1
|
David T, du Roure PD, Mallavialle A, Laurent-Matha V, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Cathepsins: Novel opportunities for antibody therapeutics in cancer. Br J Pharmacol 2025. [PMID: 39834229 DOI: 10.1111/bph.17437] [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: 08/08/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/22/2025] Open
Abstract
Cathepsins, the most abundant lysosomal proteases, have key functions in cell maintenance and homeostasis. They are overexpressed and hypersecreted in cancer and associated with poor prognosis. Secreted cathepsins display pro-tumour activities in the tumour microenvironment and thus represent interesting molecular targets in oncology. Recently, several antibody-based cancer therapies have targeted the pro-tumour activity of the extracellular cathepsin pool, altering several cancer hallmarks, but not the intracellular cathepsin levels that are often crucial for cell homeostasis. In this mini-review, we describe advances in antibodies against extracellular cathepsins in cancer, and their effect on the proteolytic cascade, matrix remodelling, proliferation, and modulation of the anti-cancer immune response. We also discuss the add-on value of combination strategies (anti-cathepsin antibodies with chemotherapy and/or biologics) that make anti-cathepsin antibodies a new opportunity for disease management.
Collapse
Affiliation(s)
- Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Centre National de la Recherche Scientifique, CNRS, Paris, France
| | | |
Collapse
|
2
|
Desroys du Roure P, David T, Mallavialle A, Laurent-Matha V, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Antibodies against the multifaceted cathepsin D protein open new avenues for TNBC immunotherapy. J Immunother Cancer 2025; 13:e009548. [PMID: 39800383 PMCID: PMC11748927 DOI: 10.1136/jitc-2024-009548] [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: 04/26/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous breast cancer subtype characterized by aggressive clinical behavior and poor prognosis. The immune landscape associated with TNBC often reveals high immunogenicity. Therefore, immunotherapy, which has demonstrated its efficacy in different cancer types, could be a promising strategy for TNBC, given the limited therapeutic options currently available besides conventional chemotherapy. The aspartic protease cathepsin D (cath-D) is a tumor cell-associated extracellular protein with protumor activity, a marker of poor prognosis, and a target for antibody-based therapy in TNBC. This commentary provides a synopsis/narrative summary of the development of anti-cath-D antibodies in different formats, their key roles in restoring the antitumor immunity, particularly via activation of tumor-infiltrating natural killer cells, and their dual antitumor effects on cancer cells and stromal cancer-associated fibroblasts, suggesting their interest for clinical use in the light of the current clinical knowledge on TNBC.
Collapse
Affiliation(s)
| | - Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre National de la Recherche Scientifique, Paris, F-75016, France
| | | |
Collapse
|
3
|
Nambiar SS, Ghosh SS, Saini GK. Gliotoxin triggers cell death through multifaceted targeting of cancer-inducing genes in breast cancer therapy. Comput Biol Chem 2024; 112:108170. [PMID: 39146703 DOI: 10.1016/j.compbiolchem.2024.108170] [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: 06/18/2024] [Revised: 08/03/2024] [Accepted: 08/03/2024] [Indexed: 08/17/2024]
Abstract
Fungal secondary metabolites have a long history of contributing to pharmaceuticals, notably in the development of antibiotics and immunosuppressants. Harnessing their potent bioactivities, these compounds are now being explored for cancer therapy, by targeting and disrupting the genes that induce cancer progression. The current study explores the anticancer potential of gliotoxin, a fungal secondary metabolite, which encompasses a multi-faceted approach integrating computational predictions, molecular dynamics simulations, and comprehensive experimental validations. In-silico studies have identified potential gliotoxin targets, including MAPK1, NFKB1, HIF1A, TDP1, TRIM24, and CTSD which are involved in critical pathways in cancer such as the NF-κB signaling pathway, MAPK/ERK signaling pathway, hypoxia signaling pathway, Wnt/β-catenin pathway, and other essential cellular processes. The gene expression analysis results indicated all the identified targets are overexpressed in various breast cancer subtypes. Subsequent molecular docking and dynamics simulations have revealed stable binding of gliotoxin with TDP1 and HIF1A. Cell viability assays exhibited a dose-dependent decreasing pattern with its remarkable IC50 values of 0.32, 0.14, and 0.53 μM for MDA-MB-231, MDA-MB-468, and MCF-7 cells, respectively. Likewise, in 3D tumor spheroids, gliotoxin exhibited a notable decrease in viability indicating its effectiveness against solid tumors. Furthermore, gene expression studies using Real-time PCR revealed a reduction of expression of cancer-inducing genes, MAPK1, HIF1A, TDP1, and TRIM24 upon gliotoxin treatment. These findings collectively underscore the promising anticancer potential of gliotoxin through multi-targeting cancer-promoting genes, positioning it as a promising therapeutic option for breast cancer.
