1
|
Moniot A, Schneider C, Chardin L, Yaniz-Galende E, Genestie C, Etiennot M, Henry A, Drelon C, Le Formal A, Langlois B, Venat L, Louvet C, Favier L, Lortholary A, Berton-Rigaud D, Dohollou N, Desauw C, Fabbro M, Malaurie E, Dubot C, Kurtz JE, Bonichon Lamichhane N, Pujade-Lauraine É, Jeanne A, Leary A, Dedieu S. The CD47/TSP-1 axis: a promising avenue for ovarian cancer treatment and biomarker research. Mol Cancer 2024; 23:166. [PMID: 39138571 PMCID: PMC11323699 DOI: 10.1186/s12943-024-02073-0] [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: 05/13/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) remains one of the most challenging and deadly malignancies facing women today. While PARP inhibitors (PARPis) have transformed the treatment landscape for women with advanced OC, many patients will relapse and the PARPi-resistant setting is an area of unmet medical need. Traditional immunotherapies targeting PD-1/PD-L1 have failed to show any benefit in OC. The CD47/TSP-1 axis may be relevant in OC. We aimed to describe changes in CD47 expression with platinum therapy and their relationship with immune features and prognosis. METHODS Tumor and blood samples collected from OC patients in the CHIVA trial were assessed for CD47 and TSP-1 before and after neoadjuvant chemotherapy (NACT) and multiplex analysis was used to investigate immune markers. Considering the therapeutic relevance of targeting the CD47/TSP-1 axis, we used the CD47-derived TAX2 peptide to selectively antagonize it in a preclinical model of aggressive ovarian carcinoma. RESULTS Significant reductions in CD47 expression were observed post NACT. Tumor patients having the highest CD47 expression profile at baseline showed the greatest CD4+ and CD8+ T-cell influx post NACT and displayed a better prognosis. In addition, TSP-1 plasma levels decreased significantly under NACT, and high TSP-1 was associated with a worse prognosis. We demonstrated that TAX2 exhibited a selective and favorable biodistribution profile in mice, localizing at the tumor sites. Using a relevant peritoneal carcinomatosis model displaying PARPi resistance, we demonstrated that post-olaparib (post-PARPi) administration of TAX2 significantly reduced tumor burden and prolonged survival. Remarkably, TAX2 used sequentially was also able to increase animal survival even under treatment conditions allowing olaparib efficacy. CONCLUSIONS Our study thus (1) proposes a CD47-based stratification of patients who may be most likely to benefit from postoperative immunotherapy, and (2) suggests that TAX2 is a potential alternative therapy for patients relapsing on PARP inhibitors.
Collapse
Affiliation(s)
| | | | - Laure Chardin
- Gustave-Roussy Cancer Campus Université Paris-Saclay GINECO/GINEGEPS, Inserm U981, Villejuif, France
| | - Elisa Yaniz-Galende
- Gustave-Roussy Cancer Campus Université Paris-Saclay GINECO/GINEGEPS, Inserm U981, Villejuif, France
| | - Catherine Genestie
- Gustave-Roussy Cancer Campus Université Paris-Saclay GINECO/GINEGEPS, Inserm U981, Villejuif, France
| | | | | | - Coralie Drelon
- UMR 7369 MEDyC, CNRS, Université de Reims Champagne-Ardenne, Reims, France
| | - Audrey Le Formal
- Gustave-Roussy Cancer Campus Université Paris-Saclay GINECO/GINEGEPS, Inserm U981, Villejuif, France
| | - Benoit Langlois
- UMR 7369 MEDyC, CNRS, Université de Reims Champagne-Ardenne, Reims, France
| | - Laurence Venat
- Centre Hospitalier Universitaire Dupuytren, Limoges, France
| | | | | | | | | | | | - Christophe Desauw
- Centre Hospitalier Régional Universitaire de Lille, Hôpital Huriez, Lille, France
| | | | | | - Coraline Dubot
- Institut Curie - Hôpital René Huguenin - GINECO, Saint-Cloud, France
| | | | | | | | | | - Alexandra Leary
- Gustave-Roussy Cancer Campus Université Paris-Saclay GINECO/GINEGEPS, Inserm U981, Villejuif, France
| | - Stéphane Dedieu
- UMR 7369 MEDyC, CNRS, Université de Reims Champagne-Ardenne, Reims, France.
