1
|
Catalano M, Rebuzzi SE, Maruzzo M, De Giorgi U, Buti S, Galli L, Fornarini G, Zucali PA, Claps M, Chiellino S, Zampiva I, Pipitone S, Ricotta R, Sorarù M, Mollica V, Tudini M, Fratino L, Prati V, Caffo O, Atzori F, Morelli F, Prati G, Nolè F, Vignani F, Cavo A, Di Napoli M, Malgeri A, Naglieri E, Signori A, Banna GL, Rescigno P, Cerbone L, Antonuzzo L, Roviello G. Sodium levels and immunotherapy efficacy in mRCC patients with bone metastases: sub analysis of Meet-Uro 15 study. Front Immunol 2024; 15:1361010. [PMID: 39034992 PMCID: PMC11257879 DOI: 10.3389/fimmu.2024.1361010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Background Immune-checkpoint inhibitors (ICIs) have significantly improved metastatic renal cell carcinoma (mRCC) prognosis, although their efficacy in patients with bone metastases (BMs) remains poorly understood. We investigated the prognostic role of natremia in pretreated RCC patients with BMs receiving immunotherapy. Materials and methods This retrospective multicenter study included RCC patients with BMs receiving nivolumab as second-line therapy or beyond. Inclusion criteria involved baseline sodium levels (pre-ICI) and sodium levels after 4 weeks of nivolumab initiation (post-ICI). The population was divided into two groups based on the median value, and response rates, progression-free survival (PFS), and overall survival (OS) were assessed. Results Among 120 eligible patients, those with pre-treatment sodium levels ≥140 mEq/L showed longer OS (18.7 vs. 12.0 months, p=0.04). Pre-treatment sodium levels ≥140 mEq/L were associated with better OS compared to levels <140 mE/L (18.7 vs. 12.0, p=0.04). Post-treatment sodium levels ≥140 mEq/L were associated with improved PFS (9.6 vs. 3.2 months) and OS (25.1 vs. 8.8 months) (p=0.05 and p<0.01, respectively). Patients with consistent sodium levels ≥140 mEq/L at both time points exhibited the best outcomes compared to those with lower values (PFS 11.5 vs. 3.3 months and OS 42.2 vs. 9.0 months, respectively, p<0.01). Disease control rate was significantly higher in the latter group (p<0.01). Multivariate analysis confirmed the prognostic significance of sodium levels. Conclusion Elevated sodium levels (≥140 mEq/L) pre- and post-ICI treatment correlate with better survival outcomes in mRCC patients with BMs. This finding suggests sodium level assessment as a potential prognostic factor in these patients and warrants further investigation, particularly in combination immunotherapy settings.
Collapse
Affiliation(s)
- Martina Catalano
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze, Italy
| | - Sara Elena Rebuzzi
- Medical Oncology Unit, Ospedale San Paolo, Savona, Italy
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
| | - Marco Maruzzo
- Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto (IOV) - Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Padova, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sebastiano Buti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Luca Galli
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino of Genova, Genova, Italy
| | - Paolo Andrea Zucali
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Oncology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Melanie Claps
- SS Oncologia Medica Genitourinaria, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Silvia Chiellino
- Medical Oncology Unit, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ilaria Zampiva
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy
| | - Stefania Pipitone
- Medical Oncology Unit, Department of Oncology and Hemathology, University Hospital of Modena, Modena, Italy
| | - Riccardo Ricotta
- Oncology Unit, IRCCS MultiMedica, Sesto san Giovanni, Milan, Italy
| | - Mariella Sorarù
- UOC Oncologia, AULSS 6 Euganea, Ospedale di Camposampiero, Padova, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Lucia Fratino
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano CRO-IRCCS, Aviano, Italy
| | - Veronica Prati
- Oncology Unit, Michele e Pietro Ferrero Hospital, Azienda Sanitaria Locale (ASL) CN 2, Verduno, Italy
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | - Francesco Atzori
- SSD Oncologia Medica, Azienda Sanitaria Locale (ASL) Sulcis, Cagliari, Italy
| | - Franco Morelli
- Medical Oncology Department, Casa Sollievo Della Sofferenza Hospital, IRCCS, San Giovanni Rotondo, Italy
| | - Giuseppe Prati
