44 results on '"Roberta Sommaggio"'
Search Results
2. 424 Adoptive cell therapy with cytokine-induces killer cells armed with immunotools against Her-2 expressing breast cancer
- Author
-
Gaia Griguolo, Anna Dalla Pietà, Elisa Cappuzzello, Annavera Ventura, Roberta Sommaggio, Antonio Rosato, Hieu Trong Ngo, Emilia Vigolo, Giulia D’Accardio, Sara Perpinello, and Matthias Peipp
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. How can Cytokine-induced killer cells overcome CAR-T cell limits
- Author
-
Elisa Cappuzzello, Emilia Vigolo, Giulia D’Accardio, Giuseppe Astori, Antonio Rosato, and Roberta Sommaggio
- Subjects
CIK (cytokine-induced killer) cells ,CAR (chimeric antigen receptor) T cells ,ATMP ,adoptive cell immunotherapy ,hematological malignancies ,solid tumors ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The successful treatment of patients affected by B-cell malignancies with Chimeric Antigen Receptor (CAR)-T cells represented a breakthrough in the field of adoptive cell therapy (ACT). However, CAR-T therapy is not an option for every patient, and several needs remain unmet. In particular, the production of CAR-T cells is expensive, labor-intensive and logistically challenging; additionally, the toxicities deriving from CAR-T cells infusion, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been documented extensively. Alternative cellular therapy products such as Cytokine-induced killer (CIK) cells have the potential to overcome some of these obstacles. CIK cells are a heterogeneous population of polyclonal CD3+CD56+ T cells with phenotypic and functional properties of NK cells. CIK cell cytotoxicity is exerted in a major histocompatibility complex (MHC)-unrestricted manner through the engagement of natural killer group 2 member D (NKG2D) molecules, against a wide range of hematological and solid tumors without the need for prior antigen exposure or priming. The foremost potential of CIK cells lies in the very limited ability to induce graft-versus-host disease (GvHD) reactions in the allogeneic setting. CIK cells are produced with a simple and extremely efficient expansion protocol, which leads to a massive expansion of effector cells and requires a lower financial commitment compared to CAR-T cells. Indeed, CAR-T manufacturing involves the engineering with expensive GMP-grade viral vectors in centralized manufacturing facilities, whereas CIK cell production is successfully performed in local academic GMP facilities, and CIK cell treatment is now licensed in many countries. Moreover, the toxicities observed for CAR-T cells are not present in CIK cell-treated patients, thus further reducing the costs associated with hospitalization and post-infusion monitoring of patients, and ultimately encouraging the delivery of cell therapies in the outpatient setting. This review aims to give an overview of the limitations of CAR-T cell therapy and outline how the use of CIK cells could overcome such drawbacks thanks to their unique features. We highlight the undeniable advantages of using CIK cells as a therapeutic product, underlying the opportunity for further research on the topic.
- Published
- 2023
- Full Text
- View/download PDF
4. Mutant p53 induces Golgi tubulo-vesiculation driving a prometastatic secretome
- Author
-
Valeria Capaci, Lorenzo Bascetta, Marco Fantuz, Galina V. Beznoussenko, Roberta Sommaggio, Valeria Cancila, Andrea Bisso, Elena Campaner, Alexander A. Mironov, Jacek R. Wiśniewski, Luisa Ulloa Severino, Denis Scaini, Fleur Bossi, Jodi Lees, Noa Alon, Ledia Brunga, David Malkin, Silvano Piazza, Licio Collavin, Antonio Rosato, Silvio Bicciato, Claudio Tripodo, Fiamma Mantovani, and Giannino Del Sal
- Subjects
Science - Abstract
p53 mutants can promote tumorigenesis by affecting fundamental cellular pathways and functions. In this study, the authors demonstrate a novel mutant-p53/HIF1α/miR-30d axis that impacts Golgi structure, trafficking, and secretion of proteins essential for tumor growth and metastasis.
- Published
- 2020
- Full Text
- View/download PDF
5. Innovative therapeutic strategy for B-cell malignancies that combines obinutuzumab and cytokine-induced killer cells
- Author
-
Elisa Cappuzzello, Pierangela Palmerini, Annavera Ventura, Andrea Visentin, Giuseppe Astori, Katia Chieregato, Valentina Mozzo, Omar Perbellini, Maria Chiara Tisi, Livio Trentin, Carlo Visco, Marco Ruggeri, Roberta Sommaggio, Antonio Rosato, and Anna Dalla Pietà
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Patients affected by aggressive B-cell malignancies who are resistant to primary or salvage chemoimmunotherapy have an extremely poor prognosis and limited therapeutic options. Promising therapeutic success has been achieved with the infusion of CD19 chimeric antigen receptor-T cells, but several limits still restrain the administration to a limited proportion of patients. This unmet clinical need might be fulfilled by an adoptive immunotherapy approach that combines cytokine-induced killer (CIK) cells and monoclonal antibodies (mAb) to the CD20 antigen. Indeed, CIK cells are an effector population endowed with antitumor activity, which can be further improved and antigen-specifically redirected by clinical-grade mAb triggering antibody-dependent cell-mediated cytotoxicity.Methods CIK cells were generated from peripheral blood of patients affected by different B-cell malignancies using a blinatumomab-based cell culture protocol. Effector cells were combined with the anti-CD20 mAb obinutuzumab and their therapeutic activity was assessed both in vitro and in vivo.Results CIK cells were successfully expanded in clinically relevant numbers, starting from small volumes of peripheral blood with extremely low CD3+ counts and high tumor burden. This relied on the addition of blinatumumab in culture, which leads to the simultaneous expansion of effector cells and the complete elimination of the neoplastic component. Moreover, CIK cells were highly cytotoxic in vitro against both B-cell tumor cell lines and autologous neoplastic targets, and had a significant therapeutic efficacy against a B-cell malignancy patient-derived xenograft on in vivo transfer.Conclusions The combination of an easily expandable CIK cell effector population with a mAb already in clinical use establishes a tumor antigen-specific redirection strategy that can be rapidly translated into clinical practice, providing an effective therapeutic alternative for B-cell malignancies without any need for genetic modifications. Additionally, the approach can be potentially applied to an extremely vast array of different tumors by simply substituting the targeting mAb.