| |
Collapse
|
2
|
Xing L, Wang Z, Feng Y, Luo H, Dai G, Sang L, Zhang C, Qian J. The biological roles of CD47 in ovarian cancer progression. Cancer Immunol Immunother 2024; 73:145. [PMID: 38832992 PMCID: PMC11150368 DOI: 10.1007/s00262-024-03708-3] [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: 03/18/2024] [Accepted: 04/19/2024] [Indexed: 06/06/2024]
Abstract
Ovarian cancer is one of the most lethal malignant tumors, characterized by high incidence and poor prognosis. Patients relapse occurred in 65-80% after initial treatment. To date, no effective treatment has been established for these patients. Recently, CD47 has been considered as a promising immunotherapy target. In this paper, we reviewed the biological roles of CD47 in ovarian cancer and summarized the related mechanisms. For most types of cancers, the CD47/Sirpα immune checkpoint has attracted the most attention in immunotherapy. Notably, CD47 monoclonal antibodies and related molecules are promising in the immunotherapy of ovarian cancer, and further research is needed. In the future, new immunotherapy regimens targeting CD47 can be applied to the clinical treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Linan Xing
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Zhao Wang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Yue Feng
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Haixia Luo
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Guijiang Dai
- Department of Comprehensive Office, The Second Affiliated Hospital of MuDanjiang Medical University, Mudanjiang, 157009, People's Republic of China
| | - Lin Sang
- Department of Obstetrics and Gynecology, People's Hospital of Anji, Huzhou, 310022, People's Republic of China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
3
|
Ashaq MS, Zhang S, Xu M, Li Y, Zhao B. The regulatory role of CD36 in hematopoiesis beyond fatty acid uptake. Life Sci 2024; 339:122442. [PMID: 38244916 DOI: 10.1016/j.lfs.2024.122442] [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: 11/20/2023] [Revised: 01/07/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
CD36 is a transmembrane glycoprotein, located on surface of numerous cell types. This review is aimed to explore regulatory role of CD36 in hematopoiesis beyond fatty acid uptake. CD36 acts as a pattern recognition receptor, regulates cellular fatty acid homeostasis, and negatively monitors angiogenesis. CD36 also mediates free fatty acid transportation to hematopoietic stem cells in response to infections. During normal physiology and pathophysiology, CD36 significantly participates in the activation and metabolic needs of platelets, macrophages, monocytes, T cells, B cells, and dendritic cells. CD36 has shown a unique relationship with Plasmodium falciparum-infected erythrocytes (PfIEs) as a beneficiary for both parasite and host. CD36 actively participates in pathogenesis of various hematological cancers as a significant prognostic biomarker including AML, HL, and NHL. CD36-targeting antibodies, CD36 antagonists (small molecules), and CD36 expression inhibitors/modulators are used to target CD36, depicting its therapeutic potential. Many preclinical studies or clinical trials were performed to assess CD36 as a therapeutic target; some are still under investigation. This review reflects the role of CD36 in hematopoiesis which requires more consideration in future research.
Collapse
Affiliation(s)
- Muhammad Sameer Ashaq
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shujing Zhang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Miaomiao Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuan Li
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Baobing Zhao
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
4
|
Mackert JD, Stirling ER, Wilson AS, Westwood B, Zhao D, Lo HW, Metheny-Barlow L, Cook KL, Lesser GJ, Soto-Pantoja DR. Anti-CD47 immunotherapy as a therapeutic strategy for the treatment of breast cancer brain metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550566. [PMID: 37546807 PMCID: PMC10402073 DOI: 10.1101/2023.07.25.550566] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The presence of cell surface protein CD47 allows cancer cells to evade innate and adaptive immune surveillance resulting in metastatic spread. CD47 binds to and activates SIRPα on the surface of myeloid cells, inhibiting their phagocytic activity. On the other hand, CD47 binds the matricellular protein Thrombospondin-1, limiting T-cell activation. Thus, blocking CD47 is a potential therapeutic strategy for preventing brain metastasis. To test this hypothesis, breast cancer patient biopsies were stained with antibodies against CD47 to determine differences in protein expression. An anti-CD47 antibody was used in a syngeneic orthotopic triple-negative breast cancer model, and CD47 null mice were used in a breast cancer brain metastasis model by intracardiac injection of the E0771-Br-Luc cell line. Immunohistochemical staining of patient biopsies revealed an 89% increase in CD47 expression in metastatic brain tumors compared to normal adjacent tissue (p ≤ 0.05). Anti-CD47 treatment in mice bearing brain metastatic 4T1br3 orthotopic tumors reduced tumor volume and tumor weight by over 50% compared to control mice (p ≤ 0.05) and increased IBA1 macrophage/microglia marker 5-fold in tumors compared to control (p ≤ 0.05). Additionally, CD47 blockade increased the M1/M2 macrophage ratio in tumors 2.5-fold (p ≤ 0.05). CD47 null mice had an 89% decrease in metastatic brain burden (p ≤ 0.05) compared to control mice in a brain metastasis model. Additionally, RNA sequencing revealed several uniquely expressed genes and significantly enriched genes related to tissue development, cell death, and cell migration tumors treated with anti-CD47 antibodies. Thus, demonstrating that CD47 blockade affects cancer cell and tumor microenvironment signaling to limit metastatic spread and may be an effective therapeutic for triple-negative breast cancer brain metastasis.