- Department of Oncology and Advanced Technologies AUSL - IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Franco Nolè
- Medical Oncology Division of Urogenital & Head & Neck Tumors, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesca Vignani
- Division of Medical Oncology, Mauriziano Hospital, Turin, Piemont, Italy
| | - Alessia Cavo
- Oncology Unit, Villa Scassi Hospital, Genova, Italy
| | - Marilena Di Napoli
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Andrea Malgeri
- Department of Medical Oncology, Fondazione Policlinico Campus Bio-Medico, Roma, Italy
| | - Emanuele Naglieri
- Division of Medical Oncology, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Alessio Signori
- Department of Health Sciences, Section of Biostatistics, University of Genova, Genoa, Italy
| | - Giuseppe Luigi Banna
- Faculty of Science and Health, School of Pharmacy and Biomedical Sciences, Portsmouth Hospitals University NHS Trust, University of Portsmouth, Portsmouth, United Kingdom
| | | | - Linda Cerbone
- Department of Medical Oncology, San Camillo Forlanini Hospital, Rome, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze, Italy
| |
Collapse
|
2
|
Catalano M, Lavacchi D, Giommoni E, Shabani S, Guidolin A, Brugia M, Petrioli R, Ramello M, Pillozzi S, Antonuzzo L, Roviello G. Prognostic role of sodium levels in colorectal cancer patients receiving aflibercept plus FOLFIRI. Future Oncol 2023; 19:2537-2546. [PMID: 38050741 DOI: 10.2217/fon-2023-0610] [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] [Indexed: 12/06/2023] Open
Abstract
Aim: To investigate the impact of natremia in metastatic colorectal cancer (mCRC) patients treated with aflibercept plus folinic acid, 5-fluorouracil, oxaliplatin and irinotecan (FOLFIRI). Patients & methods: A total of 84 mCRC patients receiving aflibercept plus FOLFIRI as second-line treatment were enrolled and divided into two groups based on their median sodium value. Progression-free survival and overall survival were analyzed. Results: Patients with sodium levels ≥140 mEq/l had significantly longer median progression-free survival (4.1 vs 2 months; p < 0.01) and median overall survival (12 vs 7.3 months; p < 0.01) compared with those with lower levels. Conclusion: This study suggests that higher pretreatment serum sodium levels are associated with improved outcomes in mCRC patients receiving aflibercept and FOLFIRI, potentially serving as a prognostic marker to aid treatment management.
Collapse
Affiliation(s)
- Martina Catalano
- Department of Health Science, University of Florence, Florence, 50134, Italy
| | - Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50134, Italy
| | - Elisa Giommoni
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50134, Italy
| | - Sonia Shabani
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50134, Italy
| | - Alessia Guidolin
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50134, Italy
| | - Marco Brugia
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50134, Italy
| | - Roberto Petrioli
- Oncology Unit, Azienda Ospedaliera Universitaria Senese, Siena, 53100, Italy
| | - Monica Ramello
- Oncology Unit, Department of Medical, Surgical & Health Sciences, University of Trieste, Trieste, 34129, Italy
| | - Serena Pillozzi
- Department of Experimental & Clinical Medicine, University of Florence, Florence, 34129, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, 50134, Italy
- Department of Experimental & Clinical Medicine, University of Florence, Florence, 34129, Italy
| | | |
Collapse
|
3
|
Pukkanasut P, Whitt J, Guenter R, Lynch SE, Gallegos C, Rosendo-Pineda MJ, Gomora JC, Chen H, Lin D, Sorace A, Jaskula-Sztul R, Velu SE. Voltage-Gated Sodium Channel Na V1.7 Inhibitors with Potent Anticancer Activities in Medullary Thyroid Cancer Cells. Cancers (Basel) 2023; 15:2806. [PMID: 37345144 PMCID: PMC10216335 DOI: 10.3390/cancers15102806] [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: 03/13/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Our results from quantitative RT-PCR, Western blotting, immunohistochemistry, and the tissue microarray of medullary thyroid cancer (MTC) cell lines and patient specimens confirm that VGSC subtype NaV1.7 is uniquely expressed in aggressive MTC and not expressed in normal thyroid cells and tissues. We establish the druggability of NaV1.7 in MTC by identifying a novel inhibitor (SV188) and investigate its