progression (ICAM-1, CD70, CXCL10, CCL2, and sIL-2Rα) and an upregulation of the death receptor Fas. In a co-culture of immune and MM cells, LOAd viruses promoted activation of cytotoxic T cells as seen by higher CD69, CD107a, and IFNγ expression. This was most prominent with LOAd703. In conclusion, LOAd viruses are of interest for MM therapy.
Collapse
Affiliation(s)
- Jessica Wenthe
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| | - Sedigheh Naseri
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ann-Charlotte Hellström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Helena Jernberg Wiklund
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Emma Eriksson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Angelica Loskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Lokon Pharma AB, Uppsala, Sweden
| |
Collapse
|
22
|
Mechanistic basis of co-stimulatory CD40-CD40L ligation mediated regulation of immune responses in cancer and autoimmune disorders. Immunobiology 2019; 225:151899. [PMID: 31899051 DOI: 10.1016/j.imbio.2019.151899] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/12/2019] [Accepted: 12/14/2019] [Indexed: 02/08/2023]
Abstract
Generation of an accurate humoral and a cell mediated adaptive immune responsesare dictated by binding of an antigen to a T- and a B-cell receptor, respectively (first signal) followed by ligation of costimulatory molecules (second signal). CD40, a costimulatory receptor molecule, expressed mainly on antigen presenting cells, some non-immune cells and tumors, binds to CD40 ligand molecule expressed transiently on T-cells and non-immune cells under inflammatory conditions. In the past decade, the CD40-CD40L interaction has emerged as an immune-potentiating system that governs and regulates host immune response against various diseases and pathogens, failing of which results in detrimental patho-physiologies including cancer and autoimmune disorders. CD40-CD40L transduces immune signals intracellularly via TRAF-dependent and independent mechanisms and further downstream by different MAPK pathways and transcription factors such as NF-κB, p38 etc. While CD40 signaling pathway through its cognate interaction between B and T cells promotes activation and proliferation of B-cells, Ig class switching, and generation of B cell memory; however, CD40-CD40L interaction involving other APCs and non-immune cells relay distinct cell signaling resulting in production of a variety of cytokines/chemokines and cell adhesion molecules ultimately conferring host defense against pathogen. In cancer and autoimmune disorders, CD40-CD40L interaction is also responsible for aberrant expression of many disease specific markers, class I/II MHC molecules and other co-stimulatory molecules such as B7 and CD28 in cell- and disease-specific manner. In the present review, the current state of understanding about the CD40-CD40L mediated regulation of immune and non-immune cells is presented. The current paradigm is to target CD40 using agonist anti-CD40 mAbs alone or in synergistic combination with chemotherapy in order to harness or confer anti-tumor and anti-inflammatory immunity.
Collapse
|
23
|
Remer M, White A, Glennie M, Al-Shamkhani A, Johnson P. The Use of Anti-CD40 mAb in Cancer. Curr Top Microbiol Immunol 2019; 405:165-207. [PMID: 25651948 DOI: 10.1007/82_2014_427] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Immunomodulatory monoclonal antibody (mAb) therapy is at the forefront of developing cancer therapeutics with numerous targeted agents proving highly effective in selective patients at stimulating protective host immunity, capable of eradicating established tumours and leading to long-term disease-free states. The cell surface marker CD40 is expressed on a range of immune cells and transformed cells in malignant states whose signalling plays a critical role in modulating adaptive immune responses. Anti-CD40 mAb therapy acts via multiple mechanisms to stimulate anti-tumour immunity across a broad range of lymphoid and solid malignancies. A wealth of preclinical research in this field has led to the successful development of multiple anti-CD40 mAb agents that have shown promise in early-phase clinical trials. Significant progress has been made to enhance the engagement of antibodies with immune effectors through their interactions with Fcγ receptors (FcγRs) by the process of Fc engineering. As more is understood about how to best optimise these agents, principally through the fine-tuning of mAb structure and choice of synergistic partnerships, our ability to generate robust, clinically beneficial anti-tumour activity will form the foundation for the next generation of cancer therapeutics.
Collapse
Affiliation(s)
- Marcus Remer
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.
| | - Ann White
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Martin Glennie
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Aymen Al-Shamkhani
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Peter Johnson
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| |
Collapse
|
24
|
Pantoja-Escobar G, Morales-Martínez M, Vega GG, Castro-Escarpulli G, Vega MI. Cytotoxic effect caspase activation dependent of a genetically engineered fusion protein with a CD154 peptide mimetic (OmpC-CD154 p) on B-NHL cell lines is mediated by the inhibition of bcl-6 and YY1 through MAPK p38 activation. Leuk Lymphoma 2019; 60:1062-1070. [PMID: 30277117 DOI: 10.1080/10428194.2018.1516286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/21/2018] [Accepted: 08/08/2018] [Indexed: 10/28/2022]
Abstract
The interaction between CD40, and its ligand, CD154, is essential for the development of humoral and cellular immune responses. The selective inhibition or activation of this pathway forms the basis for the development of new therapeutics against immunologically based diseases and malignancies. We are developing a gene fusion of Salmonella typhi OmpC protein expressing the CD154 Tyr140-Ser-149 amino acid strand. This OmpC-CD154 binds CD40 and activates B cells. In this study, we demonstrate that OmpC-CD154p treatment inhibits cell growth, proliferation and induced apoptosis in the B-NHL cell lines Raji and Ramos. The Bcl-2 family proteins were regulated and the Bcl-6 and YY1 oncoproteins were inhibited. p38 MAPK activation is an important mechanism underlying the effect on proliferation and apoptosis mediated by this fusion protein. This study establishes a basis for the possible use of fusion protein OmpC-CD154 as an alternative treatment for B-NHL.
Collapse
Affiliation(s)
- Gerardo Pantoja-Escobar
- a Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas , Laboratorio de Investigación Clínica y Ambiental , Mexico City , Mexico
- b Molecular Signal Pathways in Cancer Laboratory , UIMEO, Oncology Hospital, Siglo XXI National Medical Center, IMSS , Mexico City , Mexico
| | - Mario Morales-Martínez
- b Molecular Signal Pathways in Cancer Laboratory , UIMEO, Oncology Hospital, Siglo XXI National Medical Center, IMSS , Mexico City , Mexico
| | - Gabriel G Vega
- b Molecular Signal Pathways in Cancer Laboratory , UIMEO, Oncology Hospital, Siglo XXI National Medical Center, IMSS , Mexico City , Mexico
| | - Graciela Castro-Escarpulli
- a Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas , Laboratorio de Investigación Clínica y Ambiental , Mexico City , Mexico
| | - Mario I Vega
- b Molecular Signal Pathways in Cancer Laboratory , UIMEO, Oncology Hospital, Siglo XXI National Medical Center, IMSS , Mexico City , Mexico
- c Department of Medicine , Hematology-Oncology Division VA West Los Angeles Medical Center BBRI UCLA Medical Center Jonsson Comprehensive Cancer Center , Los Angeles , CA , USA
| |
Collapse
|
25
|
Nanofluidic drug-eluting seed for sustained intratumoral immunotherapy in triple negative breast cancer. J Control Release 2018; 285:23-34. [DOI: 10.1016/j.jconrel.2018.06.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/15/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
|
26
|
The promise and challenges of immune agonist antibody development in cancer. Nat Rev Drug Discov 2018; 17:509-527. [PMID: 29904196 DOI: 10.1038/nrd.2018.75] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune cell functions are regulated by co-inhibitory and co-stimulatory receptors. The first two generations of cancer immunotherapy agents consist primarily of antagonist antibodies that block negative immune checkpoints, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte protein 4 (CTLA4). Looking ahead, there is substantial promise in targeting co-stimulatory receptors with agonist antibodies, and a growing number of these agents are making their way through various stages of development. This Review discusses the key considerations and potential pitfalls of immune agonist antibody design and development, their differentiating features from antagonist antibodies and the landscape of agonist antibodies in clinical development for cancer treatment.