Collapse
|
5
|
Rubanov A, Berico P, Hernando E. Epigenetic Mechanisms Underlying Melanoma Resistance to Immune and Targeted Therapies. Cancers (Basel) 2022; 14:cancers14235858. [PMID: 36497341 PMCID: PMC9738385 DOI: 10.3390/cancers14235858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is an aggressive skin cancer reliant on early detection for high likelihood of successful treatment. Solar UV exposure transforms melanocytes into highly mutated tumor cells that metastasize to the liver, lungs, and brain. Even upon resection of the primary tumor, almost thirty percent of patients succumb to melanoma within twenty years. Identification of key melanoma genetic drivers led to the development of pharmacological BRAFV600E and MEK inhibitors, significantly improving metastatic patient outcomes over traditional cytotoxic chemotherapy or pioneering IFN-α and IL-2 immune therapies. Checkpoint blockade inhibitors releasing the immunosuppressive effects of CTLA-4 or PD-1 proved to be even more effective and are the standard first-line treatment. Despite these major improvements, durable responses to immunotherapy and targeted therapy have been hindered by intrinsic or acquired resistance. In addition to gained or selected genetic alterations, cellular plasticity conferred by epigenetic reprogramming is emerging as a driver of therapy resistance. Epigenetic regulation of chromatin accessibility drives gene expression and establishes distinct transcriptional cell states. Here we review how aberrant chromatin, transcriptional, and epigenetic regulation contribute to therapy resistance and discuss how targeting these programs sensitizes melanoma cells to immune and targeted therapies.
Collapse
Affiliation(s)
- Andrey Rubanov
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Pietro Berico
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
6
|
Stirling ER, Terabe M, Wilson AS, Kooshki M, Yamaleyeva LM, Alexander-Miller MA, Zhang W, Miller LD, Triozzi PL, Soto-Pantoja DR. Targeting the CD47/thrombospondin-1 signaling axis regulates immune cell bioenergetics in the tumor microenvironment to potentiate antitumor immune response. J Immunother Cancer 2022; 10:e004712. [PMID: 36418073 PMCID: PMC9685258 DOI: 10.1136/jitc-2022-004712] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND CD47 is an integral membrane protein that alters adaptive immunosurveillance when bound to the matricellular glycoprotein thrombospondin-1 (TSP1). We examined the impact of the CD47/TSP1 signaling axis on melanoma patient response to anti-PD-1 therapy due to alterations in T cell activation, proliferation, effector function, and bioenergetics. METHODS A syngeneic B16 mouse melanoma model was performed to determine if targeting CD47 as monotherapy or in combination with anti-PD-1 impacted tumor burden. Cytotoxic (CD8+) T cells from Pmel-1 transgenic mice were used for T cell activation, cytotoxic T lymphocyte, and cellular bioenergetic assays. Single-cell RNA-sequencing, ELISA, and flow cytometry was performed on peripheral blood mononuclear cells and plasma of melanoma patients receiving anti-PD-1 therapy to examine CD47/TSP1 expression. RESULTS Human malignant melanoma tissue had increased CD47 and TSP1 expression within the tumor microenvironment compared with benign tissue. Due to the negative implications CD47/TSP1 can have on antitumor immune responses, we targeted CD47 in a melanoma model and observed a decrease in tumor burden due to increased tumor oxygen saturation and granzyme B secreting CD8+ T cells compared with wild-type tumors. Additionally, Pmel-1 CD8+ T cells exposed to TSP1 had reduced activation, proliferation, and effector function against B16 melanoma cells. Targeting CD47 allowed CD8+ T cells to overcome this TSP1 interaction to sustain these functions. TSP1 exposed CD8+ T cells have a decreased rate of glycolysis; however, targeting CD47 restored glycolysis when CD8+ T cells were exposed to TSP1, suggesting CD47 mediated metabolic reprogramming of T cells. Additionally, non-responding patients to anti-PD-1 therapy had increased T cells expressing CD47 and circulating levels of TSP1 compared with responding patients. Since CD47/TSP1 signaling axis negatively impacts CD8+ T cells and non-responding patients to anti-PD-1 therapy have increased CD47/TSP1 expression, we targeted CD47 in combination with anti-PD-1 in a melanoma model. Targeting CD47 in combination with anti-PD-1 treatment further decreased tumor burden compared with monotherapy and control. CONCLUSION CD47/TSP1 expression could serve as a marker to predict patient response to immune checkpoint blockade treatment, and targeting this pathway may preserve T cell activation, proliferation, effector function, and bioenergetics to reduce tumor burden as a monotherapy or in combination with anti-PD-1.