mode of binding and ability to inhibit INa current in NaV1.7. The whole-cell patch-clamp studies of the SV188 in the NaV1.7 channels expressed in HEK-293 cells show that SV188 inhibited the INa current in NaV1.7 with an IC50 value of 3.6 µM by a voltage- and use-dependent blockade mechanism, and the maximum inhibitory effect is observed when the channel is open. SV188 inhibited the viability of MTC cell lines, MZ-CRC-1 and TT, with IC50 values of 8.47 μM and 9.32 μM, respectively, and significantly inhibited the invasion of MZ-CRC-1 cells by 35% and 52% at 3 μM and 6 μM, respectively. In contrast, SV188 had no effect on the invasion of TT cells derived from primary tumor, which have lower basal expression of NaV1.7. In addition, SV188 at 3 μM significantly inhibited the migration of MZ-CRC-1 and TT cells by 27% and 57%, respectively.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Jason Whitt
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
| | - Rachael Guenter
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
| | - Shannon E. Lynch
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
| | - Carlos Gallegos
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Margarita Jacaranda Rosendo-Pineda
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.J.R.-P.); (J.C.G.)
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.J.R.-P.); (J.C.G.)
| | - Herbert Chen
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Diana Lin
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Anna Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
4
|
He J, Xu J, Chang Z, Yan J, Zhang L, Qin Y. NALCN is a potential biomarker and therapeutic target in human cancers. Front Genet 2023; 14:1164707. [PMID: 37152978 PMCID: PMC10154523 DOI: 10.3389/fgene.2023.1164707] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Background: Sodium leak channel non-selective (NALCN), known as a voltage-independent Na+ channel, is increasingly considered to play vital roles in tumorigenesis and metastasis of human cancers. However, no comprehensive pan-cancer analysis of NALCN has been conducted. Our study aims to explore the potential diagnostic, prognostic and therapeutic value of NALCN in human cancers. Methods: Through comprehensive application of datasets from Human Protein Atlas (HPA), The Cancer Genome Atlas (TCGA), Cancer Cell Line Encyclopedia (CCLE), Enhanced Version of Tumor Immune Estimation Resource (TIMER2.0), Tumor and Immune System Interaction Database (TISIDB), The University of Alabama at Birmingham Cancer data analysis Portal (UALCAN), cBioPortal, GeneMANIA and Search Tool for the Retrieval of Interaction Gene/Proteins (STRING) databases, we explored the potential roles of NALCN in different cancers. The differential expression, prognostic implications, pathological stages and grades, molecular and immune subtypes, diagnostic accuracy, tumor mutation burden (TMB), microsatellite instability (MSI), mismatch repair (MMR) genes, immune checkpoint genes, chemokine genes, major histocompatibility complex (MHC)-related genes, tumor-infiltrating immune cells (TIICs), promoter methylation, mutations, copy number alteration (CNA), and functional enrichment related to NALCN were analyzed. Results: Most cancers lowly expressed NALCN. Upregulated NALCN expression was associated with poor or better prognosis in different cancers. Moreover, NALCN was correlated with clinicopathological features in multiple cancers. NALCN showed high diagnostic accuracy in 5 caner types. NALCN is highly linked with immune-related biomarkers, immune-related genes and TIICs. Significant methylation changes and genetic alteration of NALCN can be observed in many cancers. Enrichment analysis showed that NALCN is closely related to multiple tumor-related signaling pathways. Conclusion: Our study revealed the vital involvement of NALCN in cancer. NALCN can be used as a prognostic biomarker for immune infiltration and clinical outcomes, and has potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Jian He
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Zhiwei Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaqin Yan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Limin Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yanru Qin,
| |
Collapse
|
5
|