Collapse
Affiliation(s)
- Sujisha S Nambiar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahat, Assam 39, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahat, Assam 39, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam 39, India
| | - Gurvinder Kaur Saini
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahat, Assam 39, India.
| |
Collapse
|
4
|
Pulido-Capiz A, Chimal-Vega B, Avila-Barrientos LP, Campos-Valenzuela A, Díaz-Molina R, Muñiz-Salazar R, Galindo-Hernández O, García-González V. Auraptene Boosts the Efficacy of the Tamoxifen Metabolites Endoxifen and 4-OH-Tamoxifen in a Chemoresistant ER+ Breast Cancer Model. Pharmaceutics 2024; 16:1179. [PMID: 39339215 PMCID: PMC11435248 DOI: 10.3390/pharmaceutics16091179] [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: 07/07/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Approximately 80% of breast cancer (BC) cases are estrogen receptor positive (ER+) and sensitive to hormone treatment; Tamoxifen is a prodrug, and its main plasmatic active metabolites are 4-hydroxytamoxifen (4-OH Tam) and endoxifen. Despite the effectiveness of tamoxifen therapy, resistance can be developed. An increment in eukaryotic initiation factor-4A complex (eIF4A) activity can result in tamoxifen-resistant tumor cells. For this work, we developed a cell variant resistant to 4-OH Tam and endoxifen, denominated MCF-7Var E; then, the aim of this research was to reverse the acquired resistance of this variant to tamoxifen metabolites by incorporating the natural compound auraptene. Combination treatments of tamoxifen derivatives and auraptene successfully sensitized the chemoresistant MCF-7Var E. Our data suggest a dual regulation of eIF4A and ER by auraptene. Joint treatments of 4-OH Tam and endoxifen with auraptene identified a novel focus for chemoresistance disruption. Synergy was observed using the auraptene molecule and tamoxifen-derived metabolites, which induced a sensitization in MCF-7Var E cells and ERα parental cells that was not observed in triple-negative breast cancer cells (TNBC). Our results suggest a synergistic effect between auraptene and tamoxifen metabolites in a resistant ER+ breast cancer model, which could represent the first step to achieving a pharmacologic strategy.
Collapse
Affiliation(s)
- Angel Pulido-Capiz
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Brenda Chimal-Vega
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | | | - Alondra Campos-Valenzuela
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Raúl Díaz-Molina
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Raquel Muñiz-Salazar
- Escuela de Ciencias de la Salud, Universidad Autónoma de Baja California, Campus Ensenada, Ensenada 22890, Mexico;
| | - Octavio Galindo-Hernández
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Victor García-González
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| |
Collapse
|
5
|
Alhudiri I, Nolan C, Ellis I, Elzagheid A, Green A, Chapman C. Expression of Cathepsin D in early-stage breast cancer and its prognostic and predictive value. Breast Cancer Res Treat 2024; 206:143-153. [PMID: 38578521 PMCID: PMC11182851 DOI: 10.1007/s10549-024-07293-y] [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: 12/09/2023] [Accepted: 02/08/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE Cathepsin D is a proteolytic enzyme that is normally localized in the lysosomes and is involved in the malignant progression of breast cancer. There are conflicting results regarding Cathepsin D significance as prognostic and predictor marker in breast cancer. This study aimed to evaluate the expression and prognostic significance of Cathepsin D in early-stage breast cancer. METHODS Expression of Cathepsin D was assessed by immunohistochemical staining of tissue microarrays, in a large well-characterized series of early-stage operable breast cancer (n = 954) from Nottingham Primary Breast Carcinoma Series between the period of 1988 and 1998 who underwent primary surgery. Correlation of Cathepsin D expression with clinicopathological parameters and prognosis was evaluated. RESULTS Cathepsin D expression was positive in 71.2% (679/954) of breast cancer tumours. Positive expression of Cathepsin D was significantly associated with high histological grade (p = 0.007), pleomorphism (p = 0.002), poor Nottingham Prognostic Index (NPI) score (p < 0.002), recurrence (p = 0.005) and distant metastasis (p < 0.0001). Kaplan-Meier analysis showed that Cathepsin D expression was significantly associated with shorter breast cancer-specific survival (p = 0.001), higher risk of recurrence (p = 0.001) and distant metastasis (p < 0.0001). ER-positive tumours expressing Cathepsin D and treated with tamoxifen demonstrated a significantly higher risk of distant metastasis. CONCLUSION Cathepsin D expression significantly predicts poor prognosis in breast cancer and is associated with variables of poor prognosis and shorter outcome. The strong association of Cathepsin D with aggressive tumour characteristics and poor outcomes warrants further research of its potential as a therapeutic target The results also suggest a possible interaction between Cathepsin D and tamoxifen therapy in ER-positive breast cancer which needs further investigation to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Inas Alhudiri
- Breast Pathology Research Group, Nottingham Breast Cancer Research Centre, Biodiscovery Institute, Faculty of Medicine, The University of Nottingham, Nottingham, UK
- Genetic Engineering Department, Libyan Biotechnology Research Centre, Tripoli, Libya
| | - Christopher Nolan
- Breast Pathology Research Group, Nottingham Breast Cancer Research Centre, Biodiscovery Institute, Faculty of Medicine, The University of Nottingham, Nottingham, UK
| | - Ian Ellis
- Breast Pathology Research Group, Nottingham Breast Cancer Research Centre, Biodiscovery Institute, Faculty of Medicine, The University of Nottingham, Nottingham, UK
| | - Adam Elzagheid
- Genetic Engineering Department, Libyan Biotechnology Research Centre, Tripoli, Libya
| | - Andrew Green
- Breast Pathology Research Group, Nottingham Breast Cancer Research Centre, Biodiscovery Institute, Faculty of Medicine, The University of Nottingham, Nottingham, UK.