Collapse
Affiliation(s)
- Elizabeth R Stirling
- Department of Cancer Biology, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
| | - Masaki Terabe
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Adam S Wilson
- Department of Surgery, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
| | - Mitra Kooshki
- Department of Hematology & Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Radiation Oncology, Wake Forest University School of Medicine, WInston-Salem, North Carolina, USA
| | - Liliya M Yamaleyeva
- Department of Surgery, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
| | - Martha A Alexander-Miller
- Department of Microbiology & Immunology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Wei Zhang
- Department of Cancer Biology, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
- Atrium Health Wake Forest Baptist Medical Center, Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
- Atrium Health Wake Forest Baptist Medical Center, Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| | - Pierre L Triozzi
- Department of Cancer Biology, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
- Department of Hematology & Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Atrium Health Wake Forest Baptist Medical Center, Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| | - David R Soto-Pantoja
- Department of Cancer Biology, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
- Department of Surgery, Wake Forest Univerisity School of Medicine, Winston-Salem, North Carolina, USA
- Department of Radiation Oncology, Wake Forest University School of Medicine, WInston-Salem, North Carolina, USA
- Atrium Health Wake Forest Baptist Medical Center, Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| |
Collapse
|
7
|
Witkowski J, Polak S, Rogulski Z, Pawelec D. In Vitro/In Vivo Translation of Synergistic Combination of MDM2 and MEK Inhibitors in Melanoma Using PBPK/PD Modelling: Part II. Int J Mol Sci 2022; 23:11939. [PMID: 36233247 PMCID: PMC9570053 DOI: 10.3390/ijms231911939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/23/2022] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
Abstract
The development of in vitro/in vivo translational methods for synergistically acting drug combinations is needed to identify the most effective therapeutic strategies. We performed PBPK/PD modelling for siremadlin, trametinib, and their combination at various dose levels and dosing schedules in an A375 xenografted mouse model (melanoma cells). In this study, we built models based on in vitro ADME and in vivo PK/PD data determined from the literature or estimated by the Simcyp Animal simulator (V21). The developed PBPK/PD models allowed us to account for the interactions between siremadlin and trametinib at PK and PD levels. The interaction at the PK level was described by an interplay between absorption and tumour disposition levels, whereas the PD interaction was based on the in vitro results. This approach allowed us to reasonably estimate the most synergistic and efficacious dosing schedules and dose levels for combinations of siremadlin and trametinib in mice. PBPK/PD modelling is a powerful tool that allows researchers to properly estimate the in vivo efficacy of the anticancer drug combination based on the results of in vitro studies. Such an approach based on in vitro and in vivo extrapolation may help researchers determine the most efficacious dosing strategies and will allow for the extrapolation of animal PBPK/PD models into clinical settings.
Collapse
Affiliation(s)
- Jakub Witkowski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
- Adamed Pharma S.A., Adamkiewicza 6a, 05-152 Czosnów, Poland
| | - Sebastian Polak
- Faculty of Pharmacy, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland
- Simcyp Division, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Zbigniew Rogulski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | | |
Collapse
|
8
|
Targeting Ovarian Carcinoma with TSP-1:CD47 Antagonist TAX2 Activates Anti-Tumor Immunity. Cancers (Basel) 2021; 13:cancers13195019. [PMID: 34638503 PMCID: PMC8508526 DOI: 10.3390/cancers13195019] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/02/2021] [Accepted: 10/03/2021] [Indexed: 11/17/2022] Open
Abstract
TAX2 peptide is a cyclic peptide that acts as an orthosteric antagonist for thrombospondin-1 (TSP-1) interaction with CD47. TAX2 was first described for its anti-angiogenic activities and showed anti-cancer efficacy in numerous preclinical models. Here, we aimed at providing an extensive molecular characterization of TAX2 mode of action, while evaluating its potential in ovarian cancer therapy. Multidisciplinary approaches were used to qualify a TAX2 drug candidate in terms of stability, solubility and potency. Then, efficacy studies, together with benchmark experiments, were performed in relevant mouse models of ovarian carcinoma. TAX2 peptide appears to be stable and soluble in clinically relevant solvents, while displaying a favorable safety profile. Moreover, clinical data mining allowed for the identification of TSP-1 as a relevant pharmacological target in ovarian cancer. In mice, TAX2 therapy inhibits ovarian tumor growth and metastatic dissemination, while activating anti-cancer adaptive immunity. Interestingly, TAX2 also synergizes when administered in combination with anti-PD-1 immune checkpoint inhibitiors. Altogether, our data expose TAX2 as an optimized candidate with advanced preclinical characterization. Using relevant syngeneic ovarian carcinoma models, we highlighted TAX2's ability to convert poorly immunogenic tumors into ones displaying effective anti-tumor T-cell immunity.
