Introduction. Pancreatic cancer (PC), and in particular the ductal adenocarcinoma, has still a poor response to available therapies. A hallmark of this malignancy is the pronounced fibrotic stroma, composed by tumor-associated fibroblasts (TAF), immune cells, cytokines and growth factors stored in the extracellular matrix (ECM), with a key role in PC progression and drug resistance (Korc et al., 2007). For these reasons, efforts have been focused on the development of novel therapies able to target both malignant cells and TAF (Sun et al., 2018). Since years, we have developed an anticancer gene therapy based on human adipose mesenchymal stromal/stem cells (AD-MSC) secreting the pro-apoptotic soluble (s)-TRAIL to mostly target PC malignant cells (Spano et al., 2019; Rossignoli et al., 2019). Here, by in vitro and in vivo models, we investigated the potential synergistic impact of a novel combinatory approach between chemotherapeutic Gemcitabine (Gem) and AD-MSC sTRAIL on both PC tumor and TAF. Moreover, to initially verify the safety of this strategy, we assessed the impact of AD-MSC sTRAIL on normal tissues and in particular against white blood cells. Material and methods. In vitro assay on PC cell lines: sensitivity of PC cell lines (BxPC-3, MIA PaCa-2 and PA-TU-8988T) to sTRAIL, alone or in combination with Gem, was evaluated by CellTiter Glo assay in 2D. PC in vivo-like microenvironment was reproduced loading PC cell lines in 3D bioreactor and cell death was quantified after Gem & AD-MSC sTRAIL combinatory treatment. In vivo animal model: PC orthotopic xenotransplant murine models were developed by implantation of PC cell lines in the pancreas of NOD/SCID mice. Synergistic effect of the combinatory approach on tumor growth was verified by Ultrasound imaging. Paraffin embedded tumor sections were evaluated by H&E staining and Cytokeratin 7 and 8/18 IHC. Cytotoxicity study on PC TAF: primary TAF isolated by mechanical and enzymatic digestion were tested for sensitivity to sTRAIL, alone or in combination with Gem, by CellTiter Glo. RNA-Seq on PC microenvironment: TAF were co-cultured with a PC cell line, subsequently sorted by FACS and analysed by RNA-Seq. Immunotoxicity study: viability and proliferative rate of immune cells (monocytes, neutrophils and T lymphocytes) isolated from peripheral blood of healthy donors and treated with sTRAIL or co-cultured with AD-MSC sTRAIL were evaluated by metabolic assays and FACS. Results. PC cell lines showed different sensitivity levels to Gem or sTRAIL alone. Gem & sTRAIL combinatory approach revealed a synergistic cytotoxic effect on PC cells both in 2D and 3D systems. Synergy between Gem and AD-MSC sTRAIL was confirmed in the orthotopic PC murine models, showing a significant tumor tissue degeneration compared to control and Gem alone. Primary PC TAF were refractory to the sTRAIL cytotoxic effect as single agent, however pre-treatment with Gem was able to restore sTRAIL sensitivity. Investigating tumor-stroma crosstalk, gene expression profiling revealed a significant increase in expression of genes related to ECM, such as MMP2, POSTN, COL1A1 and COL12A1, in PC tumor cells after co-culture with TAF. Finally, immunological safety study confirmed that white blood cells were refractory to the pro-apoptotic effect displayed by sTRAIL and the direct cell-to-cell contact with AD-MSC sTRAIL had negligible impact on monocyte and T cell viability. Similarly, proliferation capability of T cells was not significantly affected by sTRAIL alone, as well as by co-culture with AD-MSC sTRAIL. Conclusions. These data demonstrated the safety and therapeutic potential of combining a gene therapy approach with chemotherapy to target both the tumor and stromal compartment in pancreatic ductal adenocarcinoma.