| | - Caroline Chapman
- Eastern Bowel Cancer Screening Hub, Nottingham University Hospitals, NHS Trust, Nottingham, UK
| |
Collapse
|
6
|
Pathan SU, Kharwar A, Ibrahim MA, Singh SB, Bajaj P. Enzymes as indispensable markers in disease diagnosis. Bioanalysis 2024; 16:485-497. [PMID: 38530222 PMCID: PMC11216522 DOI: 10.4155/bio-2023-0207] [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: 10/10/2023] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
Enzymes have been used for disease diagnosis for many decades; however, advancements in technology like ELISA and flow cytometry-based detection have significantly increased their use and have increased the sensitivity of detection. Technological advancements in recombinant enzyme production have increased enzymatic stability, and the use of colorimetric-based and florescence-based assays has led to their increased use as biomarkers for disease detection. Enzymes like acid phosphatase, cathepsin, lactate dehydrogenase, thymidine kinase and creatine kinase are indispensable markers for diagnosing cancer, cardiovascular diseases and others. This minireview summarizes various enzymes used in disease diagnosis, their metabolic role, market value and potential as disease markers across various metabolic and other disorders.
Collapse
Affiliation(s)
- Shehabaz Usman Pathan
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Akash Kharwar
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Madaje Amir Ibrahim
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Shashi Bala Singh
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Priyanka Bajaj
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| |
Collapse
|
7
|
Huang X, Deng H, Zhang B, Wang K, Qu Y, Li T, Liu T. The causal relationship between cathepsins and digestive system tumors: a Mendelian randomization study. Front Oncol 2024; 14:1365138. [PMID: 38590662 PMCID: PMC10999587 DOI: 10.3389/fonc.2024.1365138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/14/2024] [Indexed: 04/10/2024] Open
Abstract
Background Multiple studies have confirmed the significant role of cathepsins in the development and progression of digestive system tumors. However, further investigation is needed to determine the causal relationships. Methods We conducted a two-sample bidirectional Mendelian randomization (MR) study using pooled data from a genome-wide association study (GWAS) to assess the causal associations between nine cathepsins (cathepsin B, E, F, G, H, L2, O, S, and Z) and six types of digestive system tumors, including hepatocellular carcinoma (HCC), pancreatic cancer (PCa), biliary tract cancer (BTC), colorectal cancer (CRC), gastric carcinoma (GC), and esophageal cancer (EC). We employed the following methods including inverse variance weighting (IVW), MR-Egger, weighted median (WM), Cochran's Q, MR-PRESSO, MR-Egger intercept test and leave-one-out sensitivity analysis. The STROBE-MR checklist for the reporting of MR studies was used in this study. Results The risk of HCC increased with high levels of cathepsin G (IVW: p = 0.029, odds ratio (OR) = 1.369, 95% confidence interval (CI) = 1.033-1.814). Similarly, BTC was associated with elevated cathepsin B levels (IVW: p = 0.025, OR = 1.693, 95% CI = 1.070-2.681). Conversely, a reduction in PCa risk was associated with increased cathepsin H levels (IVW: p = 0.027, OR = 0.896, 95% CI = 0.812-0.988). Lastly, high levels of cathepsin L2 were found to lower the risk of CRC (IVW: p = 0.034, OR = 0.814, 95% CI = 0.674-0.985). Conclusion Our findings confirm the causal relationship between cathepsins and digestive system tumors, which can offer valuable insights for the diagnosis and treatment of digestive system tumors.