Collapse
|
9
|
Fuselier C, Quemener S, Dufay E, Bour C, Boulagnon-Rombi C, Bouland N, Djermoune EH, Devy J, Martiny L, Schneider C. Anti-Tumoral and Anti-Angiogenic Effects of Low-Diluted Phenacetinum on Melanoma. Front Oncol 2021; 11:597503. [PMID: 33747916 PMCID: PMC7966719 DOI: 10.3389/fonc.2021.597503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/07/2021] [Indexed: 01/11/2023] Open
Abstract
Melanoma is the most aggressive form of skin cancer and the most rapidly expanding cancer in terms of worldwide incidence. If primary cutaneous melanoma is mostly treated with a curative wide local excision, malignant melanoma has a poor prognosis and needs other therapeutic approaches. Angiogenesis is a normal physiological process essential in growth and development, but it also plays a crucial role in crossing from benign to advanced state in cancer. In melanoma progression, angiogenesis is widely involved during the vertical growth phase. Currently, no anti-angiogenic agents are efficient on their own, and combination of treatments will probably be the key to success. In the past, phenacetin was used as an analgesic to relieve pain, causing side effects at large dose and tumor-inducing in humans and animals. By contrast, Phenacetinum low-dilution is often used in skin febrile exanthema, patches profusely scattered on limbs, headache, or flushed face without side effects. Herein are described the in vitro, in vivo, and ex vivo anti-angiogenic and anti-tumoral potentials of Phenacetinum low-dilution in a B16F1 tumor model and endothelial cells. We demonstrate that low-diluted Phenacetinum inhibits in vivo tumor growth and tumor vascularization and thus increases the survival time of B16F1 melanoma induced-C57BL/6 mice. Moreover, Phenacetinum modulates the lung metastasis in a B16F10 induced model. Ex vivo and in vitro, we evidence that low-diluted Phenacetinum inhibits the migration and the recruitment of endothelial cells and leads to an imbalance in the pro-tumoral macrophages and to a structural malformation of the vascular network. All together these results demonstrate highly hopeful anti-tumoral, anti-metastatic, and anti-angiogenic effects of Phenacetinum low-dilution on melanoma. Continued studies are needed to preclinically validate Phenacetinum low-dilution as a complementary or therapeutic strategy for melanoma treatment.
Collapse
Affiliation(s)
- Camille Fuselier
- Université de Reims-Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Sandrine Quemener
- Université de Lille, Institut Pasteur de Lille, U1011 INSERM, Lille, France
| | - Eleonore Dufay
- Université de Reims-Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Camille Bour
- Université de Reims-Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Camille Boulagnon-Rombi
- Université de Reims-Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
- Centre Hospitalier et Université de Reims Champagne-Ardenne, laboratoire de Biopathologie, Reims, France
| | - Nicole Bouland
- Université de Reims Champagne-Ardenne, laboratoire d’Anatomie Pathologie, Reims, France
| | | | - Jérôme Devy
- Université de Reims-Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Laurent Martiny
- Université de Reims-Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Christophe Schneider
- Université de Reims-Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| |
Collapse
|
10
|
Jeanne A, Sarazin T, Charlé M, Kawecki C, Kauskot A, Hedtke T, Schmelzer CEH, Martiny L, Maurice P, Dedieu S. Towards the Therapeutic Use of Thrombospondin 1/CD47 Targeting TAX2 Peptide as an Antithrombotic Agent. Arterioscler Thromb Vasc Biol 2021; 41:e1-e17. [PMID: 33232198 DOI: 10.1161/atvbaha.120.314571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE TSP-1 (thrombospondin 1) is one of the most expressed proteins in platelet α-granules and plays an important role in the regulation of hemostasis and thrombosis. Interaction of released TSP-1 with CD47 membrane receptor has been shown to regulate major events leading to thrombus formation, such as, platelet adhesion to vascular endothelium, nitric oxide/cGMP (cyclic guanosine monophosphate) signaling, platelet activation as well as aggregation. Therefore, targeting TSP-1:CD47 axis may represent a promising antithrombotic strategy. Approach and Results: A CD47-derived cyclic peptide was engineered, namely TAX2, that targets TSP-1 and selectively prevents TSP-1:CD47 interaction. Here, we demonstrate for the first time that TAX2 peptide strongly decreases platelet aggregation and interaction with collagen under arterial shear conditions. TAX2 also delays time for complete thrombotic occlusion in 2 mouse models of arterial thrombosis following chemical injury, while Thbs1-/- mice recapitulate TAX2 effects. Importantly, TAX2 administration is not associated with increased bleeding risk or modification of hematologic parameters. CONCLUSIONS Overall, this study sheds light on the major contribution of TSP-1:CD47 interaction in platelet activation and thrombus formation while putting forward TAX2 as an innovative antithrombotic agent with high added-value.