Collapse
|
27
|
Jiang E, He X, Chen X, Sun G, Wu H, Wei Y, Zhao X. Expression of CD40 in Ovarian Cancer and Adenovirus-Mediated CD40 Ligand Therapy on Ovarian Cancer in Vitro. TUMORI JOURNAL 2018; 94:356-61. [DOI: 10.1177/030089160809400312] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aims and Background To test the expression level of CD40 on ovarian cancer tissues and its correlation to clinicopathological features of patients and to evaluate the therapeutic effectiveness of adenovirus-mediated CD40 ligand on ovarian cancer in vitro. Material and Methods The expression of CD40 on paraffin-embedded ovarian cancer tissues (n = 58) and normal ovarian tissues (n = 15) was tested by immunohistochemistry, and CD40 expression on ovarian cancer cells derived from fresh surgical specimens was tested by flow cytometry analysis. The apoptosis-inducing effects of adenovirus-mediated CD40 ligand therapy on ovarian cancer cells derived from fresh surgical specimens were analyzed by flow cytometry analysis and TUNEL assay. Results CD40 expression was detected in 60.3% (35/58) of paraffin-embedded ovarian cancer tissues and 73.3% (11/15) of fresh ovarian cancer tissues, but not in normal ovarian tissues (n = 15). CD40 expression was significantly correlated with FIGO stage of ovarian cancer. Adenovirus-mediated CD40 ligand therapy induced significant apoptosis effects on ovarian cancer cells derived from fresh surgical specimens in vitro compared to null adenovirus vector and phosphate-buffered saline. Conclusions Our results suggested the therapeutic potential of adenovirus-mediated CD40 ligand on ovarian cancer, especially on the late stage of ovarian cancer.
Collapse
Affiliation(s)
- Enli Jiang
- Department of Obstetrics and Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002
| | - Xiang He
- Department of Gynecology and Obstetrics, Second West China Hospital of Sichuan University, Chengdu, Sichuan, 610041
| | - Xiancheng Chen
- Department of Gynecology and Obstetrics, Second West China Hospital of Sichuan University, Chengdu, Sichuan, 610041
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Guojuan Sun
- Department of Gynecology and Obstetrics, Second West China Hospital of Sichuan University, Chengdu, Sichuan, 610041
| | - Hongbing Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Second West China Hospital of Sichuan University, Chengdu, Sichuan, 610041
| |
Collapse
|
28
|
RIP1 has a role in CD40-mediated apoptosis in human follicular lymphoma cells. Immunobiology 2017; 222:998-1003. [PMID: 28610909 DOI: 10.1016/j.imbio.2017.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/05/2017] [Indexed: 01/27/2023]
Abstract
CD40 is a cell surface receptor which belongs to tumor necrosis factor receptor (TNFR) family members. It transmits signals that regulate diverse cellular responses such as proliferation, differentiation, adhesion molecule expression and apoptosis. Unlike other TNFR family members (TRAIL-R, Fas-R and TNFR1), the CD40 cytoplasmic tail lacks death domain. However, CD40 is capable of inducing apoptosis in different types of cancer cells including lymphoma. The apoptotic effect of CD40 is linked to the involvement of Fas, TRAIL or receptor interacting protein 1 (RIP1) kinase. We have previously shown that CD40 activation has anti-apoptotic or apoptotic effect in follicular lymphoma (FL) cell lines. In this study, we investigated the mechanism by which CD40 mediates apoptosis in a follicular lymphoma cell line, HF4.9. We show here that CD40-induced apoptosis was dependent on caspase-8 activation because caspase-8 specific inhibitor, Z-IETD-FMK completely prevented apoptosis. Therefore, the involvement of TRAIL, Fas and RIP1 in caspase-8 activation was examined. The exogenous TRAIL-induced apoptosis was fully prevented by anti-TRAIL neutralizing antibody. However, the antibody had no effect on CD40-induced apoptosis indicating that CD40 did not induce the expression of endogenous TRAIL in HF4.9 cells. Moreover, the cells were not sensitive to Fas-mediated apoptosis. Interestingly, RIP1 specific inhibitor, necrostatin-1 decreased CD40-induced apoptosis, which showed that RIP1 has a role in caspase-8 activation. In conclusion, the survival or apoptotic effects of CD40-mediated signaling might be related to the differentiation stages of FL cells.
Collapse
|
29
|
Du L, Che Z, Wang-Gillam A. Promising therapeutics of gastrointestinal cancers in clinical trials. J Gastrointest Oncol 2017; 8:524-533. [PMID: 28736639 DOI: 10.21037/jgo.2017.01.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Many novel therapeutics are being developed for patients with cancers along the gastrointestinal (GI) tract. These emerging agents are frequently classified by their biological targets such as tumor growth pathways, tumor metabolism, microenvironment, etc. Some agents targeting cancer growth pathways are based on existing clinically validated therapeutic targets, such as regorafenib for hepatocellular carcinoma (HCC), while other agents focus on newly identified targets, such as FGFR fusions in cholangiocarcinoma. Drugs modifying the immunosuppressive tumor microenvironment have emerged as an attractive area of clinical investigation. Moreover, drugs targeting the stem-cell like qualities of cancer and the tight junction protein claudin 18.2 have generated quite a lot of excitement in the field. In this paper, we will systemically review the recent promising agents and therapeutic strategies in GI cancers.