Collapse
Affiliation(s)
- Xupeng Huang
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Houbo Deng
- Department of Hepatology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Bo Zhang
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Kuisong Wang
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yi Qu
- Graduate School, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Ting Li
- Department of Hepatology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Tiejun Liu
- Department of Hepatology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
8
|
Lee SG, Woo SM, Seo SU, Lee CH, Baek MC, Jang SH, Park ZY, Yook S, Nam JO, Kwon TK. Cathepsin D promotes polarization of tumor-associated macrophages and metastasis through TGFBI-CCL20 signaling. Exp Mol Med 2024; 56:383-394. [PMID: 38297161 PMCID: PMC10907383 DOI: 10.1038/s12276-024-01163-9] [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: 02/11/2023] [Revised: 10/23/2023] [Accepted: 11/23/2023] [Indexed: 02/02/2024] Open
Abstract
M2-like tumor-associated macrophages (TAMs) are risk factors for cancer progression and metastasis. However, the mechanisms underlying their polarization are still not fully understood. Although cathepsin D (Cat D) has been reported as a procarcinogenic factor, little is known about the functional role of Cat D in the tumor microenvironment (TME). This study aimed to explore the effect and molecular mechanisms of Cat D in the TME. Cat D knockout (KO) altered the cytokine secretion pattern and induced TAM reprogramming from the M2 to M1 subtype, thereby preventing epithelial-mesenchymal transition and tumor metastasis. Mechanistically, we identified transforming growth factor beta-induced protein (TGFBI) as a Cat D target protein that is specifically associated with TAM polarization. Elevated TGFBI expression in Cat D KO cancer cells resulted in a decline in M2-like TAM polarization. Our RNA-sequencing results indicated that the cancer cell-secreted chemokine CCL20 is a major secretory chemokine for Cat D-TGFBI-mediated TAM polarization. In contrast, Cat D overexpression accelerated TAM polarization into M2-like cells by suppressing TGFBI expression. In addition, the double Cat D and TGFBI KO rescued the inhibitory effects of Cat D KO on tumor metastasis by controlling TAM and T-cell activation. These findings indicated that Cat D contributes to cancer metastasis through TGFBI-mediated TAM reprogramming. Cat D deletion inhibits M2-like TAM polarization through TGFBI-mediated CCL20 expression, reprogramming the immunosuppressive TME. Our results open a potential new avenue for therapy focused on eliminating tumor metastasis.
Collapse
Affiliation(s)
- Seul Gi Lee
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Seon Min Woo
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Seung Un Seo
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Chan-Hyeong Lee
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Se Hwan Jang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Zee Yong Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ju-Ock Nam
- Department of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea.
- Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, Republic of Korea.
| |
Collapse
|
9
|
Desroys du Roure P, Lajoie L, Mallavialle A, Alcaraz LB, Mansouri H, Fenou L, Garambois V, Rubio L, David T, Coenon L, Boissière-Michot F, Chateau MC, Ngo G, Jarlier M, Villalba M, Martineau P, Laurent-Matha V, Roger P, Guiu S, Chardès T, Gros L, Liaudet-Coopman E. A novel Fc-engineered cathepsin D-targeting antibody enhances ADCC, triggers tumor-infiltrating NK cell recruitment, and improves treatment with paclitaxel and enzalutamide in triple-negative breast cancer. J Immunother Cancer 2024; 12:e007135. [PMID: 38290768 PMCID: PMC10828871 DOI: 10.1136/jitc-2023-007135] [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] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) prognosis is poor. Immunotherapies to enhance the antibody-induced natural killer (NK) cell antitumor activity are emerging for TNBC that is frequently immunogenic. The aspartic protease cathepsin D (cath-D), a tumor cell-associated extracellular protein with protumor activity and a poor prognosis marker in TNBC, is a prime target for antibody-based therapy to induce NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC). This study investigated whether Fc-engineered anti-cath-D antibodies trigger ADCC, their impact on antitumor efficacy and tumor-infiltrating NK cells, and their relevance for combinatory therapy in TNBC. METHODS Cath-D expression and localization in TNBC samples were evaluated by western blotting, immunofluorescence, and immunohistochemistry. The binding of human anti-cath-D F1M1 and Fc-engineered antibody variants, which enhance (F1M1-Fc+) or prevent (F1M1-Fc-) affinity for CD16a, to secreted human and murine cath-D was analyzed by ELISA, and to CD16a by surface plasmon resonance and flow cytometry. NK cell activation was investigated by flow cytometry, and ADCC by lactate dehydrogenase release. The antitumor efficacy of F1M1 Fc-variants was investigated using TNBC cell xenografts in nude mice. NK cell recruitment, activation, and cytotoxic activity were analyzed in MDA-MB-231 cell xenografts by immunophenotyping and RT-qPCR. NK cells were depleted using an anti-asialo GM1 antibody. F1M1-Fc+ antitumor effect was assessed in TNBC patient-derived xenografts (PDXs) and TNBC SUM159 cell xenografts, and in combination with paclitaxel or enzalutamide. RESULTS Cath-D expression on the TNBC cell surface could be exploited to induce ADCC. F1M1 Fc-variants recognized human and mouse cath-D. F1M1-Fc+ activated NK cells in vitro and induced ADCC against TNBC cells and cancer-associated fibroblasts more efficiently than F1M1. F1M1-Fc- was ineffective. In the MDA-MB-231 cell xenograft model, F1M1-Fc+ displayed higher antitumor activity than F1M1, whereas F1M1-Fc- was less effective, reflecting the importance of Fc-dependent mechanisms in vivo. F1M1-Fc+ triggered tumor-infiltrating NK cell recruitment, activation and cytotoxic activity in MDA-MB-231 cell xenografts. NK cell depletion impaired F1M1-Fc+ antitumor activity, demonstrating their key role. F1M1-Fc+ inhibited growth of SUM159 cell xenografts and two TNBC PDXs. In combination therapy, F1M1-Fc+ improved paclitaxel and enzalutamide therapeutic efficacy without toxicity. CONCLUSIONS F1M1-Fc+ is a promising immunotherapy for TNBC that could be combined with conventional regimens, including chemotherapy or antiandrogens.