Collapse
Affiliation(s)
- Albin Jeanne
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
- SATT Nord, Lille, France (A.J.)
- Apmonia Therapeutics, Reims, France (A.J., S.D.)
| | - Thomas Sarazin
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Magalie Charlé
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Charlotte Kawecki
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Alexandre Kauskot
- HITh, UMR_S 1176, INSERM Univ. Paris-Sud, Université Paris-Saclay, France (A.K.)
| | - Tobias Hedtke
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany (T.H., C.E.H.S.)
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany (T.H., C.E.H.S.)
| | - Laurent Martiny
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Stéphane Dedieu
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
- Apmonia Therapeutics, Reims, France (A.J., S.D.)
| |
Collapse
|
11
|
Wang J, Li Y. CD36 tango in cancer: signaling pathways and functions. Theranostics 2019; 9:4893-4908. [PMID: 31410189 PMCID: PMC6691380 DOI: 10.7150/thno.36037] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/12/2019] [Indexed: 12/22/2022] Open
Abstract
CD36, a scavenger receptor expressed in multiple cell types, mediates lipid uptake, immunological recognition, inflammation, molecular adhesion, and apoptosis. CD36 is a transmembrane glycoprotein that contains several posttranslational modification sites and binds to diverse ligands, including apoptotic cells, thrombospondin-1 (TSP-1), and fatty acids (FAs). Beyond fueling tumor metastasis and therapy resistance by enhancing lipid uptake and FA oxidation, CD36 attenuates angiogenesis by binding to TSP-1 and thereby inducing apoptosis or blocking the vascular endothelial growth factor receptor 2 pathway in tumor microvascular endothelial cells. Moreover, CD36-driven lipid metabolic reprogramming and functions in tumor-associated immune cells lead to tumor immune tolerance and cancer development. Notable advances have been made in demonstrating the regulatory networks that govern distinct physiological properties of CD36, and this has identified targeting CD36 as a potential strategy for cancer treatment. Here, we provide an overview on the structure, regulation, ligands, functions, and clinical trials of CD36 in cancer.
Collapse
|
12
|
Orouji E, Federico A, Larribère L, Novak D, Lipka DB, Assenov Y, Sachindra S, Hüser L, Granados K, Gebhardt C, Plass C, Umansky V, Utikal J. Histone methyltransferase SETDB1 contributes to melanoma tumorigenesis and serves as a new potential therapeutic target. Int J Cancer 2019; 145:3462-3477. [PMID: 31131878 DOI: 10.1002/ijc.32432] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/06/2019] [Indexed: 12/29/2022]
Abstract
Alterations in histone modifications play a crucial role in the progression of various types of cancer. The histone methyltransferase SETDB1 catalyzes the addition of methyl groups to histone H3 at lysine 9. Here, we describe how overexpression of SETDB1 contributes to melanoma tumorigenesis. SETDB1 is highly amplified in melanoma cells and in the patient tumors. Increased expression of SETDB1, which correlates with SETDB1 amplification, is associated with a more aggressive phenotype in in vitro and in vivo studies. Mechanistically, SETDB1 implements its effects via regulation of thrombospondin 1, and the SET-domain of SETDB1 is essential for the maintenance of its tumorigenic activity. Inhibition of SETDB1 reduces cell growth in melanomas resistant to targeted treatments. Our results indicate that SETDB1 is a major driver of melanoma development and may serve as a potential future target for the treatment of this disease.