- Published
- 2021
- Full Text
- View/download PDF
6. Adoptive cell therapy of triple negative breast cancer with redirected cytokine-induced killer cells
- Author
-
Roberta Sommaggio, Elisa Cappuzzello, Anna Dalla Pietà, Anna Tosi, Pierangela Palmerini, Debora Carpanese, Lorenzo Nicolè, and Antonio Rosato
- Subjects
cytokine-induced killer cells (cik) ,adoptive cells therapy (act) ,triple negative breast cancer (tnbc) ,tnbc mouse models ,cetuximab ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Cytokine-Induced Killer (CIK) cells share several functional and phenotypical properties of both T and natural killer (NK) cells. They represent an attractive approach for cell-based immunotherapy, as they do not require antigen-specific priming for tumor cell recognition, and can be rapidly expanded in vitro. Their relevant expression of FcγRIIIa (CD16a) can be exploited in combination with clinical-grade monoclonal antibodies (mAbs) to redirect their lytic activity in an antigen-specific manner. Here, we report the efficacy of this combined approach against triple negative breast cancer (TNBC), an aggressive tumor that still requires therapeutic options. Different primitive and metastatic TNBC cancer mouse models were established in NSG mice, either by implanting patient-derived TNBC samples or injecting MDA-MB-231 cells orthotopically or intravenously. The combined treatment consisted in the repeated intratumoral or intravenous injection of CIK cells and cetuximab. Tumor growth and metastasis were monitored by bioluminescence or immunohistochemistry, and survival was recorded. CIK cells plus cetuximab significantly restrained primitive tumor growth in mice, either in patient-derived tumor xenografts or MDA-MB-231 cell line models. Moreover, this approach almost completely abolished metastasis spreading and dramatically improved survival. The antigen-specific mAb favored tumor and metastasis tissue infiltration by CIK cells, and led to an enrichment of the CD16a+ subset. Data highlight the potentiality of this novel immunotherapy strategy where a nonspecific cytotoxic cell population can be converted into tumor-specific effectors with clinical-grade antibodies, thus providing not only a therapeutic option for TNBC but also a valid alternative to more complex approaches based on chimeric antigen receptor-engineered cells. List of abbreviations ACT, Adoptive Cell Transfer; ADCC, Antibody-Dependent Cell-mediated Cytotoxicity; ADP, Adenosine diphosphate; BLI, Bioluminescence Imaging; CAR, Chimeric Antigen Receptor; CIK, Cytokine Induced Killer cells; CTX, Cetuximab; DMEM, Dulbecco’s Modified Eagle Medium; EGFR, Human Epidermal Growth Factor 1; ER, Estrogen; FBS, Fetal Bovine Serum; FFPE, Formalin-Fixed Paraffin-Embedded; GMP, Good Manufacturing Practices; GVHD, Graft Versus Host Disease; HER2, Human Epidermal Growth Factor 2; HRP, Horseradish Peroxidase; IFN-γ, Interferon-γ; IHC, Immunohistochemistry; IL-2, Interleukin-2; ISO, Irrelevant antibody; i.t., intratumoral; i.v., intravenous, mAbs, Monoclonal Antibodies; mIHC, Multiplex Fluorescence Immunohistochemistry; MHC, Major Histocompatibility Complex; NK, Natural Killer; NKG2D, Natural-Killer group 2 member D; NSG, NOD/SCID common γ chain knockout; PARP, Poly ADP-ribose polymerase; PBMCs, Peripheral Blood Mononuclear Cells; PBS, Phosphate-buffered saline; PDX, Patient-derived xenograft; PR, Progesterone; rhIFN-γ, Recombinant Human Interferon-γ; RPMI, Roswell Park Memorial Institute; STR, Short tandem Repeat; TCR, T Cell Receptor; TNBC, Triple Negative Breast Cancer; TSA, Tyramide Signal Amplification
- Published
- 2020
- Full Text
- View/download PDF
7. The mitochondrial calcium uniporter regulates breast cancer progression via HIF‐1α
- Author
-
Anna Tosatto, Roberta Sommaggio, Carsten Kummerow, Robert B Bentham, Thomas S Blacker, Tunde Berecz, Michael R Duchen, Antonio Rosato, Ivan Bogeski, Gyorgy Szabadkai, Rosario Rizzuto, and Cristina Mammucari
- Subjects
breast cancer ,HIF‐1α ,metastasis ,mitochondrial Ca2+ uptake ,reactive oxygen species ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Triple‐negative breast cancer (TNBC) represents the most aggressive breast tumor subtype. However, the molecular determinants responsible for the metastatic TNBC phenotype are only partially understood. We here show that expression of the mitochondrial calcium uniporter (MCU), the selective channel responsible for mitochondrial Ca2+ uptake, correlates with tumor size and lymph node infiltration, suggesting that mitochondrial Ca2+ uptake might be instrumental for tumor growth and metastatic formation. Accordingly, MCU downregulation hampered cell motility and invasiveness and reduced tumor growth, lymph node infiltration, and lung metastasis in TNBC xenografts. In MCU‐silenced cells, production of mitochondrial reactive oxygen species (mROS) is blunted and expression of the hypoxia‐inducible factor‐1α (HIF‐1α) is reduced, suggesting a signaling role for mROS and HIF‐1α, downstream of mitochondrial Ca2+. Finally, in breast cancer mRNA samples, a positive correlation of MCU expression with HIF‐1α signaling route is present. Our results indicate that MCU plays a central role in TNBC growth and metastasis formation and suggest that mitochondrial Ca2+ uptake is a potential novel therapeutic target for clinical intervention.
- Published
- 2016
- Full Text
- View/download PDF
8. Identification of a HLA-A*0201-restricted immunogenic epitope from the universal tumor antigen DEPDC1
- Author
-
Anna Tosi, Silvia Dalla Santa, Elisa Cappuzzello, Carolina Marotta, Dawid Walerich, Giannino Del Sal, Paola Zanovello, Roberta Sommaggio, and Antonio Rosato
- Subjects
cancer immunotherapy ,cytotoxic t lymphocytes ,depdc1 ,tumor antigen ,triple negative breast cancer ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The identification of universal tumor-specific antigens shared between multiple patients and/or multiple tumors is of great importance to overcome the practical limitations of personalized cancer immunotherapy. Recent studies support the involvement of DEPDC1 in many aspects of cancer traits, such as cell proliferation, resistance to induction of apoptosis and cell invasion, suggesting that it may play key roles in the oncogenic process. In this study, we report that DEPDC1 expression is upregulated in most types of human tumors, and closely linked to a poorer prognosis; therefore, it might be regarded as a novel universal oncoantigen potentially suitable for targeting many different cancers. In this regard, we report the identification of a HLA-A*0201 allele-restricted immunogenic DEPDC1-derived epitope, which is able to induce cytotoxic T lymphocytes (CTL) exerting a strong and specific functional response in vitro toward not only peptide-loaded cells but also triple negative breast cancer (TNBC) cells endogenously expressing the DEPDC1 protein. Such CTL are also therapeutically active against human TNBC xenografts in vivo upon adoptive transfer in immunodeficient mice. Overall, these data provide evidence that this DEPDC1-derived antigenic epitope can be exploited as a new tool for developing immunotherapeutic strategies for HLA-A*0201 patients with TNBC, and potentially many other cancers.
- Published
- 2017
- Full Text
- View/download PDF
9. A BARF1-specific mAb as a new immunotherapeutic tool for the management of EBV-related tumors
- Author
-
Riccardo Turrini, Anna Merlo, Debora Martorelli, Damiana Antonia Faè, Roberta Sommaggio, Isabella Monia Montagner, Vito Barbieri, Oriano Marin, Paola Zanovello, Riccardo Dolcetti, and Antonio Rosato
- Subjects
adcc ,barf1 ,cdc ,epstein–barr virus ,immunotherapy ,monoclonal antibody ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The use of monoclonal antibodies (mAb) for the diagnosis and treatment of malignancies is acquiring an increasing clinical importance, thanks to their specificity, efficacy and relative easiness of use. However, in the context of Epstein-Barr virus (EBV)-related malignancies, only cancers of B-cell origin can benefit from therapeutic mAb targeting specific B-cell lineage antigens. To overcome this limitation, we generated a new mAb specific for BARF1, an EBV-encoded protein with transforming and immune-modulating properties. BARF1 is expressed as a latent protein in nasopharyngeal (NPC) and gastric carcinoma (GC), and also in neoplastic B cells mainly upon lytic cycle induction, thus representing a potential target for all EBV-related malignancies. Considering that BARF1 is largely but not exclusively secreted, the BARF1 mAb was selected on the basis of its ability to bind a domain of the protein retained at the cell surface of tumor cells. In vitro, the newly generated mAb recognized the target molecule in its native conformation, and was highly effective in mediating both ADCC and CDC against BARF1-positive tumor cells. In vivo, biodistribution analysis in mice engrafted with BARF1-positive and -negative tumor cells confirmed its high specificity for the target. More importantly, the mAb disclosed a relevant antitumor potential in preclinical models of NPC and lymphoma, as evaluated in terms of both reduction of tumor masses and long-term survival. Taken together, these data not only confirm BARF1 as a promising target for immunotherapeutic interventions, but also pave the way for a successful translation of this new mAb to the clinical use.