Collapse
Affiliation(s)
| | - Laurie Lajoie
- Université de Tours - INRAE, UMR1282, Infectiologie et Santé Publique (ISP), équipe BioMédicaments Anti-Parasitaires (BioMAP), Tours, France
| | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Lindsay B Alcaraz
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Hanane Mansouri
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- RHEM, IRCM, Montpellier, France
| | - Lise Fenou
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Lucie Rubio
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Loïs Coenon
- IRMB, University of Montpellier, INSERM, CNRS, CHU Montpellier, Montpellier, France
| | | | | | - Giang Ngo
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Martin Villalba
- IRMB, University of Montpellier, INSERM, CNRS, CHU Montpellier, Montpellier, France
- Institut du Cancer Avignon-Provence Sainte Catherine, Avignon, France
| | - Pierre Martineau
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nimes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre national de la recherche Scientifique, Paris, F-75016, France
| | - Laurent Gros
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre national de la recherche Scientifique, Paris, F-75016, France
| | | |
Collapse
|
10
|
Barman SK, Sen MK, Mahns DA, Wu MJ, Malladi CS. Molecular Insights into the Breast and Prostate Cancer Cells in Response to the Change of Extracellular Zinc. JOURNAL OF ONCOLOGY 2024; 2024:9925970. [PMID: 38249992 PMCID: PMC10798840 DOI: 10.1155/2024/9925970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024]
Abstract
Zinc dyshomeostasis is manifested in breast and prostate cancer cells. This study attempted to uncover the molecular details prodded by the change of extracellular zinc by employing a panel of normal and cancerous breast and prostate cell lines coupled with the top-down proteomics with two-dimensional gel electrophoresis followed by liquid chromatography-tandem mass spectrometry. The protein samples were generated from MCF-7 breast cancer cells, MCF10A normal breast cells, PC3 prostate cancer cells, and RWPE-1 normal prostate cells with or without exogenous zinc exposure in a time course (T0 and T120). By comparing the cancer cells vs respective normal epithelial cells without zinc treatment (T0), differentially expressed proteins (23 upregulated and 18 downregulated in MCF-7 cells; 14 upregulated and 30 downregulated in PC3 cells) were identified, which provides insights into the intrinsic differences of breast and prostate cancer cells. The dynamic protein landscapes in the cancer cells prodded by the extracellular zinc treatment reveal the potential roles of the identified zinc-responsive proteins (e.g., triosephosphate isomerase, S100A13, tumour proteins hD53 and hD54, and tumour suppressor prohibitin) in breast and prostate cancers. This study, for the first time, simultaneously investigated the two kinds of cancer cells related to zinc dyshomeostasis, and the findings shed light on the molecular understanding of the breast and prostate cancer cells in response to extracellular zinc variation.
Collapse
Affiliation(s)
- Shital K. Barman
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751, Australia
| | - Monokesh K. Sen
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown 2006, NSW, Australia
| | - David A. Mahns
- School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751, Australia
| | - Ming J. Wu
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751, Australia
| | - Chandra S. Malladi
- Proteomics and Lipidomics Lab, School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW 2751, Australia
| |
Collapse
|
11
|
David T, Mallavialle A, Faget J, Alcaraz LB, Lapierre M, du Roure PD, Laurent-Matha V, Mansouri H, Jarlier M, Martineau P, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Anti-cathepsin D immunotherapy triggers both innate and adaptive anti-tumour immunity in breast cancer. Br J Pharmacol 2023. [PMID: 38030588 DOI: 10.1111/bph.16291] [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: 06/28/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Triple-negative breast cancer (TNBC) has poorer outcomes than other breast cancers (BC), including HER2+ BC. Cathepsin D (CathD) is a poor prognosis marker overproduced by BC cells, hypersecreted in the tumour microenvironment with tumour-promoting activity. Here, we characterized the immunomodulatory activity of the anti-CathD antibody F1 and its improved Fab-aglycosylated version (F1M1) in immunocompetent mouse models of TNBC (C57BL/6 mice harbouring E0771 cell grafts) and HER2-amplified BC (BALB/c mice harbouring TUBO cell grafts). EXPERIMENTAL APPROACH CathD expression was evaluated by western blotting and immunofluorescence, and antibody binding to CathD by ELISA. Antibody anti-tumour efficacy was investigated in mouse models. Immune cell recruitment and activation were assessed by immunohistochemistry, immunophenotyping, and RT-qPCR. KEY RESULTS F1 and F1M1 antibodies remodelled the tumour immune landscape. Both antibodies promoted innate antitumour immunity by preventing the recruitment of immunosuppressive M2-polarized tumour-associated macrophages (TAMs) and by activating natural killer cells in the tumour microenvironment of both models. This translated into a reduction of T-cell exhaustion markers in the tumour microenvironment that could be locally supported by enhanced activation of anti-tumour antigen-presenting cell (M1-polarized TAMs and cDC1 cells) functions. Both antibodies inhibited tumour growth in the highly-immunogenic E0771 model, but only marginally in the immune-excluded TUBO model, indicating that anti-CathD immunotherapy is more relevant for BC with a high immune cell infiltrate, as often observed in TNBC. CONCLUSION AND IMPLICATION Anti-CathD antibody-based therapy triggers the anti-tumour innate and adaptive immunity in preclinical models of BC and is a promising immunotherapy for immunogenic TNBC.