Collapse
Affiliation(s)
- Lingling Du
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Andrea Wang-Gillam
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
30
|
Thind K, Padrnos LJ, Ramanathan RK, Borad MJ. Immunotherapy in pancreatic cancer treatment: a new frontier. Therap Adv Gastroenterol 2017; 10:168-194. [PMID: 28286568 PMCID: PMC5330603 DOI: 10.1177/1756283x16667909] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Pancreatic cancer is a highly aggressive and lethal cancer characterized by high invasiveness, local and extensive dissemination at time of diagnosis and resistance to treatment. Few therapies have shown efficacy in the past and even standard of care therapies yield only modest improvements in the mortality of patients with advanced or metastatic disease. Efforts have been undertaken to study the pancreatic tumor microenvironment and have established its complex and immunosuppressive nature which could explain the high resistance to chemotherapy. Novel therapies targeting the tumor microenvironment with an aim to decrease this resistance, improve immune tolerance and increase the efficacy of the current treatment have shown some promising preliminary results in preclinical and clinical trials. We review the current advances in the field of immunotherapy and their effectiveness as a potential treatment strategy in the pancreatic cancer.
Collapse
Affiliation(s)
- Komal Thind
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, OH, USA
| | - Leslie J. Padrnos
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | | | - Mitesh J. Borad
- Division of Hematology/Oncology, Mayo Clinic Arizona, 5777 E. Mayo Boulevard, Phoenix, AZ 85054, USA
| |
Collapse
|
31
|
Zafar S, Parviainen S, Siurala M, Hemminki O, Havunen R, Tähtinen S, Bramante S, Vassilev L, Wang H, Lieber A, Hemmi S, de Gruijl T, Kanerva A, Hemminki A. Intravenously usable fully serotype 3 oncolytic adenovirus coding for CD40L as an enabler of dendritic cell therapy. Oncoimmunology 2016; 6:e1265717. [PMID: 28344872 DOI: 10.1080/2162402x.2016.1265717] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 01/01/2023] Open
Abstract
Vaccination with dendritic cells (DCs), the most potent professional antigen-presenting cells in the body, is a promising approach in cancer immunotherapy. However, tumors induce immunosuppression in their microenvironment that suppresses and impairs the function of DCs. Therefore, human clinical trials with DC therapy have often been disappointing. To improve the therapeutic efficacy and to overcome the major obstacles of DC therapy, we generated a novel adenovirus, Ad3-hTERT-CMV-hCD40L, which is fully serotype 3 and expresses hCD40L for induction of antitumor immune response. The specific aim is to enhance DCs function. Data from a human cancer patient indicated that this capsid allows effective transduction of distant tumors through the intravenous route. Moreover, patient data suggested that virally produced hCD40L can activate DCs in situ. The virus was efficient in vitro and had potent antitumor activity in vivo. In a syngeneic model, tumors treated with Ad5/3-CMV-mCD40L virus plus DCs elicited greater antitumor effect as compared with either treatment alone. Moreover, virally coded CD40L induced activation of DCs, which in turn, lead to the induction of a Th1 immune response and increased tumor-specific T cells. In conclusion, Ad3-hTERT-CMV-hCD40L is promising for translation into human trials. In particular, this virus could enable successful dendritic cell therapy in cancer patients.
Collapse
Affiliation(s)
- Sadia Zafar
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki , Helsinki, Finland
| | - Suvi Parviainen
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Otto Hemminki
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki , Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki , Helsinki, Finland
| | - Siri Tähtinen
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki , Helsinki, Finland
| | - Simona Bramante
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki , Helsinki, Finland
| | - Lotta Vassilev
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki , Helsinki, Finland
| | - Hongjie Wang
- Division of Medical Genetics, University of Washington , Seattle, WA, USA
| | - Andre Lieber
- Division of Medical Genetics, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA
| | - Silvio Hemmi
- Institute of Molecular Life Sciences, University of Zurich , Zurich, Switzerland
| | | | - Anna Kanerva
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki, Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Department of Oncology, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland; Helsinki University Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
32
|
Albarbar B, Dunnill C, Georgopoulos NT. Regulation of cell fate by lymphotoxin (LT) receptor signalling: Functional differences and similarities of the LT system to other TNF superfamily (TNFSF) members. Cytokine Growth Factor Rev 2015; 26:659-71. [DOI: 10.1016/j.cytogfr.2015.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 05/10/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
|
33
|
Angelis D, Fontánez Nieves TD, Delivoria-Papadopoulos M. Temporal Changes in Caspase-1 and Caspase-8 Activities Following Brain Hypoxia With and Without Src kinase Inhibition in a Piglet Animal Model. Neurochem Res 2015; 40:2270-9. [PMID: 26342830 DOI: 10.1007/s11064-015-1717-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 08/08/2015] [Accepted: 09/01/2015] [Indexed: 12/16/2022]
Abstract
The Src family kinases are a family of intracellular, non-receptor tyrosine kinases that are involved in a variety of cellular functions including the regulation of inflammation and apoptosis after brain hypoxia. Caspase-1 (C1) activates IL-1β through the formation of complex structures, the inflammasomes, while caspase-8 (C8) is part of the extrinsic apoptotic pathway. C8 has been found to directly activate the production of IL-1β. Previously, we observed that C1 and IL-1β are increased in the acute phase after hypoxia in the brain of piglets, but they follow a different pattern long term, with C1 remaining activated throughout the period of observation, while IL-1β returning to baseline at 15 days. Src kinase inhibition ameliorated the activation of C1 and IL-1β early, but did not appear to have any effect long term. Prompted by these findings, we assessed the changes that occur over time (1 h and 15 days) in C1 and C8 activities after brain hypoxia as well as the effect of pretreatment with a Src kinase inhibitor, PP2 on these biochemical markers. Enzymatic activities were determined by spectrophotometry with measurements of C1 and C8 in each cytosolic brain sample (N = 4 in each group). We found that C1 and C8 activities increase in the acute phase following hypoxia in the brain of newborn piglets, with C8 relatively more than C1 (C8/C1 ratio increased from 2:1 as baseline to 3:1 in hypoxia). Fifteen days after hypoxia C8/C1 ratio decreased to about 1:1. In piglets that were pretreated with a Src kinase selective inhibitor (PP2) and then subjected to hypoxia, the C8/C1 ratio early increase was not observed. Immediately after hypoxia C8 and C1 follow a similar pattern of increase while long term this appears to dissociate. We propose that following this experimental methodology, the previously observed IL-1β production after hypoxia might be associated with C8 rather than C1 and that Src kinase is involved in the above process.
Collapse
Affiliation(s)
- Dimitrios Angelis
- Division of Neonatology, Department of Pediatrics, Texas Tech University Health Sciences Center, Odessa, TX, 79763, USA.