- Published
- 2017
- Full Text
- View/download PDF
10. Prolyl‐isomerase Pin1 controls normal and cancer stem cells of the breast
- Author
-
Alessandra Rustighi, Alessandro Zannini, Luca Tiberi, Roberta Sommaggio, Silvano Piazza, Giovanni Sorrentino, Simona Nuzzo, Antonella Tuscano, Vincenzo Eterno, Federica Benvenuti, Libero Santarpia, Iannis Aifantis, Antonio Rosato, Silvio Bicciato, Alberto Zambelli, and Giannino Del Sal
- Subjects
breast cancer ,Fbxw7 E3 ubiquitin‐ligase ,Notch ,prolyl‐isomerase Pin1 ,stem cells ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Mammary epithelial stem cells are fundamental to maintain tissue integrity. Cancer stem cells (CSCs) are implicated in both treatment resistance and disease relapse, and the molecular bases of their malignant properties are still poorly understood. Here we show that both normal stem cells and CSCs of the breast are controlled by the prolyl‐isomerase Pin1. Mechanistically, following interaction with Pin1, Notch1 and Notch4, key regulators of cell fate, escape from proteasomal degradation by their major ubiquitin‐ligase Fbxw7α. Functionally, we show that Fbxw7α acts as an essential negative regulator of breast CSCs' expansion by restraining Notch activity, but the establishment of a Notch/Pin1 active circuitry opposes this effect, thus promoting breast CSCs self‐renewal, tumor growth and metastasis in vivo. In human breast cancers, despite Fbxw7α expression, high levels of Pin1 sustain Notch signaling, which correlates with poor prognosis. Suppression of Pin1 holds promise in reverting aggressive phenotypes, through CSC exhaustion as well as recovered drug sensitivity carrying relevant implications for therapy of breast cancers.
- Published
- 2013
- Full Text
- View/download PDF
11. TNF, Pig CD86, and VCAM-1 Identified as Potential Targets for Intervention in Xenotransplantation of Pig Chondrocytes
- Author
-
Roberta Sommaggio, Rafael Máñez, and Cristina Costa
- Subjects
Medicine - Abstract
Xenotransplantation of genetically engineered porcine chondrocytes may benefit many patients who suffer cartilage defects. In this work, we sought to elucidate the molecular bases of the cellular response to xenogeneic cartilage. To this end, we isolated pig costal chondrocytes (PCC) and conducted a series of functional studies. First, we determined by flow cytometry the cell surface expression of multiple immunoregulatory proteins in resting conditions or after treatment with human TNF-α, IL-1α, or IL-1β, which did not induce apoptosis. TNF-α and to a lesser extent IL-1α led to a marked upregulation of SLA I, VCAM-1, and ICAM-1 on PCC. SLA II and E-selectin remained undetectable in all the conditions assayed. Notably, CD86 was constitutively expressed at moderate levels, whereas CD80 and CD40 were barely detected. To assess their function, we next studied the interaction of PCC with human monoblastic U937 and Jurkat T cells. U937 cells adhered to resting and in a greater proportion to cytokine-stimulated PCC. Consistent with its expression pattern, pig VCAM-1 was key, mediating the increased adhesion after cytokine stimulation. We also conducted coculture experiments with U937 and PCC and measured the release of pig and human cytokines. Stimulated PCC secreted IL-6 and IL-8, whereas U937 secreted IL-8 in response to PCC. Finally, coculture of PCC with Jurkat in the presence of PHA led to a marked Jurkat activation as determined by the increase in IL-2 secretion. This process was dramatically reduced by blocking pig CD86. In summary, CD86 and VCAM-1 on pig chondrocytes may be important triggers of the xenogeneic cellular immune response. These molecules together with TNF could be considered potential targets for intervention in order to develop xenogeneic therapies for cartilage repair.
- Published
- 2009
- Full Text
- View/download PDF
12. Erratum To: Prolyl‐isomerase Pin1 controls normal and cancer stem cells of the breast
- Author
-
Alessandra Rustighi, Alessandro Zannini, Luca Tiberi, Roberta Sommaggio, Silvano Piazza, Giovanni Sorrentino, Simona Nuzzo, Antonella Tuscano, Vincenzo Eterno, Federica Benvenuti, Libero Santarpia, Iannis Aifantis, Antonio Rosato, Silvio Bicciato, Alberto Zambelli, and Giannino Del Sal
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2014
- Full Text
- View/download PDF
13. 377 Adoptive cell therapy with cytokine-induced killer cells retargeted with immunotools against HER-2 positive breast cancer
- Author
-
Roberta Sommaggio, Elisa Cappuzzello, Annavera Ventura, Sara Perpinello, Anna Dalla Pieta, Emilia Vigolo, Giulia D’accardio, Pierangela Palmerini, and Antonio Rosato
- Published
- 2022
- Full Text
- View/download PDF
14. P09.13 Optimization of a GMP-grade large-scale expansion protocol for cytokine-induced killer cells using gas-permeable static culture flasks
- Author
-
Carlo Visco, Annavera Ventura, Marco Ruggeri, Katia Chieregato, Omar Perbellini, Giuseppe Astori, Roberta Sommaggio, P Palmerini, Antonio Rosato, A Dalla Pietà, Maria Chiara Tisi, and Elisa Cappuzzello
- Subjects
Cell therapy ,Antigen ,Cytokine-induced killer cell ,medicine.diagnostic_test ,Chemistry ,Cell culture ,medicine ,Cytotoxic T cell ,NKG2D ,Molecular biology ,Ex vivo ,Flow cytometry - Abstract
Background Cytokine-Induced Killer (CIK) cells are ex vivo expanded T cells with NK cell phenotype. They express both CD3 and CD56 antigens, and exert a potent antitumor activity against a variety of tumors. Several clinical trials demonstrated the safety and the feasibility of CIK cell therapy, with very low side effects and minimal graft-versus-host toxicity. In this study, we developed a GMP-compliant protocol for robust large-scale expansion of CIK cells using G-Rex® gas-permeable static culture flasks. Materials and Methods CIK cells were obtained by stimulating healthy donor PBMCs with GMP-grade IFN-γ, IL-2 and CD3 mAbs, and were cultured in G-Rex6® or G-Rex®6M well plates. CIK cells in G-Rex6® were split only once at day 7 to reduce cell density, whereas the number of CIK cells culterd in G-Rex®6M was not adjusted. In both culture conditions, fresh IL-2 was provided every 3–4 days. We compared these two culture protocols with the culture in standard flasks. Phenotype was analyzed by flow cytometry and cytotoxicity was assessed against several tumor cell lines by calcein-release assay. Results CIK cells cultured in G-Rex6® well plates showed an outstanding cell expansion compared to G-Rex®6M well plates or standard culture flasks, with a 400-fold expansion and a mean of 109 total cells obtained per single well in 14 days, starting from just 2.5 × 106 cells per well. Moreover, the cultures in G-Rex6® were characterized by an higher percentage of CD3+CD56+ cells, as compared to G-Rex®6M or standard culture flasks. Cells cultured in all devices had a comparable expression of NKG2D, NKp30, NKp44, 2B4 receptors. Importantly, CIK cells expanded in G-Rex®6 were as cytotoxic as cells expanded in standard culture flasks. Conversely, CIK cells cultured in G-Rex®6M showed a remarkable reduction of cytotoxicity against tumor cell targets, thus suggesting that cell density during expansion could affect CIK cell activity. Conclusions We propose a GMP-compliant protocol for robust large-scale production of CIK cells. G-Rex® system allows to obtain large amounts of CIK cells highly enriched in the CD3+CD56+ subset and endowed with high cytotoxic activity; this can be accomplished with just a single cell culture split at day 7, which dramatically reduces the culture manipulation as compared to the standard culture flasks. Notably, this strategy can be further and easily scalable to produce CIK cells for clinical immunotherapy applications. Disclosure Information A. Ventura: None. P. Palmerini: None. A. Dalla Pieta: None. R. Sommaggio: None. G. Astori: None. K. Chieregato: None. M. Tisi: None. C. Visco: None. O. Perbellini: None. M. Ruggeri: None. E. Cappuzzello: None. A. Rosato: None.