Collapse
Affiliation(s)
- Timothée David
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | - Julien Faget
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | - Marion Lapierre
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | | | - Hanane Mansouri
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- RHEM, IRCM, Montpellier, France
| | | | | | - Pascal Roger
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Centre national de la recherche Scientifique, CNRS, Paris, France
| | | |
Collapse
|
12
|
Singh S, Maurya AK, Meena A, Mishra N, Luqman S. Myricitrin from bayberry as a potential inhibitor of cathepsin-D: Prospects for squamous lung carcinoma prevention. Food Chem Toxicol 2023; 179:113988. [PMID: 37586679 DOI: 10.1016/j.fct.2023.113988] [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: 04/18/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
Cathepsin-D (CATD) inhibitors' design and development drawn interest due to their potential therapeutic applications in managing different cancer types, including lung cancer. This study investigated myricitrin, a flavonol-3-O-rhamnoside, for its binding affinity to CATD. Molecular docking experiments revealed a strong binding affinity (-7.8 kcal/mol). Molecular dynamics (MD) simulation confirmed the complex's stability, while enzyme activity studies showed inhibitory concentration (IC50) of 35.14 ± 6.08 μM (in cell-free) and 16.00 ± 3.48 μM (in cell-based) test systems. Expression analysis indicated downregulation of CATD with a fold change of 1.35. Myricitrin demonstrated antiproliferative effects on NCIH-520 cells [IC50: 64.11 μM in Sulphorhodamine B (SRB), 24.44 μM in 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], but did not affect healthy CHANG cells. It also prolonged the G2/M phase (at 10 μM: 1.19-fold; at 100 μM: 1.13-fold) and increased sub-diploid population by 1.35-fold. Based on the analysis done using SwissADME program, it is predicted that myricitrin is not a cytochrome p450s (CYPs) inhibitor, followed the rule of Ghose and found not permeable to the blood-brain barrier (BBB) which suggests it as a safe molecule. In summary, the experimental findings may establish the foundation for myricitrin and its analogues to be used therapeutically in CATD-mediated lung cancer prevention.
Collapse
Affiliation(s)
- Shilpi Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, Uttar Pradesh, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Akhilesh Kumar Maurya
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, Uttar Pradesh, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, Uttar Pradesh, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Nidhi Mishra
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, Uttar Pradesh, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, Uttar Pradesh, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
13
|
Jeong HW, Chang DS, Kim JS, Hwang YS. Role of cathepsin D induced by Porphyromonas gingivalis lipopolysaccharide in periodontitis. Eur J Oral Sci 2023; 131:e12923. [PMID: 36788303 DOI: 10.1111/eos.12923] [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: 08/11/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023]
Abstract
Periodontitis is an inflammatory disease of tooth-supporting tissues caused by oral bacteria. Periodontal ligament loss and alveolar bone destruction occur in progressive periodontitis. Since gingival crevicular fluids (GCF) reflects the inflammatory environment of the periodontal pocket, it is a very important specimen for developing targets for periodontitis diagnosis. An antibody array was performed using GCF collected from healthy participants and patients with periodontitis to identify the proteolytic enzymes involved in periodontitis. Of 21 targets on the antibody array membrane, kallikrein 6 (KLK6), kallikrein 10 (KLK10), cathepsin A (CathA), and cathepsin D (CathD) showed higher levels in periodontitis GCF than in GCF from healthy participants. Lipopolysaccharide stimulation of Porphyromonas gingivalis (PG-LPS) in immortalized gingival fibroblasts only increased CathD protein levels among the four targets. The substrate cleavage activity of CathD was increased in PG-LPS-treated immortalized gingival fibroblast extract. The PG-LPS-induced substrate cleavage effect was abolished by the CathD inhibitor pepstatin A. Osteoclast formation was promoted by treatment with conditioned media from PG-LPS- treated immortalized gingival fibroblasts but inhibited by the CathD inhibitor pepstatin A. These results suggest that PG-LPS affected the osteoclast formation process by increasing CathD expression in cells around the alveolar bone, thereby participating in periodontitis progression.