- Department of Pediatrics, Drexel University and St. Christopher's Hospital for Children, Philadelphia, PA, USA.
| | - Tania D Fontánez Nieves
- Department of Pediatrics, Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
- Department of Pediatrics, Drexel University and St. Christopher's Hospital for Children, Philadelphia, PA, USA
| | | |
Collapse
|
34
|
CD40 ligand induces RIP1-dependent, necroptosis-like cell death in low-grade serous but not serous borderline ovarian tumor cells. Cell Death Dis 2015; 6:e1864. [PMID: 26313915 PMCID: PMC4558516 DOI: 10.1038/cddis.2015.229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 07/03/2015] [Accepted: 07/15/2015] [Indexed: 01/28/2023]
Abstract
Ovarian high-grade serous carcinomas (HGSCs) and invasive low-grade serous carcinomas (LGSCs) are considered to be distinct entities. In particular, LGSCs are thought to arise from non-invasive serous borderline ovarian tumors (SBOTs) and show poor responsiveness to conventional chemotherapy. The pro-apoptotic effects of CD40 ligand (CD40L) have been demonstrated in HGSC, though the underlying mechanisms are not fully understood. Conversely, the therapeutic potential of the CD40L-CD40 system has yet to be evaluated in LGSC. We now show that CD40 protein is focally expressed on tumor cells in two of five primary LGSCs compared with no expression in eight primary SBOTs. Treatment with CD40L or agonistic CD40 antibody decreased the viability of LGSC-derived MPSC1 and VOA1312 cells, but not SBOT3.1 cells. Small interfering RNA (siRNA) targeting CD40 was used to show that it is required for these reductions in cell viability. CD40L treatment increased cleaved caspase-3 levels in MPSC1 cells though, surprisingly, neither pan-caspase inhibitor nor caspase-3 siRNA reversed or even attenuated CD40L-induced cell death. In addition, CD40-induced cell death was not affected by knockdown of the mitochondrial proteins apoptosis-inducing factor (AIF) and endonuclease G (EndoG). Interestingly, CD40L-induced cell death was blocked by necrostatin-1, an inhibitor of receptor-interacting protein 1 (RIP1), and attenuated by inhibitors of RIP3 (GSK'872) or MLKL (mixed lineage kinase domain-like; necrosulfonamide). Our results indicate that the upregulation of CD40 may be relatively common in LGSC and that CD40 activation induces RIP1-dependent, necroptosis-like cell death in LGSC cells.
Collapse
|
35
|
Coler RN, Hudson T, Hughes S, Huang PWD, Beebe EA, Orr MT. Vaccination Produces CD4 T Cells with a Novel CD154-CD40-Dependent Cytolytic Mechanism. THE JOURNAL OF IMMUNOLOGY 2015; 195:3190-7. [PMID: 26297758 DOI: 10.4049/jimmunol.1501118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/22/2015] [Indexed: 11/19/2022]
Abstract
The discovery of new vaccines against infectious diseases and cancer requires the development of novel adjuvants with well-defined activities. The TLR4 agonist adjuvant GLA-SE elicits robust Th1 responses to a variety of vaccine Ags and is in clinical development for both infectious diseases and cancer. We demonstrate that immunization with a recombinant protein Ag and GLA-SE also induces granzyme A expression in CD4 T cells and produces cytolytic cells that can be detected in vivo. Surprisingly, these in vivo CTLs were CD4 T cells, not CD8 T cells, and this cytolytic activity was not dependent on granzyme A/B or perforin. Unlike previously reported CD4 CTLs, the transcription factors Tbet and Eomes were not necessary for their development. CTL activity was also independent of the Fas ligand-Fas, TRAIL-DR5, and canonical death pathways, indicating a novel mechanism of CTL activity. Rather, the in vivo CD4 CTL activity induced by vaccination required T cell expression of CD154 (CD40L) and target cell expression of CD40. Thus, vaccination with a TLR4 agonist adjuvant induces CD4 CTLs, which kill through a previously unknown CD154-dependent mechanism.
Collapse
Affiliation(s)
- Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA 98102; Department of Global Health, University of Washington, Seattle, WA 98105; and PAI Life Sciences, Seattle, WA 98102
| | - Thomas Hudson
- Infectious Disease Research Institute, Seattle, WA 98102
| | - Sean Hughes
- Infectious Disease Research Institute, Seattle, WA 98102
| | - Po-Wei D Huang
- Infectious Disease Research Institute, Seattle, WA 98102
| | - Elyse A Beebe
- Infectious Disease Research Institute, Seattle, WA 98102
| | - Mark T Orr
- Infectious Disease Research Institute, Seattle, WA 98102; Department of Global Health, University of Washington, Seattle, WA 98105; and
| |
Collapse
|
36
|
Kim H, Kim Y, Bae S, Kong JM, Choi J, Jang M, Choi J, Hong JM, Hwang YI, Kang JS, Lee WJ. Direct Interaction of CD40 on Tumor Cells with CD40L on T Cells Increases the Proliferation of Tumor Cells by Enhancing TGF-β Production and Th17 Differentiation. PLoS One 2015; 10:e0125742. [PMID: 25992978 PMCID: PMC4436336 DOI: 10.1371/journal.pone.0125742] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/26/2015] [Indexed: 11/18/2022] Open
Abstract
It has recently been reported that the CD40-CD40 ligand (CD40L) interaction is important in Th17 development. In addition, transforming growth factor-beta (TGF-β) promotes tumorigenesis as an immunosuppressive cytokine and is crucial in the development of Th17 cells. This study investigated the role of CD40 in breast cancer cells and its role in immunosuppressive function and tumor progression. CD40 was highly expressed in the breast cancer cell line MDA-MB231, and its stimulation with CD40 antibodies caused the up-regulation of TGF-β. Direct CD40-CD40L interaction between MDA-MB231 cells and activated T cells also increased TGF-β production and induced the production of IL-17, which accelerated the proliferation of MDA-MB231 cells through the activation of STAT3. Taken together, the direct CD40-CD40L interaction of breast tumor cells and activated T cells increases TGF-β production and the differentiation of Th17 cells, which promotes the proliferation of breast cancer cells.
Collapse
Affiliation(s)
- Hyemin Kim
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, 110–799, Korea
| | - Yejin Kim
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Seyeon Bae
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Joo Myoung Kong
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Jiwon Choi
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Mirim Jang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Jiyea Choi
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Jun-man Hong
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Young-il Hwang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| | - Jae Seung Kang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, 110–799, Korea
- * E-mail:
| | - Wang Jae Lee
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy, Seoul National University College of Medicine, Seoul, 110–799, Korea
| |
Collapse
|
37
|
Hassan GS, Stagg J, Mourad W. Role of CD154 in cancer pathogenesis and immunotherapy. Cancer Treat Rev 2015; 41:431-40. [PMID: 25843228 DOI: 10.1016/j.ctrv.2015.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/11/2023]
Abstract
Many factors and molecules have been investigated as potential players in the pathogenesis or immunosurveillance of cancer. Among these, CD154 has been recognized as a co-stimulatory molecule with high potential for treating cancer, in addition to its contribution in the development of the disease. CD154 was initially described for its pivotal role in T cell-dependent humoral responses via an interaction with its classical receptor, CD40. Subsequent studies showed that CD154 is also implicated in cell-mediated immunity and inflammation via an interaction with CD40 alone or in combination with newly identified receptors, members of the integrin family, leading to the development of chronic inflammatory and autoimmune diseases. In the current article, we present an overview of the role of CD154 as a potential etiological factor in tumors inducing proliferation of malignant cells, their rescue from apoptosis and their invasiveness. In addition, this review describes the immuno-regulatory functions of CD154 against cancer reflected by its stimulation of antigen-presenting cells and the subsequent activation of effector cells, its enhancement of malignant cells' immunogenicity, its modulation of immune settings around tumors, and its initiation of proliferation inhibiting effects in malignant cells. In vitro as well as in vivo studies are outlined and a particular attention is given to clinical studies and progress reached at this point. Findings reviewed herein will improve our knowledge of the role of the CD154 system in cancers from causative to immunotherapeutic functions, paving the way for the identification of new targets for prevention and/or treatment of malignant disorders.