- Published
- 2020
- Full Text
- View/download PDF
15. P09.01 Adoptive cell therapy of hematological malignancies using cytokine-induced killer cells retargeted with monoclonal antibodies
- Author
-
Katia Chieregato, Antonio Rosato, Omar Perbellini, Marco Ruggeri, A Dalla Pietà, Giuseppe Astori, Maria Chiara Tisi, Elisa Cappuzzello, Carlo Visco, Roberta Sommaggio, and P Palmerini
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,education.field_of_study ,Cytokine-induced killer cell ,business.industry ,medicine.drug_class ,Population ,CD16 ,Monoclonal antibody ,medicine.anatomical_structure ,Cancer research ,Medicine ,Cytotoxic T cell ,business ,education ,B cell ,CD8 - Abstract
Background Cytokine-Induced Killer (CIK) cells are a population of effector cells that represents a promising tool for adoptive cell therapy. They are easily expandable ex-vivo, safe, and exert cytotoxicity against a broad range of tumor histotypes.1 We recently reported that they have a relevant expression of FcγRIIIa (CD16a), which can be exploited in combination with clinical-grade monoclonal antibodies (mAbs) to redirect their cytotoxicity in an antigen-specific manner, to improve their antitumor activity.2 Indeed, the engagement of CD16a on CIK cells leads to a potent antibody-dependent cell-mediated cytotoxicity (ADCC) against ovarian cancer both in vitro and in vivo. Based on this observation, we investigated whether CIK cells can be specifically retargeted against B-cell malignancies by combination with anti-CD20 mAbs, namely Rituximab® (RTX) and Obinutuzumab® (OBI). Materials and Methods CIK cells were obtained from peripheral blood mononuclear cells of healthy donors, and stimulated in vitro with IFN-γ, CD3 mAb and IL-2 for 14 days; fresh IL-2 was provided every 3–4 days. CIK cell phenotype was analyzed by multicolor flow cytometry; cytotoxic activity was assessed by calcein AM-release assay against B-cell lines, primary samples and patient-derived xenografts (PDX) obtained from B-cell lymphoma patients after written informed consent. Results The combination with both RTX and OBI significantly increased specific CIK cells lysis against several CD20-expressing lymphoma B cell lines, primary tumors from B-cell lymphoma patients and an established PDX, compared to the combination with a control mAb (cetuximab, CTX). NK-depletion demonstrated that the mAb-mediated cytotoxicity is accountable to the CIK cells fraction within the bulk population since no difference in the lytic activity was detectd in the absence of NK cells. In addition, these results are further supported by in vivo preliminary experiments where the treatment with CIK cells in combination with OBI extensively reduced the growth of PDX and increased mice survival, compared to CIK cells or OBI administered alone. Conclusions Here we proved that CIK cells can be retargeted with clinical-grade mAbs against CD20-expressing lymphomas. These data indicate that the combination of CIK cells with mAbs can represent a novel approach for the treatment of haematological malignancies. References Franceschetti M, Pievani A, Borleri G, Vago L, Fleischhauer K, Golay J, et al. Cytokine-induced killer cells are terminally differentiated activated CD8 cytotoxic T-EMRA lymphocytes. Exp Hematol 2009;37:616–28. Cappuzzello E, Tosi A, Zanovello P, Sommaggio R, Rosato A. Retargeting cytokine-induced killer cell activity by CD16 engagement with clinical-grade antibodies. Oncoimmunology 2016 Aug;5(8):e1199311. The research leading to these results has received funding from Fondazione AIRC under IG 2018 - ID. 21354 project - P.I. Rosato Antonio Disclosure Information A. Dalla Pieta: None. E. Cappuzzello: None. P. Palmerini: None. R. Sommaggio: None. G. Astori: None. K. Chieregato: None. O. Perbellini: None. M. Tisi: None. C. Visco: None. M. Ruggeri: None. A. Rosato: None.
- Published
- 2020
- Full Text
- View/download PDF
16. P06.06 Adoptive cell therapy of triple negative breast cancer with redirected cytokine-induced killer cells
- Author
-
Antonio Rosato, A Dalla Pietà, Roberta Sommaggio, Elisa Cappuzzello, P Palmerini, Debora Carpanese, Anna Tosi, and Lorenzo Nicolè
- Subjects
education.field_of_study ,Cetuximab ,Cytokine-induced killer cell ,business.industry ,medicine.medical_treatment ,Population ,Immunotherapy ,medicine.disease ,Metastasis ,Cell therapy ,Antigen ,medicine ,Cancer research ,Cytotoxic T cell ,education ,business ,medicine.drug - Abstract
Background Cytokine-Induced Killer (CIK) cells share several functional and phenotypical properties of both T and natural killer (NK) cells, and represent an attractive approach for cell-based immunotherapy as they do not require antigen-specific priming for tumor cell recognition, and can be efficiently and rapidly expanded in vitro. We recently reported that CIK cells have a relevant expression of FcγRIIIa (CD16a), which can be exploited in combination with clinical-grade monoclonal antibodies (mAbs) to redirect their lytic activity in an antigen-specific manner. Here, we report the assessment and the efficacy of this combined approach against triple negative breast cancer (TNBC), an aggressive tumor that still requires reliable therapeutic options. Materials and methods Different primitive and metastatic TNBC cancer mouse models were established in NSG mice, either by implanting patient-derived TNBC samples or MDA-MB-231 cells subcutaneously or orthotopically into the mammary fat pad, or by injecting MDA-MB-231 cells intravenously. The combined treatment consisted in the repeated intratumoral or intravenous injection of CIK cells and cetuximab, while the mAb alone or CIK cells plus Rituximab served as control treatments. Tumor growth and metastasis were monitored by bioluminescence or immunohistochemistry, and survival was recorded. Results CIK cells plus cetuximab significantly restrained primitive tumor growth in mice, either implanted with TNBC patient-derived tumor xenografts or injected with MDA-MB-231 TNBC cell line. Moreover, in both experimental and spontaneous metastatic models the combined approach almost completely abolished metastasis spreading and dramatically improved survival. The antigen-specific mAb favored tumor and metastasis tissue infiltration by CIK cells, and in particular led to an enrichment of the CD16a+ subset. Conclusions Data highlight the potentiality of a novel immunotherapy approach where a non-specific cytotoxic cell population can be converted into tumor-specific effectors with clinical-grade antibodies, thus providing not only a therapeutic option for TNBC but also a valid alternative to more complex approaches based on chimeric antigen receptor-engineered cells. Disclosure Information R. Sommaggio: None. E. Cappuzzello: None. A. Dalla Pieta: None. P. Palmerini: None. A. Tosi: None. D. Carpanese: None. L. Nicole: None. A. Rosato: None.