Conrad C, Conway J, Polacheck WJ, Rizvi I, Scarcelli G. Water transport regulates nucleus volume, cell density, Young's modulus, and E-cadherin expression in tumor spheroids. Eur J Cell Biol 2022; 101:151278. [PMID: 36306595 DOI: 10.1016/j.ejcb.2022.151278] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 12/14/2022] Open
Abstract
Cell volume is maintained by the balance of water and solutes across the cell membrane and plays an important role in mechanics and biochemical signaling in cells. Here, we assess the relationship between cell volume, mechanical properties, and E-cadherin expression in three-dimensional cultures for ovarian cancer. To determine the effect of water transport in multi-cellular tumors, ovarian cancer spheroids were subjected to hypotonic and hypertonic shock using water and sucrose mixtures, respectively. Increased osmolality resulted in decreased nucleus volume, increased Young's modulus, and increased tumor cell density in ovarian cancer spheroids. Next, we looked at the reversibility of mechanics and morphology after 5 min of osmotic shock and found that spheroids had a robust ability to return to their original state. Finally, we quantified the size of E-cadherin clusters at cell-cell junctions and observed a significant increase in aggregate size following 30 min of hypertonic and hypotonic osmotic shocks. Yet, these effects were not apparent after 5 min of osmotic shock, illustrating a temporal difference between E-cadherin regulation and the immediate mechanical and morphology changes. Still, the osmotically induced E-cadherin aggregates which formed at the 30-minute timepoint was reversible when spheroids were replenished with isotonic medium. Altogether, this work demonstrated an important role of osmolality in transforming mechanical, morphology, and molecular states.
Collapse
Affiliation(s)
- Christina Conrad
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Jessica Conway
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
| |
Collapse
|
6
|
Huang Q, Li X, Sun J, Zhou Y. Tumor-derived endomucin promotes colorectal cancer proliferation and metastasis. Cancer Med 2022; 12:3222-3236. [PMID: 35971319 PMCID: PMC9939191 DOI: 10.1002/cam4.5055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 05/31/2022] [Accepted: 07/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Endomucin (EMCN) is a type I transmembrane glycoprotein and a mucin-like component of the endothelial cell glycocalyx. The mechanism of EMCN action in colorectal cancer (CRC) remains unclear. AIMS Our aim was to explore the role of EMCN in the progression of CRC. MATERIALS & METHODS We examined EMCN expression in CRC tissues and normal para-carcinoma tissues. The function and mechanisms of EMCN were checked in CRC cell lines and in mouse xenograft. Additionally, we used co-immunoprecipitation and mass spectrometry to identify the potential EMCN-binding proteins. Functional annotation analysis showed where these genes were enriched. RESULTS We found that EMCN was overexpressed in tumor tissues compared with that in normal para-carcinoma tissues. We also found that overexpression of EMCN induced CRC proliferation and metastasis both in vitro and in vivo. EMCN knockdown prevents epithelial-mesenchymal transition in vitro. We identified 178 potential EMCN-binding partners. Furthermore, functional annotation analysis indicated that these genes were considerably enriched in carcinogenic-related functions and pathways. Collectively, the identification of EMCN-binding partners enhanced our understanding of the mechanism of EMCN-mediated malignant phenotypes, and this research may provide valuable insights into the molecular mechanisms underlying CRC. CONCLUSION Tumor-derived endomucin promotes colorectal cancer proliferation and metastasis. We identified 178 EMCN-binding proteins and initially screened three potential EMCN-interacting proteins: NALCN, and TPM2, ANKK1. Our study provides valuable insights into the molecular mechanisms underlying CRC development.
Collapse
Affiliation(s)
- Qi Huang
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaMianyangSichuanPR China
| | - Xue‐mei Li
- The First Affiliated Hospital of Chengdu Medical College, Clinical Medical CollegeChengdu Medical CollegeChengduSichuanChina
| | - Jing‐ping Sun
- The First Affiliated Hospital of Chengdu Medical College, Clinical Medical CollegeChengdu Medical CollegeChengduSichuanChina
| | - Yan Zhou
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaMianyangSichuanPR China
| |
Collapse
|