Collapse
Affiliation(s)
- Ghada S Hassan
- Centre de Recherche-Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, Quebec, Canada
| | - John Stagg
- Centre de Recherche-Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, Quebec, Canada
| | - Walid Mourad
- Centre de Recherche-Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, Quebec, Canada.
| |
Collapse
|
38
|
O'Brien MA, Power DG, Clover AJP, Bird B, Soden DM, Forde PF. Local tumour ablative therapies: Opportunities for maximising immune engagement and activation. Biochim Biophys Acta Rev Cancer 2014; 1846:510-23. [DOI: 10.1016/j.bbcan.2014.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/05/2014] [Accepted: 09/20/2014] [Indexed: 12/12/2022]
|
39
|
Tin AS, Park AH, Sundar SN, Firestone GL. Essential role of the cancer stem/progenitor cell marker nucleostemin for indole-3-carbinol anti-proliferative responsiveness in human breast cancer cells. BMC Biol 2014; 12:72. [PMID: 25209720 PMCID: PMC4180847 DOI: 10.1186/s12915-014-0072-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Nucleostemin is a nucleolus residing GTPase that is considered to be an important cancer stem/progenitor cell marker protein due to its high expression levels in breast cancer stem cells and its role in tumor-initiation of human mammary tumor cells. It has been proposed that nucleostemin may represent a valuable therapeutic target for breast cancer; however, to date evidence supporting the cellular mechanism has not been elucidated. RESULTS Expression of exogenous HER2, a member of the EGF receptor gene family, in the human MCF-10AT preneoplastic mammary epithelial cell line formed a new breast cancer cell line, 10AT-Her2, which is highly enriched in cells with stem/progenitor cell-like character. 10AT-Her2 cells display a CD44+/CD24-/low phenotype with high levels of the cancer stem/progenitor cell marker proteins nucleostemin, and active aldehyde dehydrogenase-1. The overall expression pattern of HER2 protein and the stem/progenitor cell marker proteins in the 10AT-Her2 cell population is similar to that of the luminal HER2+ SKBR3 human breast cancer cell line, whereas, both MCF-7 and MDA-MB-231 cells display reduced levels of nucleostemin and no detectable expression of ALDH-1. Importantly, in contrast to the other well-established human breast cancer cell lines, 10AT-Her2 cells efficiently form tumorspheres in suspension cultures and initiate tumor xenograft formation in athymic mice at low cell numbers. Furthermore, 10AT-Her2 cells are highly sensitive to the anti-proliferative apoptotic effects of indole-3-carbinol (I3C), a natural anti-cancer indolecarbinol from cruciferous vegetables of the Brassica genus such as broccoli and cabbage. I3C promotes the interaction of nucleostemin with MDM2 (Murine Double Mutant 2), an inhibitor of the p53 tumor suppressor, and disrupts the MDM2 interaction with p53. I3C also induced nucleostemin to sequester MDM2 in a nucleolus compartment, thereby freeing p53 to mediate its apoptotic activity. siRNA knockdown of nucleostemin functionally documented that nucleostemin is required for I3C to trigger its cellular anti-proliferative responses, inhibit tumorsphere formation, and disrupt MDM2-p53 protein-protein interactions. Furthermore, expression of an I3C-resistant form of elastase, the only known target protein for I3C, prevented I3C anti-proliferative responses in cells and in tumor xenografts in vivo, as well as disrupt the I3C stimulated nucleostemin-MDM2 interactions. CONCLUSIONS Our results provide the first evidence that a natural anti-cancer compound mediates its cellular and in vivo tumor anti-proliferative responses by selectively stimulating cellular interactions of the stem/progenitor cell marker nucleostemin with MDM2, which frees p53 to trigger its apoptotic response. Furthermore, our study provides a new mechanistic template that can be potentially exploited for the development of cancer stem/progenitor cell targeted therapeutic strategies.
Collapse
|
40
|
Song L, Ma A, Dun H, Hu Y, Zeng L, Bai J, Zhang G, Kinugasa F, Sudo Y, Miyao Y, Okimura K, Miura T, Daloze P, Chen H. Effects of ASKP1240 combined with tacrolimus or mycophenolate mofetil on renal allograft survival in Cynomolgus monkeys. Transplantation 2014; 98:267-76. [PMID: 24992357 PMCID: PMC4175122 DOI: 10.1097/tp.0000000000000236] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 04/04/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND Blocking the CD40-CD154 signal pathway has previously shown promise as a strategy to prevent allograft rejection. In this study, the efficacy of a novel fully human anti-CD40 monoclonal antibody-ASKP1240, administered as a monotherapy or combination therapy (subtherapeutic dose of tacrolimus or mycophenolate mofetil), on the prevention of renal allograft rejection was evaluated in Cynomolgus monkeys. METHODS Heterotopic kidney transplants were performed in ABO-compatible, stimulation index 2.5 or higher in the two-way mixed lymphocyte reaction monkey pairs. Animals were divided into 12 groups and observed for a maximum of 180 days. Histopathologic, hematology, and biochemistry analyses were conducted in all groups. Cytokine release (interleukin [IL]-2, IL-4, IL-5, IL-6, tumor necrosis factor, and interferon-γ) was investigated in several groups. RESULTS ASKP1240 prolonged renal allograft survival in a dose-dependent manner in monotherapy. Low-dose (2 mg/kg) or high-dose (5 mg/kg) ASKP1240, in combination with mycophenolate mofetil (15 mg/kg) or tacrolimus (1 mg/kg), showed a significantly longer allograft survival time compared with monotherapy groups. No obvious side effects including drug-related thromboembolic complications were found. Cytokine release was not induced by ASKP1240 administration. CONCLUSION The present study indicates that ASKP1240, alone or in combination with other immunosuppressive drugs, could be a promising antirejection agent in organ transplantation.