- Published
- 2020
- Full Text
- View/download PDF
17. Cell-Based Assays for Modeling Xenogeneic Immune Responses
- Author
-
Kelly, Casós, Roberta, Sommaggio, Magdiel, Pérez-Cruz, and Cristina, Costa
- Subjects
Cytotoxicity, Immunologic ,Graft Rejection ,Swine ,T-Lymphocytes ,Transplantation, Heterologous ,Cell Culture Techniques ,Coculture Techniques ,Killer Cells, Natural ,Transplantation Immunology ,Antigens, Heterophile ,Animals ,Cytokines ,Heterografts ,Humans ,Biological Assay - Abstract
Research in xenotransplantation implies a high experimental complexity comprising molecular, cellular, and in vivo studies to investigate the mechanisms of xenograft immune rejection and functional failure, as well as the strategies to counteract them. After major advances associated with the identification of the carbohydrate xenoantigens and their elimination through genomic edition of the source pigs, the study of the cellular immune response against the xenograft is gaining particular attention. Xenogeneic cell-based assays that put together pig cells and human leukocytes such as monocytes, NK cells, and T cells are relevant to address this hurdle. Thus, we describe here coculture, co-stimulatory, and cytotoxicity assays for investigating the cellular and molecular mechanisms of xenograft rejection. These techniques allow elucidating the key pathways that take place during the xenogeneic immune response in a simplified setting. Treatment with either pro-inflammatory or anti-inflammatory cytokines can be used for studying the regulation of adhesion, co-stimulatory molecules, and receptors involved in triggering the immune response under various conditions. Furthermore, these assays can be used for the follow-up of the immune response of in vivo studies as well as for the development of tolerogenic approaches that promote xenograft survival.
- Published
- 2020
18. Mutant p53 induces Golgi tubulo-vesiculation driving a prometastatic secretome
- Author
-
Antonio Rosato, Noa Alon, Lorenzo Bascetta, Silvio Bicciato, Licio Collavin, Alexander A. Mironov, Giannino Del Sal, Fiamma Mantovani, Luisa Ulloa Severino, Fleur Bossi, Ledia Brunga, Andrea Bisso, Valeria Cancila, Denis Scaini, Galina V. Beznoussenko, Roberta Sommaggio, Jodi Lees, Marco Fantuz, Jacek R. Wiśniewski, Elena Campaner, Claudio Tripodo, Silvano Piazza, Valeria Capaci, David Malkin, Capaci V., Bascetta L., Fantuz M., Beznoussenko G.V., Sommaggio R., Cancila V., Bisso A., Campaner E., Mironov A.A., Wisniewski J.R., Ulloa Severino L., Scaini D., Bossi F., Lees J., Alon N., Brunga L., Malkin D., Piazza S., Collavin L., Rosato A., Bicciato S., Tripodo C., Mantovani F., Del Sal G., Capaci, V., Bascetta, L., Fantuz, M., Beznoussenko, G. V., Sommaggio, R., Cancila, V., Bisso, A., Campaner, E., Mironov, A. A., Wisniewski, J. R., Ulloa Severino, L., Scaini, D., Bossi, F., Lees, J., Alon, N., Brunga, L., Malkin, D., Piazza, S., Collavin, L., Rosato, A., Bicciato, S., Tripodo, C., Mantovani, F., and Del Sal, G.
- Subjects
0301 basic medicine ,Biopsy ,General Physics and Astronomy ,Golgi Apparatus ,Animals, Biopsy, Breast Neoplasms, Cell Line, Tumor, Cell Transformation, Neoplastic, Female, Fibroblasts, Gene Expression Regulation, Neoplastic, Golgi Apparatus, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Li-Fraumeni Syndrome, Mice, MicroRNAs, Microtubules, Mutation, Primary Cell Culture, Secretory Vesicles, Signal Transduction,Skin, Tumor Microenvironment, Tumor Suppressor Protein p53, Xenograft Model Antitumor Assays ,02 engineering and technology ,medicine.disease_cause ,Cell Transformation ,Microtubules ,Settore BIO/09 - Fisiologia ,Metastasis ,Li-Fraumeni Syndrome ,Mice ,Tumor Microenvironment ,Golgi ,secretory machinery ,Super-resolution microscopy ,Animals ,Breast Neoplasms ,Cell Line, Tumor ,Cell Transformation, Neoplastic ,Female ,Fibroblasts ,Gene Expression Regulation, Neoplastic ,Humans ,Hypoxia-Inducible Factor 1, alpha Subunit ,MicroRNAs ,Mutation ,Primary Cell Culture ,Secretory Vesicles ,Signal Transduction ,Skin ,Tumor Suppressor Protein p53 ,Xenograft Model Antitumor Assays ,lcsh:Science ,Multidisciplinary ,Tumor ,Chemistry ,mutant p53 ,Cell migration ,MicroRNA ,Secretomics ,021001 nanoscience & nanotechnology ,Cell biology ,symbols ,Fibroblast ,miR-30d ,Hypoxia-Inducible Factor 1 ,0210 nano-technology ,Breast Neoplasm ,Human ,Cancer microenvironment ,Stromal cell ,Secretory Vesicle ,Science ,Microtubule ,Golgi Apparatu ,Settore MED/08 - Anatomia Patologica ,alpha Subunit ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,symbols.namesake ,medicine ,Settore MED/05 - Patologia Clinica ,Secretion ,Tumor microenvironment ,Neoplastic ,Animal ,General Chemistry ,Oncogenes ,Golgi apparatus ,HDAC6 ,Microreview ,microenvironment ,030104 developmental biology ,Gene Expression Regulation ,lcsh:Q ,Carcinogenesis - Abstract
TP53 missense mutations leading to the expression of mutant p53 oncoproteins are frequent driver events during tumorigenesis. p53 mutants promote tumor growth, metastasis and chemoresistance by affecting fundamental cellular pathways and functions. Here, we demonstrate that p53 mutants modify structure and function of the Golgi apparatus, culminating in the increased release of a pro-malignant secretome by tumor cells and primary fibroblasts from patients with Li-Fraumeni cancer predisposition syndrome. Mechanistically, interacting with the hypoxia responsive factor HIF1α, mutant p53 induces the expression of miR-30d, which in turn causes tubulo-vesiculation of the Golgi apparatus, leading to enhanced vesicular trafficking and secretion. The mut-p53/HIF1α/miR-30d axis potentiates the release of soluble factors and the deposition and remodeling of the ECM, affecting mechano-signaling and stromal cells activation within the tumor microenvironment, thereby enhancing tumor growth and metastatic colonization., p53 mutants can promote tumorigenesis by affecting fundamental cellular pathways and functions. In this study, the authors demonstrate a novel mutant-p53/HIF1α/miR-30d axis that impacts Golgi structure, trafficking, and secretion of proteins essential for tumor growth and metastasis.
- Published
- 2020
19. Adoptive cell therapy of hematological malignancies using cytokine-induced killer cells retargeted with monoclonal antibodies
- Author
-
Antonio Rosato, Omar Perbellini, A Dalla Pietà, Maria Chiara Tisi, Elisa Cappuzzello, Giuseppe Astori, P Palmerini, Carlo Visco, Katia Chieregato, Roberta Sommaggio, and Marco Ruggeri
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,Transplantation ,education.field_of_study ,Cytokine-induced killer cell ,medicine.drug_class ,business.industry ,Immunology ,Population ,Cell Biology ,Monoclonal antibody ,Chimeric antigen receptor ,Cell therapy ,Oncology ,Cancer research ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,education ,business ,Cytotoxicity ,Genetics (clinical) - Abstract
Background & Aim Cytokine-Induced Killer (CIK) cells are a population of effector cells that represent a promising tool for adoptive cell therapy. From a technical and methodological point of view, they are easily expandable ex-vivo and safe, and demonstrated efficacy against hematological malignancies. CIK cells exert cytotoxicity against a broad range of tumor histotypes but not against normal tissues and hematopoietic precursors. We recently reported that they have a relevant expression of FcγRIIIa (CD16a), which can be exploited in combination with clinical-grade monoclonal antibodies (mAbs) to redirect their cytotoxicity in an antigen-specific manner, to improve their antitumor activity. Indeed, the engagement of CD16a on CIK cells leads to a potent antibody-dependent cell-mediated cytotoxicity (ADCC) against ovarian cancer both in vitro and in vivo. Based on this observation, we investigated whether CIK cells could be specifically retargeted against B-cell malignancies by combination with anti-CD20 mAbs, namely Rituximab® and Obinutuzumab®. Methods, Results & Conclusion CIK cells were obtained from peripheral blood mononucleated cells of healthy donors, and stimulated in vitro with IFN-γ, CD3 mAb (OKT3) and IL-2 for 14 days; fresh IL-2 was provided every 3-4 days. CIK cell phenotype was analyzed by multicolor flow cytometry; cytotoxic activity was assessed by calcein AM-release assay against EHEB (CLL), Raji (Burkitt lymphoma), RCK-8, TMB-8, Karpas 422 (DLBCL) tumor cell lines, and primary samples obtained from CLL and DLBCL patients after written informed consent. The combination with either the mAbs significantly increased CIK cell cytotoxicity not only against lymphoma cell lines, but also against the primary tumor samples. Depletion of NK cells from CIK cell bulk cultures did not affect target cell lysis, thus confirming that the antibody-mediated increase of cytotoxicity was mainly accountable to the CIK cell fraction. Taken together, these data indicate that the combination treatment with CIK cells and anti-CD20 mAbs could represent a novel approach for the treatment of hematological malignancies, alternative to the use of chimeric antigen receptor (CAR)-T cells.