Collapse
Affiliation(s)
- Elias Orouji
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Aniello Federico
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Lionel Larribère
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Daniel Novak
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Daniel B Lipka
- Division of Epigenomics and Cancer Risk Factors, DKFZ, Heidelberg, Baden Württemberg, Germany
| | - Yassen Assenov
- Division of Epigenomics and Cancer Risk Factors, DKFZ, Heidelberg, Baden Württemberg, Germany
| | - Sachindra Sachindra
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Laura Hüser
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Karol Granados
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Christoffer Gebhardt
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany.,Department of Dermatology and Venereology, University Hospital Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Christoph Plass
- Division of Epigenomics and Cancer Risk Factors, DKFZ, Heidelberg, Baden Württemberg, Germany
| | - Viktor Umansky
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| | - Jochen Utikal
- German Cancer Research Center (DKFZ), Skin Cancer Unit, Heidelberg, Baden Württemberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Baden Württemberg, Germany
| |
Collapse
|
13
|
Zhao X, Song X, Bai X, Tan Z, Ma X, Guo J, Zhang Z, Du Q, Huang Y, Tong D. microRNA-222 Attenuates Mitochondrial Dysfunction During Transmissible Gastroenteritis Virus Infection. Mol Cell Proteomics 2019; 18:51-64. [PMID: 30257878 PMCID: PMC6317483 DOI: 10.1074/mcp.ra118.000808] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/13/2018] [Indexed: 12/30/2022] Open
Abstract
Transmissible gastroenteritis virus (TGEV) is a member of Coronaviridae family. Our previous research showed that TGEV infection could induce mitochondrial dysfunction and upregulate miR-222 level. Therefore, we presumed that miR-222 might be implicated in regulating mitochondrial dysfunction induced by TGEV infection. To verify the hypothesis, the effect of miR-222 on mitochondrial dysfunction was tested and we showed that miR-222 attenuated TGEV-induced mitochondrial dysfunction. To investigate the underlying molecular mechanism of miR-222 in TGEV-induced mitochondrial dysfunction, a quantitative proteomic analysis of PK-15 cells that were transfected with miR-222 mimics and infected with TGEV was performed. In total, 4151 proteins were quantified and 100 differentially expressed proteins were obtained (57 upregulated, 43 downregulated), among which thrombospondin-1 (THBS1) and cluster of differentiation 47 (CD47) were downregulated. THBS1 was identified as the target of miR-222. Knockdown of THBS1 and CD47 decreased mitochondrial Ca2+ level and increased mitochondrial membrane potential (MMP) level. Reversely, overexpression of THBS1 and CD47 elevated mitochondrial Ca2+ level and reduced mitochondrial membrane potential (MMP) level. Together, our data establish a significant role of miR-222 in regulating mitochondrial dysfunction in response to TGEV infection.
Collapse
Affiliation(s)
- Xiaomin Zhao
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Xiangjun Song
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Xiaoyuan Bai
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Zhanhang Tan
- §Huyi District Center for Animal Disease Control and Prevention, Xi'an, Shaanxi 710300, P.R. China
| | - Xuelian Ma
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Jianxiong Guo
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Zhichao Zhang
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Qian Du
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Yong Huang
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Dewen Tong
- From the ‡College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China;.
| |
Collapse
|
14
|
Zhou XQ, Ren J, Yang S. [Relationship between thrombospondin-1 and the occurrence and development of oral and maxillofacial malignancy]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2018; 36:686-690. [PMID: 30593119 DOI: 10.7518/hxkq.2018.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Thrombospondin-1 (TSP-1) is widely distributed in human tissues and is important in inhibiting angiogenesis.It also occupies an indispensable position in the formation, growth, differentiation, and metastasis of tumors in different tissues.TSP-1 plays an important role in the occurrence and development of various types of tumors. The inhibitory effect of TSP-1 on the angiogenesis and tumor development of oral and maxillofacial malignant tumors has been demonstrated in recent years. This paper reviews the findings and progress of TSP-1 research involving all kinds of tumors as well as oral and maxillofacial malignancies.