Collapse
Affiliation(s)
- Lijun Song
- 1 Department of Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montreal, Montreal, Quebec, Canada. 2 Laboratory Animals Center, the Academy of Military Medical Sciences, Beijing, China. 3 Translational and Development Pharmacology-US, Drug Discovery Research, Astellas Research Institute of America LLC, Northbrook, IL. 4 Astellas Research Technology Inc., Osaka, Japan. 5 Drug Metabolism Research Labs. Astellas Pharma Inc., Osaka, Japan. 6 Pharmacological Research Labs., Kyowa Hakko Kirin Co., Ltd., Shizuoka, Japan. 7 Address correspondence to: Huifang Chen, M.D., Ph.D., Laboratory of Experimental Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montréal, 2099 Alexandre de Sève, Montréal, Room Y1611, Québec, Canada H2L 2W5
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Wang W, Meng M, Zhang Y, Wei C, Xie Y, Jiang L, Wang C, Yang F, Tang W, Jin X, Chen D, Zong J, Hou Z, Li R. Global transcriptome-wide analysis of CIK cells identify distinct roles of IL-2 and IL-15 in acquisition of cytotoxic capacity against tumor. BMC Med Genomics 2014; 7:49. [PMID: 25108500 PMCID: PMC4134122 DOI: 10.1186/1755-8794-7-49] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 08/05/2014] [Indexed: 12/13/2022] Open
Abstract
Background Cytokine-induced killer (CIK) cells are an emerging approach of cancer treatment. Our previous study have shown that CIK cells stimulated with combination of IL-2 and IL-15 displayed improved proliferation capacity and tumor cytotoxicity. However, the mechanisms of CIK cell proliferation and acquisition of cytolytic function against tumor induced by IL-2 and IL-15 have not been well elucidated yet. Methods CIKIL-2 and CIKIL-15 were generated from peripheral blood mononuclear cells primed with IFN-γ, and stimulated with IL-2 and IL-15 in combination with OKT3 respectively. RNA-seq was performed to identify differentially expressed genes, and gene ontology and pathways based analysis were used to identify the distinct roles of IL-2 and IL-15 in CIK preparation. Results The results indicated that CIKIL-15 showed improved cell proliferation capacity compared to CIKIL-2. However, CIKIL-2 has exhibited greater tumor cytotoxic effect than CIKIL-15. Employing deep sequencing, we sequenced mRNA transcripts in CIKIL-2 and CIKIL-15. A total of 374 differentially expressed genes (DEGs) were identified including 175 up-regulated genes in CIKIL-15 and 199 up-regulated genes in CIKIL-2. Among DEGs in CIKIL-15, Wnt signaling and cell adhesion were significant GO terms and pathways which related with their functions. In CIKIL-2, type I interferon signaling and cytokine-cytokine receptor interaction were significant GO terms and pathways. We found that the up-regulation of Wnt 4 and PDGFD may contribute to enhanced cell proliferation capacity of CIKIL-15, while inhibitory signal from interaction between CTLA4 and CD80 may be responsible for the weak proliferation capacity of CIKIL-2. Moreover, up-regulated expressions of CD40LG and IRF7 may make for improved tumor cytolytic function of CIKIL-2 through type I interferon signaling. Conclusions Through our findings, we have preliminarily elucidated the cells proliferation and acquisition of tumor cytotoxicity mechanism of CIKIL-15 and CIKIL-2. Better understanding of these mechanisms will help to generate novel CIK cells with greater proliferation potential and improved tumor cytolytic function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Zongliu Hou
- Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, People's Republic of China.
| | | |
Collapse
|
42
|
Weiss JM, Wiltout RH. Multifaceted antitumor responses to activating anti-CD40 antibody therapy combined with immunomodulatory or targeted agents. Oncoimmunology 2014; 3:e954483. [PMID: 25610748 DOI: 10.4161/21624011.2014.954483] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/07/2014] [Indexed: 12/15/2022] Open
Abstract
Therapeutic targeting of the CD40 pathway may be efficacious for cancer treatment. Accumulating evidence suggests synergistic and unique antitumor responses may be achieved using CD40-based therapies in combination with other immunomodulators or targeted agents.
Collapse
Affiliation(s)
- Jonathan M Weiss
- Cancer and Inflammation Program; National Cancer Institute ; Frederick, MD USA
| | - Robert H Wiltout
- Cancer and Inflammation Program; National Cancer Institute ; Frederick, MD USA
| |
Collapse
|
43
|
Al-Zoobi L, Salti S, Colavecchio A, Jundi M, Nadiri A, Hassan GS, El-Gabalawy H, Mourad W. Enhancement of Rituximab-induced cell death by the physical association of CD20 with CD40 molecules on the cell surface. Int Immunol 2014; 26:451-65. [PMID: 24894009 DOI: 10.1093/intimm/dxu046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CD20 is an attractive therapeutic target given the success of its monoclonal antibody, Rituximab, in the treatment of B-cell malignancies and B-cell-mediated autoimmune diseases. Treatment with Rituximab causes a rapid depletion of B cells and a decrease in disease symptoms. Despite the clinical efficiency of Rituximab, its mechanism of action is not completely understood. In this study, we aimed at further investigating the Rituximab-induced cell death and the factors affecting such responses. Our results indicate that Rituximab-induced cell death depends on the nature of the cells and levels of CD20 expression on the cell surface. Coexpression of CD20 with CD40, a member of the TNF receptor family that is known to be physically associated with CD20 on the cell surface, enhances the apoptotic response induced by Rituximab. Inhibiting the formation of CD40 disulfide-bound-homodimers, a process required for some CD40 signaling, further enhances Rituximab-induced cell death. Cell death induced by anti-CD40 mAb is also upregulated by the presence of CD20, suggesting a bidirectional influence of the CD20/CD40 association. Moreover, treating cells with both anti-CD20 and anti-CD40 antibodies improves the cell death response induced by a single-agent treatment. These results highlight the role of the CD20/CD40 association in triggering B-cell depletion and may pave the way for an alternative more efficient therapeutic strategy in treating B-cell-mediated disorders.
Collapse
Affiliation(s)
- Loubna Al-Zoobi
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Suzanne Salti
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Anna Colavecchio
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Malek Jundi
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Amal Nadiri
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Ghada S Hassan
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Hani El-Gabalawy
- Arthritis Centre, University of Manitoba Arthritis Centre, RR149-800 Sherbrooke Street, Winnipeg, Manitoba R3A 1M4, Canada
| | - Walid Mourad
- Laboratoire d'immunologie cellulaire et moléculaire, Centre de Recherche-Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| |
Collapse
|
44
|
CD40 expression in renal cell carcinoma is associated with tumor apoptosis, CD8(+) T cell frequency and patient survival. Hum Immunol 2014; 75:614-20. [PMID: 24801648 DOI: 10.1016/j.humimm.2014.04.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 03/17/2014] [Accepted: 04/27/2014] [Indexed: 12/11/2022]
Abstract
The co-stimulatory molecule, CD40, is expressed in renal cell carcinoma (RCC) and a variety of inflammatory diseases in the kidney. We investigated the relationship between tumor-associated CD40 expression, immune milieu of the tumor microenvironment, tumor stage and survival of patients with RCC. The expression of CD40, TUNEL and CD8 in human renal cell carcinomas was analyzed by immunohistochemistry performed on tissue samples obtained at the time of surgery. Computer-assisted quantitation of protein expression was used to analyze results in connection with patient survival and tumor stage. We show for the first time that tumor-associated CD40 expression is associated with prolonged survival in RCC patients. Tumor apoptosis (TUNEL) and CD8 immunostaining were also associated with patient survival. No relation was observed between CD40 expression and tumor stage. Our results suggest CD40 may be a prognostic biomarker indicative of prolonged RCC patient survival. Strategies that up-regulate CD40 expression in some RCC patients may thus improve survival, supporting further studies of agonistic CD40 antibodies in RCC.