- Published
- 2020
- Full Text
- View/download PDF
20. Proteasome machinery is instrumental in a common gain-of-function program of the p53 missense mutants in cancer
- Author
-
Katarzyna Rajkowska, Silvano Piazza, Giannino Del Sal, Antonio Rosato, Roberta Sommaggio, Bruno Amati, Angela Amato, Kamil Lisek, Emiliano Dalla, Alberto Zambelli, Vincenzo Eterno, Katarzyna Gaweda-Walerych, Marco J. Morelli, Claudia Tonelli, Eleonora Ingallina, Dawid Walerych, Jacek R. Wiśniewski, Yari Ciani, Walerych, D., Lisek, K., Sommaggio, R., Piazza, S., Ciani, Y., Dalla, E., Rajkowska, Katarzyna, Gaweda-Walerych, K., Ingallina, E., Tonelli, C., Morelli, M. J., Amato, A., Eterno, V., Zambelli, A., Rosato, A., Amati, B., Wisniewski, J. R., and Del Sal, G.
- Subjects
p53 ,0301 basic medicine ,Animals ,Antineoplastic Agents ,Humans ,Mice ,MicroRNAs ,Mutant Proteins ,Mutation, Missense ,NF-E2-Related Factor 2 ,Neoplasm Metastasis ,Oligopeptides ,Proteasome Endopeptidase Complex ,Proteome ,Quinuclidines ,Triple Negative Breast Neoplasms ,Tumor Suppressor Protein p53 ,Mutant ,Biology ,cancer ,proteasome ,NRF2 ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,Gene expression ,medicine ,Cell Biology ,Carfilzomib ,Cell biology ,030104 developmental biology ,chemistry ,Proteasome ,Mutation ,Cancer cell ,Proteasome inhibitor ,Missense ,Chromatin immunoprecipitation ,medicine.drug - Abstract
In cancer, the tumour suppressor gene TP53 undergoes frequent missense mutations that endow mutant p53 proteins with oncogenic properties. Until now, a universal mutant p53 gain-of-function program has not been defined. By means of multi-omics: proteome, DNA interactome (chromatin immunoprecipitation followed by sequencing) and transcriptome (RNA sequencing/microarray) analyses, we identified the proteasome machinery as a common target of p53 missense mutants. The mutant p53–proteasome axis globally affects protein homeostasis, inhibiting multiple tumour-suppressive pathways, including the anti-oncogenic KSRP–microRNA pathway. In cancer cells, p53 missense mutants cooperate with Nrf2 (NFE2L2) to activate proteasome gene transcription, resulting in resistance to the proteasome inhibitor carfilzomib. Combining the mutant p53-inactivating agent APR-246 (PRIMA-1MET) with the proteasome inhibitor carfilzomib is effective in overcoming chemoresistance in triple-negative breast cancer cells, creating a therapeutic opportunity for treatment of solid tumours and metastasis with mutant p53. Walerych et al. show that p53 missense mutants upregulate the proteasome and increase breast cancer cell resistance to proteasome inhibitors. Combined inhibition of p53 mutants and the proteasome leads to increased therapeutic efficacy.
- Published
- 2016
- Full Text
- View/download PDF
21. The mitochondrial calcium uniporter regulates breast cancer progression via HIF‐1α
- Author
-
Antonio Rosato, Robert B Bentham, Ivan Bogeski, Rosario Rizzuto, Michael R. Duchen, Tunde Berecz, Thomas S. Blacker, Anna Tosatto, Roberta Sommaggio, Cristina Mammucari, Carsten Kummerow, and Gyorgy Szabadkai
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Motility ,HIF-1α ,Triple Negative Breast Neoplasms ,Mice, SCID ,Biology ,Metastasis ,mitochondrial Ca2+ uptake ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,Downregulation and upregulation ,Cell Movement ,Internal medicine ,Cell Line, Tumor ,medicine ,Gene silencing ,metastasis ,Animals ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Neoplasm Metastasis ,Mitochondrial Ca2+ uptake ,Reactive oxygen species ,Molecular Medicine ,Research Articles ,Cancer ,Cell Proliferation ,reactive oxygen species ,Cell growth ,HIF‐1α ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Molecular medicine ,030104 developmental biology ,Endocrinology ,Metabolism ,030220 oncology & carcinogenesis ,Cancer research ,Heterografts ,Female ,Calcium Channels ,Research Article - Abstract
Triple‐negative breast cancer (TNBC) represents the most aggressive breast tumor subtype. However, the molecular determinants responsible for the metastatic TNBC phenotype are only partially understood. We here show that expression of the mitochondrial calcium uniporter (MCU), the selective channel responsible for mitochondrial Ca2+ uptake, correlates with tumor size and lymph node infiltration, suggesting that mitochondrial Ca2+ uptake might be instrumental for tumor growth and metastatic formation. Accordingly, MCU downregulation hampered cell motility and invasiveness and reduced tumor growth, lymph node infiltration, and lung metastasis in TNBC xenografts. In MCU‐silenced cells, production of mitochondrial reactive oxygen species (mROS) is blunted and expression of the hypoxia‐inducible factor‐1α (HIF‐1α) is reduced, suggesting a signaling role for mROS and HIF‐1α, downstream of mitochondrial Ca2+. Finally, in breast cancer mRNA samples, a positive correlation of MCU expression with HIF‐1α signaling route is present. Our results indicate that MCU plays a central role in TNBC growth and metastasis formation and suggest that mitochondrial Ca2+ uptake is a potential novel therapeutic target for clinical intervention.