Collapse
Affiliation(s)
- Hyun Woong Jeong
- Department of Dental Hygiene, College of Health, Science, Eulji University, Seongnam, Korea
| | - Dong Sik Chang
- Department of Otorhinolaryngology, Eulji University Hospital, Eulji University, Daejeon, Korea
| | | | - Young Sun Hwang
- Department of Dental Hygiene, College of Health, Science, Eulji University, Seongnam, Korea
| |
Collapse
|
14
|
All Roads Lead to Cathepsins: The Role of Cathepsins in Non-Alcoholic Steatohepatitis-Induced Hepatocellular Carcinoma. Biomedicines 2022; 10:biomedicines10102351. [PMID: 36289617 PMCID: PMC9598942 DOI: 10.3390/biomedicines10102351] [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: 08/16/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins are lysosomal proteases that are essential to maintain cellular physiological homeostasis and are involved in multiple processes, such as immune and energy regulation. Predominantly, cathepsins reside in the lysosomal compartment; however, they can also be secreted by cells and enter the extracellular space. Extracellular cathepsins have been linked to several pathologies, including non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). NASH is an increasingly important risk factor for the development of HCC, which is the third leading cause of cancer-related deaths and poses a great medical and economic burden. While information regarding the involvement of cathepsins in NASH-induced HCC (NASH-HCC) is limited, data to support the role of cathepsins in either NASH or HCC is accumulating. Since cathepsins play a role in both NASH and HCC, it is likely that the role of cathepsins is more significant in NASH-HCC compared to HCC derived from other etiologies. In the current review, we provide an overview on the available data regarding cathepsins in NASH and HCC, argue that cathepsins play a key role in the transition from NASH to HCC, and shed light on therapeutic options in this context.
Collapse
|
15
|
Berg AL, Rowson-Hodel A, Wheeler MR, Hu M, Free SR, Carraway KL. Engaging the Lysosome and Lysosome-Dependent Cell Death in Cancer. Breast Cancer 2022. [DOI: 10.36255/exon-publications-breast-cancer-lysosome] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
A Comparison of Various Chips Used for the Manufacture of Biosensors Applied in Non-Fluidic Array SPRi, Based on the Example of Determination of Cathepsin D. BIOSENSORS 2021; 12:bios12010021. [PMID: 35049649 PMCID: PMC8773720 DOI: 10.3390/bios12010021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 01/01/2023]
Abstract
Non-fluidic array SPR imaging (SPRi) with appropriate biosensors is a new tool for the determination of various biomarkers in body fluids. Numerous biomarkers can be determined without signal enhancement or preliminarily preconcentration. The introduction of a new material solution of the chip may increase the scope of the application of this technique. Solutions with adhesive separating foil and an Ag/Au chip were compared with the previously used two-paint separating polymer and pure gold chip. These solutions were tested using the example of a biosensor for cathepsin D (Cath D), which consisted of pepstatin A (a Cath D inhibitor) immobilized via a cysteamine linker using the NHS/EDC protocol. Four material versions of the Cath D biosensor proved adequate in terms of range of linearity, LOQ, precision and recovery. All four versions of the biosensor were used for the determination of Cath D in the blood serum patients with glioblastoma and control samples, producing very similar results and showing an elevated biomarker concentration in the case of cancer. Therefore, the problem of determining the correct level of Cath D in the serum of healthy individuals has been resolved, correcting literature data which ranged over three orders of magnitude.
Collapse
|
17
|
CSNK1G2 differently sensitizes tamoxifen-induced decrease in PI3K/AKT/mTOR/S6K and ERK signaling according to the estrogen receptor existence in breast cancer cells. PLoS One 2021; 16:e0246264. [PMID: 33861751 PMCID: PMC8051802 DOI: 10.1371/journal.pone.0246264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/17/2021] [Indexed: 11/19/2022] Open
Abstract
Tamoxifen (TAM) is a selective estrogen receptor modulator used for breast cancer patients. Prolonged use of tamoxifen is not recommended for some patients. In this study, we aimed to identify molecular targets sensitive to TAM using a genome-wide gene deletion library screening of fission yeast heterozygous mutants. From the screening, casein kinase 1 gamma 2 (CSNK1G2), a serine-/threonine protein kinase, was the most sensitive target to TAM with a significant cytotoxicity in estrogen receptor-positive (ER+) breast cancer cells but with only a slight toxicity in the case of ER- cells. In addition, tumor sphere formation and expression of breast stem cell marker genes such as CD44/CD2 were greatly inhibited by CSNK1G2 knockdown in ER+ breast cancer cells. Consistently, CSNK1G2 altered ERα activity via phosphorylation, specifically at serine (Ser)167, as well as the regulation of estrogen-responsive element (ERE) of estrogen-responsive genes such as CTSD and GREB1. However, ERα silencing almost completely blocked CSNK1G2-induced TAM sensitivity. In ER+ breast cancer cells, combined treatment with TAM and CSNK1G2 knockdown further enhanced the TAM-mediated decrease in phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR)/ribosomal protein S6 kinase (S6K) signaling but not extracellular signal-regulated kinase (ERK) signaling. Inversely, in ER- cells treated with TAM, only ERK and PI3K signaling was altered by CSNK1G2 knockdown. The CK1 inhibitor, D4476, partly mimicked the CSNK1G2 knockdown effect in ER+ breast cancer cells, but with a broader repression ranging from PI3K/AKT/mTOR/S6K to ERK signaling. Collectively, these results suggest that CSNK1G2 plays a key role in sensitizing TAM toxicity in ER+ and ER- breast cancer cells via differently regulating PI3K/AKT/mTOR/S6K and ERK signaling.