Collapse
|
45
|
Klinker MW, Lizzio V, Reed TJ, Fox DA, Lundy SK. Human B Cell-Derived Lymphoblastoid Cell Lines Constitutively Produce Fas Ligand and Secrete MHCII(+)FasL(+) Killer Exosomes. Front Immunol 2014; 5:144. [PMID: 24765093 PMCID: PMC3980107 DOI: 10.3389/fimmu.2014.00144] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 03/20/2014] [Indexed: 12/18/2022] Open
Abstract
Immune suppression mediated by exosomes is an emerging concept with potentially immense utility for immunotherapy in a variety of inflammatory contexts, including allogeneic transplantation. Exosomes containing the apoptosis-inducing molecule Fas ligand (FasL) have demonstrated efficacy in inhibiting antigen-specific immune responses upon adoptive transfer in animal models. We report here that a very high frequency of human B cell-derived lymphoblastoid cell lines (LCL) constitutively produce MHCII+FasL+ exosomes that can induce apoptosis in CD4+ T cells. All LCL tested for this study (>20 independent cell lines) showed robust expression of FasL, but had no detectable FasL on the cell surface. Given this intracellular sequestration, we hypothesized that FasL in LCL was retained in the secretory lysosome and secreted via exosomes. Indeed, we found both MHCII and FasL proteins present in LCL-derived exosomes, and using a bead-based exosome capture assay demonstrated the presence of MHCII+FasL+ exosomes among those secreted by LCL. Using two independent experimental approaches, we demonstrated that LCL-derived exosomes were capable of inducing antigen-specific apoptosis in autologous CD4+ T cells. These results suggest that LCL-derived exosomes may present a realistic source of immunosuppressive exosomes that could reduce or eliminate T cell-mediated responses against donor-derived antigens in transplant recipients.
Collapse
Affiliation(s)
- Matthew W Klinker
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Vincent Lizzio
- Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Tamra J Reed
- Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - David A Fox
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Steven K Lundy
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| |
Collapse
|
46
|
Hassan SB, Sørensen JF, Olsen BN, Pedersen AE. Anti-CD40-mediated cancer immunotherapy: an update of recent and ongoing clinical trials. Immunopharmacol Immunotoxicol 2014; 36:96-104. [PMID: 24555495 DOI: 10.3109/08923973.2014.890626] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The costimulatory molecule CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily and is expressed on various antigen presenting cells (APCs) as well as some tumor cells. The binding to the natural ligand CD40L, which is expressed on T helper cells, leads to APC activation and thus enhancement of immune responses. Treatment with anti-CD40 monoclonal antibodies has been exploited in several cancer immunotherapy studies in mice and led to the development of anti-CD40 antibodies for clinical use. Here, Dacetuzumab and Lucatumumab are in the most advanced stage and are being tested as treatment for malignancies such as chronic lymphatic leukemia (CLL), Multiple Myeloma (MM), and non-Hodgkin's lymphoma (NHL). The promising results from these early clinical trials have encouraged clinical drug development in order to investigate the effect of CD40 mAbs in combination with other cancer immunotherapies, in particular interleukin (IL)-2. An in-depth analysis of this immunotherapy is provided elsewhere. In the present review, we provide an update of the most recent clinical trials with anti-CD40 antibodies. We present and discuss recent and ongoing clinical trials in this field, including clinical studies which combine anti-CD40 treatment with other cancer-treatments, such as Rituximab and Tremelimumab.
Collapse
Affiliation(s)
- Sufia Butt Hassan
- Department of International Health, Immunology and Microbiology, University of Copenhagen , Copenhagen N , Denmark
| | | | | | | |
Collapse
|
47
|
Parviainen S, Ahonen M, Diaconu I, Hirvinen M, Karttunen Å, Vähä-Koskela M, Hemminki A, Cerullo V. CD40 ligand and tdTomato-armed vaccinia virus for induction of antitumor immune response and tumor imaging. Gene Ther 2013; 21:195-204. [PMID: 24305418 DOI: 10.1038/gt.2013.73] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 10/02/2013] [Accepted: 10/11/2013] [Indexed: 12/31/2022]
Abstract
Oncolytic vaccinia virus is an attractive platform for immunotherapy. Oncolysis releases tumor antigens and provides co-stimulatory danger signals. However, arming the virus can improve efficacy further. CD40 ligand (CD40L, CD154) can induce apoptosis of tumor cells and it also triggers several immune mechanisms. One of these is a T-helper type 1 (Th1) response that leads to activation of cytotoxic T-cells and reduction of immune suppression. Therefore, we constructed an oncolytic vaccinia virus expressing hCD40L (vvdd-hCD40L-tdTomato), which in addition features a cDNA expressing the tdTomato fluorochrome for detection of virus, potentially important for biosafety evaluation. We show effective expression of functional CD40L both in vitro and in vivo. In a xenograft model of bladder carcinoma sensitive to CD40L treatment, we show that growth of tumors was significantly inhibited by the oncolysis and apoptosis following both intravenous and intratumoral administration. In a CD40-negative model, CD40L expression did not add potency to vaccinia oncolysis. Tumors treated with vvdd-mCD40L-tdtomato showed enhanced efficacy in a syngenic mouse model and induced recruitment of antigen-presenting cells and lymphocytes at the tumor site. In summary, oncolytic vaccinia virus coding for CD40L mediates multiple antitumor effects including oncolysis, apoptosis and induction of Th1 type T-cell responses.