- Published
- 2016
22. Optimization of a gmp-grade large-scale expansion protocol for cytokine-induced killer cells using gas-permeable static culture flasks
- Author
-
Roberta Sommaggio, Giuseppe Astori, P Palmerini, Marco Ruggeri, Carlo Visco, Maria Chiara Tisi, Elisa Cappuzzello, Omar Perbellini, Katia Chieregato, Antonio Rosato, and A Dalla Pietà
- Subjects
Cancer Research ,Transplantation ,medicine.diagnostic_test ,Cytokine-induced killer cell ,Chemistry ,Immunology ,Cell Biology ,NKG2D ,Molecular biology ,Flow cytometry ,Cell therapy ,Laboratory flask ,Oncology ,Antigen ,Cell culture ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Genetics (clinical) - Abstract
Background & Aim Cytokine-Induced Killer (CIK) cells are ex vivo expanded T cells with NK cell phenotype. They express both CD3 and CD56 antigens, and exert a potent antitumor activity against a variety of tumors. Several clinical trials demonstrated the safety and the feasibility of CIK cell therapy, with very low side effects and minimal graft-versus-host toxicity. In this study, we developed a GMP-compliant protocol for robust large-scale expansion of CIK cells using G-Rex® gas-permeable static culture flasks. Methods, Results & Conclusion CIK cells were obtained by stimulating healthy donor PBMCs with GMP-grade IFN-γ, IL-2 and CD3 mAbs, and were cultured in G-Rex6® or G-Rex®6M well plates. CIK cells in G-Rex6® were split only once at day 7 to reduce cell density, whereas the number of CIK cells culterd in G-Rex®6M was not adjusted. In both culture conditions, fresh IL-2 was provided every 3-4 days. We compared these two culture protocols with the culture in standard flasks. Phenotype was analyzed by flow cytometry and cytotoxicity was assessed against several tumor cell lines by calcein-release assay. CIK cells cultured in G-Rex6® well plates showed an outstanding cell expansion compared to G-Rex®6M well plates or standard culture flasks, with a 400-fold expansion and a mean of 109 total cells obtained per single well in 14 days, starting from just 2.5 × 106 cells per well. Moreover, the cultures in G-Rex6® were characterized by an higher percentage of CD3+CD56+ cells, as compared to G-Rex®6M or standard culture flasks (38.6±14.9%, 35.0±15.5%, 24.0±17.8%, respectively). Cells cultured in all devices had a comparable expression of NKG2D, NKp30, NKp44, 2B4 receptors. Importantly, CIK cells expanded in G-Rex®6 were as cytotoxic as cells expanded in standard culture flasks. Conversely, CIK cells cultured in G-Rex®6M showed a remarkable reduction of cytotoxicity against tumor cell targets, thus suggesting that cell density during expansion could affect CIK cell activity. We propose a GMP-compliant protocol for robust large-scale production of CIK cells. G-Rex® system allows to obtain large amounts of CIK cells highly enriched in the CD3+CD56+ subset and endowed with high cytotoxic activity; this can be accomplished with just a single cell culture split at day 7, which dramatically reduces the culture manipulation as compared to the standard culture flasks. Notably, this strategy can be further and easily scalable to produce CIK cells for clinical immunotherapy applications.
- Published
- 2020
- Full Text
- View/download PDF
23. Cytokines for the induction of antitumor effectors: The paradigm of Cytokine-Induced Killer (CIK) cells
- Author
-
Antonio Rosato, Roberta Sommaggio, Paola Zanovello, and Elisa Cappuzzello
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Endocrinology, Diabetes and Metabolism ,T-Lymphocytes ,Immunology ,Graft vs Host Disease ,Biology ,Immunotherapy, Adoptive ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Cytokine-Induced Killer Cells ,Neoplasms ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Humans ,Graft-versus-Host disease ,Lymphokine-activated killer cell ,Adoptive Cell Therapy ,Cytokine-induced killer cell ,Cytokine-Induced Killer cells ,Interferon-γ ,Interleukin-2 ,MHC-unrestricted killing ,NKG2D ,Natural killer T cell ,Chimeric antigen receptor ,030104 developmental biology ,Interleukin 15 ,NK Cell Lectin-Like Receptor Subfamily K ,030220 oncology & carcinogenesis ,Cytokines - Abstract
Cytokine-Induced killer (CIK) cells are raising growing interest in cellular antitumor therapy, as they can be easily expanded with a straightforward and inexpensive protocol, and are safe requiring only GMP-grade cytokines to obtain very high amounts of cytotoxic cells. CIK cells do not need antigen-specific stimuli to be activated and proliferate, as they recognize and destroy tumor cells in an HLA-independent fashion through the engagement of NKG2D. In several preclinical studies and clinical trials, CIK cells showed a reduced alloreactivity compared to conventional T cells, even when challenged across HLA-barriers; only in a few patients, a mild GVHD occurred after treatment with allogeneic CIK cells. Additionally, their antitumor activity can be redirected and further improved with chimeric antigen receptors, clinical-grade monoclonal antibodies or immune checkpoint inhibitors. The evidence obtained from a growing body of literature support CIK cells as a very promising cell population for adoptive immunotherapy. In this review, all these aspects will be addressed with a particular emphasis on the role of the cytokines involved in CIK cell generation, expansion and functionalization.
- Published
- 2017
24. Identification of a HLA-A*0201-restricted immunogenic epitope from the universal tumor antigen DEPDC1
- Author
-
Antonio Rosato, Anna Tosi, Roberta Sommaggio, Carolina Marotta, Giannino Del Sal, Elisa Cappuzzello, Dawid Walerich, Paola Zanovello, and Silvia Dalla Santa
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Adoptive cell transfer ,medicine.medical_treatment ,Immunology ,Cancer immunotherapy ,Biology ,cytotoxic T lymphocytes ,lcsh:RC254-282 ,Epitope ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Immunology and Allergy ,DEPDC1 ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Tumor antigen ,Oncoantigen ,CTL ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,triple negative breast cancer ,tumor antigen ,lcsh:RC581-607 - Abstract
The identification of universal tumor-specific antigens shared between multiple patients and/or multiple tumors is of great importance to overcome the practical limitations of personalized cancer immunotherapy. Recent studies support the involvement of DEPDC1 in many aspects of cancer traits, such as cell proliferation, resistance to induction of apoptosis and cell invasion, suggesting that it may play key roles in the oncogenic process. In this study, we report that DEPDC1 expression is upregulated in most types of human tumors, and closely linked to a poorer prognosis; therefore, it might be regarded as a novel universal oncoantigen potentially suitable for targeting many different cancers. In this regard, we report the identification of a HLA-A*0201 allele-restricted immunogenic DEPDC1-derived epitope, which is able to induce cytotoxic T lymphocytes (CTL) exerting a strong and specific functional response in vitro toward not only peptide-loaded cells but also triple negative breast cancer (TNBC) cells endogenously expressing the DEPDC1 protein. Such CTL are also therapeutically active against human TNBC xenografts in vivo upon adoptive transfer in immunodeficient mice. Overall, these data provide evidence that this DEPDC1-derived antigenic epitope can be exploited as a new tool for developing immunotherapeutic strategies for HLA-A*0201 patients with TNBC, and potentially many other cancers.
- Published
- 2017
25. Immunotherapeutic Strategies for Gastric Carcinoma: A Review of Preclinical and Clinical Recent Development
- Author
-
Antonio Rosato, Roberta Sommaggio, and Mohamed Osman Ahmed Abozeid
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Immune checkpoint inhibitors ,lcsh:Medicine ,Gastric carcinoma ,Review Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Programmed cell death 1 ,Internal medicine ,medicine ,Animals ,Humans ,Chemotherapy ,Clinical Trials as Topic ,General Immunology and Microbiology ,biology ,business.industry ,Vaccination ,lcsh:R ,General Medicine ,Immunotherapy ,Prognosis ,Clinical trial ,030104 developmental biology ,Tolerability ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,business - Abstract
Gastric carcinoma (GC) is the 2nd most common cause of cancer-related death. Despite advances in conventional treatment and surgical interventions, a high percentage of GC patients still have poor survival. Recently, immunotherapy has become a promising approach to treat GC. Here, we present preclinical and clinical studies encouraging the use of vaccination, adoptive T-cell therapy (ACT), and immune checkpoint inhibitors, such as programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). The ongoing immunotherapy clinical trials have shown promising results in safety and tolerability even in late-stage GC patients. Moreover, we highlight that the combination of ACT with chemotherapy could be the best choice to treat GC.