Introduzione. Il tumore del pancreas (PC), ed in particolare l’adenocarcinoma duttale, mostra ancora una scarsa risposta alle terapie disponibili. Un elemento peculiare è l’abbondante stroma fibrotico, composto da fibroblasti associati al tumore (TAF), cellule immunitarie, citochine e fattori di crescita immersi nella matrice extracellulare (ECM), con un ruolo chiave nella progressione e farmacoresistenza del PC (Korc et al., 2007). Sforzi significativi si sono così focalizzati sullo sviluppo di terapie capaci di bersagliare sia cellule tumorali che TAF (Sun et al., 2018). Da anni, stiamo sviluppando una terapia genica antitumorale a base di cellule stromali/staminali mesenchimali da tessuto adiposo (AD-MSC) secernenti la molecola pro-apoptotica solubile TRAIL (sTRAIL) per bersagliare cellule tumorali di PC (Spano et al., 2019; Rossignoli et al., 2019). Qui, abbiamo investigato in vitro ed in vivo l’impatto sinergico di un approccio combinatorio tra il chemioterapico Gemcitabina (Gem) e le AD-MSC sTRAIL sia su cellule tumorali che su TAF di PC. Inoltre, per verificare la sicurezza di tale terapia, abbiamo valutato l’impatto citotossico delle AD-MSC sTRAIL su tessuti sani ed in particolare sui leucociti. Materiali e metodi. Saggi in vitro su linee di PC: la sensibilità di linee cellulari di PC (BxPC-3, MIA PaCa-2 e PA-TU-8988T) a sTRAIL, da solo o in combinazione con Gem, è stata valutata con CellTiter Glo in 2D. Un microambiente di PC in vivo-like è stato riprodotto caricando linee di PC in un bioreattore 3D e la mortalità cellulare è stata quantificata dopo trattamento con Gem & AD-MSC sTRAIL. Modelli animali in vivo: modelli murini ortotopici di PC sono stati sviluppati mediante impianto di linee di PC nel pancreas di topi NOD/SCID. L’effetto sinergico dell’approccio combinatorio sulla crescita tumorale è stato verificato mediante ecografia. Sezioni di tumore in paraffina sono state valutate con H&E e IHC per citocheratina 7 e 8/18. Citotossicità su TAF di PC: TAF primari sono stati isolati e testati per la sensibilità a sTRAIL, da solo o in combinazione con Gem, tramite CellTiter Glo. RNA-Seq sul microambiente di PC: i TAF sono stati co-coltivati con una linea di PC, sortati mediante FACS e analizzati con RNA-Seq. Immunotossicità: vitalità e proliferazione di cellule immunitarie (monociti, neutrofili e linfociti T) da sangue periferico di donatori sani trattate con sTRAIL o co-coltivate con AD-MSC sTRAIL sono state valutate con saggi metabolici e FACS. Risultati. Le linee cellulari di PC mostrano differenti livelli di sensibilità a Gem o sTRAIL da soli e la combinazione Gem & sTRAIL ha rivelato un effetto sinergico sulle cellule di PC in 2D ed in 3D. La sinergia tra Gem e AD-MSC sTRAIL è stata confermata nei modelli murini ortotopici di PC con una significativa degenerazione del tessuto tumorale rispetto al controllo e alla sola Gem. I TAF primari di PC sono resistenti all’effetto citotossico di sTRAIL da solo, mentre il pre-trattamento con Gem ne ripristina la sensibilità. Studiando l’interazione stroma-tumore, l’RNA-Seq ha rivelato un aumento significativo dell’espressione di geni correlati all’ECM, come MMP2, POSTN, COL1A1 e COL12A1, nel tumore dopo co-coltura con i TAF. Infine, studi sulla sicurezza immunologica hanno confermato che i leucociti sono refrattari all’effetto di sTRAIL e il contatto diretto con le AD-MSC sTRAIL ha un impatto trascurabile sulla vitalità di monociti e cellule T. Allo stesso modo, la proliferazione delle cellule T non è influenzata sia da sTRAIL che dalle AD-MSC sTRAIL. Conclusioni. Questi dati hanno dimostrato la sicurezza ed il potenziale terapeutico di combinare terapia genica e chemioterapia per bersagliare sia la componente tumorale che stromale dell’adenocarcinoma pancreatico duttale.

Efficacia e Sicurezza di Modelli Pre-clinici Combinatori di Chemioterapia e Terapia Genica per Bersagliare il Tumore del Pancreas ed il suo Microambiente / Giulia Casari , 2022 May 23. 34. ciclo, Anno Accademico 2020/2021.