Collapse
|
18
|
Alcaraz LB, Mallavialle A, David T, Derocq D, Delolme F, Dieryckx C, Mollevi C, Boissière-Michot F, Simony-Lafontaine J, Du Manoir S, Huesgen PF, Overall CM, Tartare-Deckert S, Jacot W, Chardès T, Guiu S, Roger P, Reinheckel T, Moali C, Liaudet-Coopman E. A 9-kDa matricellular SPARC fragment released by cathepsin D exhibits pro-tumor activity in the triple-negative breast cancer microenvironment. Am J Cancer Res 2021; 11:6173-6192. [PMID: 33995652 PMCID: PMC8120228 DOI: 10.7150/thno.58254] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/20/2021] [Indexed: 12/26/2022] Open
Abstract
Rationale: Alternative therapeutic strategies based on tumor-specific molecular targets are urgently needed for triple-negative breast cancer (TNBC). The protease cathepsin D (cath-D) is a marker of poor prognosis in TNBC and a tumor-specific extracellular target for antibody-based therapy. The identification of cath-D substrates is crucial for the mechanistic understanding of its role in the TNBC microenvironment and future therapeutic developments. Methods: The cath-D substrate repertoire was investigated by N-Terminal Amine Isotopic Labeling of Substrates (TAILS)-based degradome analysis in a co-culture assay of TNBC cells and breast fibroblasts. Substrates were validated by amino-terminal oriented mass spectrometry of substrates (ATOMS). Cath-D and SPARC expression in TNBC was examined using an online transcriptomic survival analysis, tissue micro-arrays, TNBC cell lines, patient-derived xenografts (PDX), human TNBC samples, and mammary tumors from MMTV-PyMT Ctsd-/-knock-out mice. The biological role of SPARC and its fragments in TNBC were studied using immunohistochemistry and immunofluorescence analysis, gene expression knockdown, co-culture assays, western blot analysis, RT-quantitative PCR, adhesion assays, Transwell motility, trans-endothelial migration and invasion assays. Results: TAILS analysis showed that the matricellular protein SPARC is a substrate of extracellular cath-D. In vitro, cath-D induced limited proteolysis of SPARC C-terminal extracellular Ca2+ binding domain at acidic pH, leading to the production of SPARC fragments (34-, 27-, 16-, 9-, and 6-kDa). Similarly, cath-D secreted by TNBC cells cleaved fibroblast- and cancer cell-derived SPARC at the tumor pericellular acidic pH. SPARC cleavage also occurred in TNBC tumors. Among these fragments, only the 9-kDa SPARC fragment inhibited TNBC cell adhesion and spreading on fibronectin, and stimulated their migration, endothelial transmigration, and invasion. Conclusions: Our study establishes a novel crosstalk between proteases and matricellular proteins in the tumor microenvironment through limited SPARC proteolysis, revealing a novel targetable 9-kDa bioactive SPARC fragment for new TNBC treatments. Our study will pave the way for the development of strategies for targeting bioactive fragments from matricellular proteins in TNBC.
Collapse
|
19
|
Benchmark of site- and structure-specific quantitative tissue N-glycoproteomics for discovery of potential N-glycoprotein markers: a case study of pancreatic cancer. Glycoconj J 2021; 38:213-231. [PMID: 33835347 DOI: 10.1007/s10719-021-09994-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is a highly malignant tumor of the digestive tract that is difficult to diagnose and treat. It is more common in developed countries and has become one of the main causes of death in some countries and regions. Currently, pancreatic cancer generally has a poor prognosis, partly due to the lack of symptoms in the early stages of pancreatic cancer. Therefore, most cases are diagnosed at advanced stage. With the continuous in-depth research of glycoproteomics in precision medical diagnosis, there have been some reports on quantitative analysis of cancer-related cells, plasma or tissues to find specific biomarkers for targeted therapy. This research is based on the developed complete N-linked glycopeptide database search engine GPSeeker, combined with liquid-mass spectrometry and stable diethyl isotope labeling, providing a benchmark of site- and structure-specific quantitative tissue N-glycoproteomics for discovery of potential N-glycoprotein markers. With spectrum-level FDR ≤1%, 20,038 intact N-Glycopeptides corresponding to 4518 peptide backbones, 228 N-glycan monosaccharide compositions 1026 N-glycan putative structures, 4460 N-glycosites and 3437 intact N-glycoproteins were identified. With the criteria of ≥1.5-fold change and p value<0.05, 52 differentially expressed intact N-glycopeptides (DEGPs) were found in pancreatic cancer tussues relative to control, where 38 up-regulated and 14 down-regulated, respectively.
Collapse
|
20
|
The Role of Lysosomes in the Cancer Progression: Focus on the Extracellular Matrix Degradation. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2020-5.6.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|