Collapse
Affiliation(s)
- S Parviainen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - M Ahonen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - I Diaconu
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - M Hirvinen
- Laboratory of Immunovirotherapy, Faculty of Pharmacy, Division of Biopharmaceutics and Pharmacokinetics, University of Helsinki, Helsinki Finland
| | - Å Karttunen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - M Vähä-Koskela
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - A Hemminki
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - V Cerullo
- Laboratory of Immunovirotherapy, Faculty of Pharmacy, Division of Biopharmaceutics and Pharmacokinetics, University of Helsinki, Helsinki Finland
| |
Collapse
|
48
|
Sandin LC, Orlova A, Gustafsson E, Ellmark P, Tolmachev V, Tötterman TH, Mangsbo SM. Locally delivered CD40 agonist antibody accumulates in secondary lymphoid organs and eradicates experimental disseminated bladder cancer. Cancer Immunol Res 2013; 2:80-90. [PMID: 24778163 DOI: 10.1158/2326-6066.cir-13-0067] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunotherapy with intratumoral injection of adenoviral vectors expressing CD40L has yielded positive results in experimental and clinical bladder cancer. We therefore hypothesized that anti-CD40 antibody would be effective in this setting. Agonistic CD40 antibodies were developed as vaccine adjuvants but have later been used as treatment of advanced solid tumors and hematologic cancers. Systemic anti-CD40 therapy has been associated with immune-related adverse events, such as cytokine release syndrome and liver toxicity, and local delivery is an attractive approach that could reduce toxicity. Herein, we compared local and systemic anti-CD40 antibody delivery to evaluate efficacy, toxicity, and biodistribution in the experimental MB49 bladder cancer model. Antitumor effects were confirmed in the B16 model. In terms of antitumor efficacy, local anti-CD40 antibody stimulation was superior to systemic therapy at an equivalent dose and CD8 T cells were crucial for tumor growth inhibition. Both administration routes were dependent on host CD40 expression for therapeutic efficacy. In vivo biodistribution studies revealed CD40-specific antibody accumulation in the tumor-draining lymph nodes and the spleen, most likely reflecting organs with frequent target antigen-expressing immune cells. Systemic administration led to higher antibody concentrations in the liver and blood compared with local delivery, and was associated with elevated levels of serum haptoglobin. Despite the lack of a slow-release system, local anti-CD40 therapy was dependent on tumor antigen at the injection site for clearance of distant tumors. To summarize, local low-dose administration of anti-CD40 antibody mediates antitumor effects in murine models with reduced toxicity and may represent an attractive treatment alternative in the clinic.
Collapse
Affiliation(s)
- Linda C Sandin
- Authors' Affiliations: Department of Immunotechnology, Lund University, Lund, Sweden
| | | | | | | | | | | | | |
Collapse
|
49
|
Aronin A, Amsili S, Prigozhina TB, Tzdaka K, Rachmilewitz J, Shani N, Tykocinski ML, Dranitzki Elhalel M. Fn14•TRAIL effectively inhibits hepatocellular carcinoma growth. PLoS One 2013; 8:e77050. [PMID: 24130833 PMCID: PMC3794952 DOI: 10.1371/journal.pone.0077050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/30/2013] [Indexed: 12/31/2022] Open
Abstract
Background New strategies for the treatment of hepatocellular carcinoma (HCC) are needed, given that currently available chemotherapeutics are inefficient. Since tumor growth reflects the net balance between pro-proliferative and death signaling, agents shifting the equilibrium toward the latter are of considerable interest. The TWEAK:Fn14 signaling axis promotes tumor cell proliferation and tumor angiogenesis, while TRAIL:TRAIL-receptor (TRAIL-R) interactions selectively induce apoptosis in malignant cells. Fn14•TRAIL, a fusion protein bridging these two pathways, has the potential to inhibit tumor growth, by interfering with TWEAK:Fn14 signaling, while at the same time enforcing TRAIL:TRAIL-R-mediated apoptosis. Consequently, Fn14•TRAIL's capacity to inhibit HCC growth was tested. Results Fn14•TRAIL induced robust apoptosis of multiple HCC cell lines, while sparing non-malignant hepatocyte cell lines. Differential susceptibility to this agent did not correlate with expression levels of TRAIL, TRAIL-R, TWEAK and Fn14 by these lines. Fn14•TRAIL was more potent than soluble TRAIL, soluble Fn14, or a combination of the two. The requirement of both of Fn14•TRAIL's molecular domains for function was established using blocking antibodies directed against each of them. Subcutaneous injection of Fn14•TRAIL abrogated HCC growth in a xenograft model, and was well tolerated by the mice. Conclusions In this study, Fn14•TRAIL, a multifunctional fusion protein originally designed to treat autoimmunity, was shown to inhibit the growth of HCC, both invitro and invivo. The demonstration of this fusion protein’s potent anti-tumor activity suggests that simultaneous targeting of two signaling axes by a single fusion can serve as a basis for highly effective anti-cancer therapies.
Collapse
Affiliation(s)
- Alexandra Aronin
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Tatyana B. Prigozhina
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Kobi Tzdaka
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Jacob Rachmilewitz
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Mark L. Tykocinski
- Office of the Dean, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Michal Dranitzki Elhalel
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
50
|
Klinker MW, Reed TJ, Fox DA, Lundy SK. Interleukin-5 supports the expansion of fas ligand-expressing killer B cells that induce antigen-specific apoptosis of CD4(+) T cells and secrete interleukin-10. PLoS One 2013; 8:e70131. [PMID: 23940537 PMCID: PMC3734024 DOI: 10.1371/journal.pone.0070131] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/15/2013] [Indexed: 01/19/2023] Open
Abstract
Beyond their critical role in humoral immunity, B lymphocytes can employ a variety of immunomodulatory mechanisms including expression of the apoptosis-inducing molecule Fas ligand (FasL; CD178). Here, we extensively characterized the surface phenotype of FasL+ killer B cells, showing they are enriched in the IgMhighCD5+CD1dhigh B cell subset previously reported to contain a higher frequency of B cells producing interleukin-10 (IL-10). A rare population of B cells expressing IL-10 was present among FasL+ B cells, but most FasL+ B cells did not produce IL-10. We also identify interleukin-5 (IL-5) as a novel inducer of killer B cell function. Constitutively FasL+ B cells expressed higher levels of the IL-5 receptor, and treating B cells with IL-5 and CD40L resulted in the expansion of a B cell population enriched for FasL+ cells. B cells stimulated with IL-5 and CD40L were potent inducers of apoptosis in activated primary CD4+ T cells, and this killing function was antigen-specific and dependent upon FasL. IL-5 also enhanced IL-10 secretion in B cells stimulated with CD40L. Taken together these findings elucidate the relationship of FasL+ B cells and IL-10-producing B cells and demonstrate that IL-5 can induce or enhance both killer B cell activity and IL-10 secretion in B cells. Finally, we found that the killer B cell activity induced by IL-5 was completely blocked by IL-4, suggesting the existence of a previously unknown antagonistic relationship between these type-2 cytokines in modulating the activity of killer B cells. Targeting this IL-5/IL-4 signaling axis may therefore represent a novel area of drug discovery in inflammatory disorders.
Collapse
Affiliation(s)
- Matthew W. Klinker
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Tamra J. Reed
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - David A. Fox
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Steven K. Lundy
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|