Efficacia e Sicurezza di Modelli Pre-clinici Combinatori di Chemioterapia e Terapia Genica per Bersagliare il Tumore del Pancreas ed il suo Microambiente

CASARI, GIULIA
2022

Abstract

Introduction. Pancreatic cancer (PC), and in particular the ductal adenocarcinoma, has still a poor response to available therapies. A hallmark of this malignancy is the pronounced fibrotic stroma, composed by tumor-associated fibroblasts (TAF), immune cells, cytokines and growth factors stored in the extracellular matrix (ECM), with a key role in PC progression and drug resistance (Korc et al., 2007). For these reasons, efforts have been focused on the development of novel therapies able to target both malignant cells and TAF (Sun et al., 2018). Since years, we have developed an anticancer gene therapy based on human adipose mesenchymal stromal/stem cells (AD-MSC) secreting the pro-apoptotic soluble (s)-TRAIL to mostly target PC malignant cells (Spano et al., 2019; Rossignoli et al., 2019). Here, by in vitro and in vivo models, we investigated the potential synergistic impact of a novel combinatory approach between chemotherapeutic Gemcitabine (Gem) and AD-MSC sTRAIL on both PC tumor and TAF. Moreover, to initially verify the safety of this strategy, we assessed the impact of AD-MSC sTRAIL on normal tissues and in particular against white blood cells. Material and methods. In vitro assay on PC cell lines: sensitivity of PC cell lines (BxPC-3, MIA PaCa-2 and PA-TU-8988T) to sTRAIL, alone or in combination with Gem, was evaluated by CellTiter Glo assay in 2D. PC in vivo-like microenvironment was reproduced loading PC cell lines in 3D bioreactor and cell death was quantified after Gem & AD-MSC sTRAIL combinatory treatment. In vivo animal model: PC orthotopic xenotransplant murine models were developed by implantation of PC cell lines in the pancreas of NOD/SCID mice. Synergistic effect of the combinatory approach on tumor growth was verified by Ultrasound imaging. Paraffin embedded tumor sections were evaluated by H&E staining and Cytokeratin 7 and 8/18 IHC. Cytotoxicity study on PC TAF: primary TAF isolated by mechanical and enzymatic digestion were tested for sensitivity to sTRAIL, alone or in combination with Gem, by CellTiter Glo. RNA-Seq on PC microenvironment: TAF were co-cultured with a PC cell line, subsequently sorted by FACS and analysed by RNA-Seq. Immunotoxicity study: viability and proliferative rate of immune cells (monocytes, neutrophils and T lymphocytes) isolated from peripheral blood of healthy donors and treated with sTRAIL or co-cultured with AD-MSC sTRAIL were evaluated by metabolic assays and FACS. Results. PC cell lines showed different sensitivity levels to Gem or sTRAIL alone. Gem & sTRAIL combinatory approach revealed a synergistic cytotoxic effect on PC cells both in 2D and 3D systems. Synergy between Gem and AD-MSC sTRAIL was confirmed in the orthotopic PC murine models, showing a significant tumor tissue degeneration compared to control and Gem alone. Primary PC TAF were refractory to the sTRAIL cytotoxic effect as single agent, however pre-treatment with Gem was able to restore sTRAIL sensitivity. Investigating tumor-stroma crosstalk, gene expression profiling revealed a significant increase in expression of genes related to ECM, such as MMP2, POSTN, COL1A1 and COL12A1, in PC tumor cells after co-culture with TAF. Finally, immunological safety study confirmed that white blood cells were refractory to the pro-apoptotic effect displayed by sTRAIL and the direct cell-to-cell contact with AD-MSC sTRAIL had negligible impact on monocyte and T cell viability. Similarly, proliferation capability of T cells was not significantly affected by sTRAIL alone, as well as by co-culture with AD-MSC sTRAIL. Conclusions. These data demonstrated the safety and therapeutic potential of combining a gene therapy approach with chemotherapy to target both the tumor and stromal compartment in pancreatic ductal adenocarcinoma.
Efficacy and Safety of Chemo and Gene Therapy Combinatory Pre-clinical Models Targeting Pancreatic Cancer and its Microenvironment
23-mag-2022
DOMINICI, Massimo
File in questo prodotto:
File Dimensione Formato  
PHD THESIS_GIULIA CASARI.pdf

embargo fino al 22/05/2025

Descrizione: Tesi definitiva Casari Giulia
Tipologia: Tesi di dottorato
Dimensione 3.52 MB
Formato Adobe PDF
3.52 MB Adobe PDF   Visualizza/Apri   Richiedi una copia
Pubblicazioni consigliate

Licenza Creative Commons
I metadati presenti in IRIS UNIMORE sono rilasciati con licenza Creative Commons CC0 1.0 Universal, mentre i file delle pubblicazioni sono rilasciati con licenza Attribuzione 4.0 Internazionale (CC BY 4.0), salvo diversa indicazione.
In caso di violazione di copyright, contattare Supporto Iris

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11380/1278859
Citazioni
  • ???jsp.display-item.citation.pmc??? ND
  • Scopus ND
  • ???jsp.display-item.citation.isi??? ND
social impact