Collapse
Affiliation(s)
- Xue-Qin Zhou
- Graduate School, Zunyi Medical University, Zunyi 563000, China;Dept. of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining 629000, China
| | - Jun Ren
- Graduate School, Zunyi Medical University, Zunyi 563000, China;Dept. of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining 629000, China
| | - Sen Yang
- Graduate School, Zunyi Medical University, Zunyi 563000, China;Dept. of Oral and Maxillofacial Surgery, Suining Central Hospital, Suining 629000, China
| |
Collapse
|
15
|
Abstract
Vascular remodeling defines cancer growth and aggressiveness. Although cancer cells produce pro-angiogenic signals, the fate of angiogenesis critically depends on the cancer microenvironment. Composition of the extracellular matrix (ECM) and tumor inflammation determine whether a cancer will remain dormant, will be recognized by the immune system and eliminated, or whether the tumor will develop and lead to the spread and metastasis of cancer cells. Thrombospondins (TSPs), a family of ECM proteins that has long been associated with the regulation of angiogenesis and cancer, regulate multiple physiological processes that determine cancer growth and spreading, from angiogenesis to inflammation, metabolic changes, and properties of ECM. Here, we sought to review publications that describe various functions of TSPs that link these proteins to regulation of cancer growth by modulating multiple physiological and pathological events that prevent or support tumor development. In addition to its direct effects on angiogenesis, TSPs have important roles in regulation of inflammation, immunity, ECM properties and composition, and glucose and insulin metabolism. Furthermore, TSPs have distinct roles as regulators of remodeling in tissues and tumors, such that the pathways activated by a single TSP can interact and influence each other. The complex nature of TSP interactions and functions, including their different cell- and tissue-specific effects, may lead to confusing results and controversial conclusions when taken out of the context of interdisciplinary and holistic approaches. However, studies of TSP functions and roles in different systems of the organism offer an integrative view of tumor remodeling and a potential for finding therapeutic targets that would modulate multiple complementary processes associated with cancer growth.
Collapse
Affiliation(s)
| | - Santoshi Muppala
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, 44195, USA
| | - Jasmine Gajeton
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, 44195, USA
| |
Collapse
|
16
|
Lumican delays melanoma growth in mice and drives tumor molecular assembly as well as response to matrix-targeted TAX2 therapeutic peptide. Sci Rep 2017; 7:7700. [PMID: 28794454 PMCID: PMC5550434 DOI: 10.1038/s41598-017-07043-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/21/2017] [Indexed: 02/07/2023] Open
Abstract
Lumican is a small leucine-rich proteoglycan (SLRP) being known as a key regulator of collagen fibrillogenesis. However, little attention has been given so far in studying its influence on tumor-associated matrix architecture. Here, we investigate the role of host lumican on tumor matrix organization as well as on disease progression considering an immunocompetent model of melanoma implanted in Lum -/- vs. wild type syngeneic mice. Conjointly, lumican impact on tumor response to matrix-targeted therapy was evaluated considering a previously validated peptide, namely TAX2, that targets matricellular thrombospondin-1. Analysis of available genomics and proteomics databases for melanoma first established a correlation between lumican expression and patient outcome. In the B16 melanoma allograft model, endogenous lumican inhibits tumor growth and modulates response to TAX2 peptide. Indeed, IHC analyses revealed that lumican deficiency impacts intratumoral distribution of matricellular proteins, growth factor and stromal cells. Besides, innovative imaging approaches helped demonstrating that lumican host expression drives biochemical heterogeneity of s.c. tumors, while modulating intratumoral collagen deposition as well as organization. Altogether, the results obtained present lumican as a strong endogenous inhibitor of tumor growth, while identifying for the first time this proteoglycan as a major driver of tumor matrix coherent assembly.
Collapse
|
17
|
Jeanne A, Martiny L, Dedieu S. Thrombospondin-targeting TAX2 peptide impairs tumor growth in preclinical mouse models of childhood neuroblastoma. Pediatr Res 2017; 81:480-488. [PMID: 27842053 DOI: 10.1038/pr.2016.242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/15/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND We have previously identified TAX2 peptide as an orthosteric antagonist for thrombospondin-1 (TSP-1) interaction with the cell-surface receptor CD47. TAX2 displays exciting antiangiogenic, antitumor, and antimetastatic properties in both allograft and xenograft models of melanoma as well as pancreatic carcinoma. Here, TAX2 therapeutic potential was investigated in two distinct preclinical mouse models of neuroblastoma. METHODS SK-N-BE(2) (MYCN-amplified) and SK-N-SH (MYCN-negative) human neuroblastoma cells have been implanted in outbred NMRI nude mice prior to systemic administrations of TAX2, and then tumor growth as well as intratumoral blood flow were longitudinally monitored. At study termination, subcutaneous xenografts were macroscopically and histopathologically examined. RESULTS In both models, TAX2 induced a significant inhibition of tumor burden in mice engrafted with large pre-established neuroblastoma tumors. Indeed, TAX2 administered at biologically relevant doses sharply alters xenograft vascularization as well as multiple features of tumor progression. CONCLUSION Altogether, our results present TAX2 peptide specifically targeting TSP-1:CD47 interaction as a new putative therapeutic approach for treating neuroblastoma, whether utilized alone or in combination with existing chemotherapy drugs.
Collapse
Affiliation(s)
- Albin Jeanne
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France.,SATT Nord, Lille, France
| | - Laurent Martiny
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| | - Stéphane Dedieu
- Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire, MEDyC, Reims, France
| |
Collapse
|