- Published
- 2017
26. Enhanced EGFR targeting activity of plasmonic nanostructures with engineered GE11 peptide
- Author
-
Antonio Rosato, Moreno Meneghetti, Simone Mocellin, Lucio Litti, Gianfranco Bocchinfuso, Senthilkumar Rajendran, Roberta Sommaggio, Clara Benna, Paolo Conflitti, Marina Gobbo, Donato Nitti, Antonio Palleschi, and Francesca Biscaglia
- Subjects
Molecular dynamic ,Materials science ,Biomedical Engineering ,Pharmaceutical Science ,Nanoparticle ,Metal Nanoparticles ,Cetuximab ,Peptide ,02 engineering and technology ,Polyethylene glycol ,010402 general chemistry ,01 natural sciences ,Antibodies ,Polyethylene Glycols ,Biomaterials ,chemistry.chemical_compound ,Drug Delivery Systems ,Nanostructures ,Peptides ,SERS ,Targeting activity ,Caco-2 Cells ,Humans ,Receptor, Epidermal Growth Factor ,Gold ,3003 ,PEG ratio ,Epidermal growth factor receptor ,Settore CHIM/02 - Chimica Fisica ,chemistry.chemical_classification ,biology ,Epidermal Growth Factor ,021001 nanoscience & nanotechnology ,Ligand (biochemistry) ,Small molecule ,Combinatorial chemistry ,0104 chemical sciences ,ErbB Receptors ,chemistry ,Colloidal gold ,biology.protein ,0210 nano-technology ,Receptor - Abstract
Plasmonic nanostructures show important properties for biotechnological applications, but they have to be guided on the target for exploiting their potentialities. Antibodies are the natural molecules for targeting. However, their possible adverse immunogenic activity and their cost have suggested finding other valid substitutes. Small molecules like peptides can be an alternative source of targeting agents, even if, as single molecules, their binding affinity is usually not very good. GE11 is a small dodecapeptide with specific binding to the epidermal growth factor receptor (EGFR) and low immunogenicity. The present work shows that thousands of polyethylene glycol (PEG) chains modified with lysines and functionalized with GE11 on clusters of naked gold nanoparticles, obtained by laser ablation in water, achieves a better targeting activity than that recorded with nanoparticles decorated with the specific anti-EGFR antibody Cetuximab (C225). The insertion of the cationic spacer between the polymeric part of the ligand and the targeting peptide allows for a proper presentation of GE11 on the surface of the nanosystems. Surface enhanced resonance Raman scattering signals of the plasmonic gold nanoparticles are used for quantifying the targeting activity. Molecular dynamic calculations suggest that subtle differences in the exposition of the peptide on the PEG sea are important for the targeting activity.
- Published
- 2017
27. Metabolic control of YAP and TAZ by the mevalonate pathway
- Author
-
Antonio Rosato, Miguel Mano, Roberta Sommaggio, Stefano Piccolo, Andrea Manfrin, Valeria Specchia, Silvano Piazza, Eleonora Ingallina, Naomi Ruggeri, Sirio Dupont, Michelangelo Cordenonsi, Giovanni Sorrentino, Giannino Del Sal, Sorrentino, Giovanni, Ruggeri, Naomi, Specchia, Valeria, Cordenonsi, Michelangelo, Mano, Miguel, Dupont, Sirio, Manfrin, Andrea, Ingallina, Eleonora, Sommaggio, Roberta, Piazza, Silvano, Rosato, Antonio, Piccolo, Stefano, DEL SAL, Giannino, and Del Sal, Giannino
- Subjects
TAZ ,rho GTP-Binding Proteins ,Transcription, Genetic ,Geranylgeranyl pyrophosphate ,Transcription Factor ,Pyridines ,Pyridine ,Protein-Serine-Threonine Kinase ,Mice ,chemistry.chemical_compound ,Geranylgeranylation ,HEK293 Cell ,Polyisoprenyl Phosphates ,Drosophila Proteins ,Phosphorylation ,RNA, Small Interfering ,Nuclear Protein ,Sterol Regulatory Element Binding Proteins ,Active Transport, Cell Nucleu ,Intracellular Signaling Peptides and Proteins ,Adaptor Proteins ,Nuclear Proteins ,Protein-Serine-Threonine Kinases ,Active Transport ,Cell biology ,Sterol Regulatory Element Binding Protein ,Drosophila melanogaster ,Trans-Activator ,Phosphoprotein ,Female ,RNA Interference ,YAP ,Mevalonate pathway ,Signal transduction ,Transcription ,Breast Neoplasm ,Human ,Signal Transduction ,Active Transport, Cell Nucleus ,Mevalonic Acid ,rho GTP-Binding Protein ,Breast Neoplasms ,Mevalonic acid ,Protein Serine-Threonine Kinases ,Biology ,Small Interfering ,Hydroxymethylglutaryl-CoA Reductases ,Genetic ,NAD-Dependent ,Animals ,Humans ,Polyisoprenyl Phosphate ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Cell Nucleus ,Tumor Suppressor Protein ,Hippo signaling pathway ,Animal ,Tumor Suppressor Proteins ,Signal Transducing ,YAP-Signaling Proteins ,Cell Biology ,Hydroxymethylglutaryl-CoA Reductases, NAD-Dependent ,HCT116 Cells ,Phosphoproteins ,HEK293 Cells ,chemistry ,Intracellular Signaling Peptides and Protein ,HCT116 Cell ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Trans-Activators ,Transcription Factors ,Drosophila Protein ,RNA ,Hydroxymethylglutaryl-CoA Reductase Inhibitor ,Acyltransferases - Abstract
The YAP and TAZ mediators of the Hippo pathway (hereafter called YAP/TAZ) promote tissue proliferation and organ growth. However, how their biological properties intersect with cellular metabolism remains unexplained. Here, we show that YAP/TAZ activity is controlled by the SREBP/mevalonate pathway. Inhibition of the rate-limiting enzyme of this pathway (HMG-CoA reductase) by statins opposes YAP/TAZ nuclear localization and transcriptional responses. Mechanistically, the geranylgeranyl pyrophosphate produced by the mevalonate cascade is required for activation of Rho GTPases that, in turn, activate YAP/TAZ by inhibiting their phosphorylation and promoting their nuclear accumulation. The mevalonate-YAP/TAZ axis is required for proliferation and self-renewal of breast cancer cells. In Drosophila melanogaster, inhibition of mevalonate biosynthesis and geranylgeranylation blunts the eye overgrowth induced by Yorkie, the YAP/TAZ orthologue. In tumour cells, YAP/TAZ activation is promoted by increased levels of mevalonic acid produced by SREBP transcriptional activity, which is induced by its oncogenic cofactor mutant p53. These findings reveal an additional layer of YAP/TAZ regulation by metabolic cues.
- Published
- 2014
- Full Text
- View/download PDF
28. Enhancing Cytokine-Induced Killer (CIK) cell activity with Her2-specific Fc-engineered antibodies and antibody derivatives
- Author
-
M. Peipp, Antonio Rosato, Roberta Sommaggio, Elisa Cappuzzello, and C. Kellner
- Subjects
Cancer Research ,biology ,Chemistry ,medicine.medical_treatment ,02 engineering and technology ,021001 nanoscience & nanotechnology ,Cell activity ,03 medical and health sciences ,0302 clinical medicine ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,Antibody ,0210 nano-technology - Published
- 2018
- Full Text
- View/download PDF
29. Inhibition of complement component C5 protects porcine chondrocytes from xenogeneic rejection
- Author
-
C. Costa, Rafael Mañez, Magdiel Pérez-Cruz, Roberta Sommaggio, and J.L. Brokaw
- Subjects
Cartilage, Articular ,Graft Rejection ,Swine ,Xenotransplantation ,medicine.medical_treatment ,Transplantation, Heterologous ,Complement ,Biomedical Engineering ,CD59 Antigens ,Complement C5a ,Complement receptor ,CD59 ,Complement Membrane Attack Complex ,Mice ,Chondrocytes ,Antigen ,Rheumatology ,Blocking antibody ,medicine ,Animals ,Humans ,Orthopedics and Sports Medicine ,Mice, Knockout ,biology ,Interleukin-6 ,Cartilage ,Graft Survival ,Histocompatibility Antigens Class I ,Interleukin-8 ,Histocompatibility Antigens Class II ,Antibodies, Monoclonal ,Complement C5 ,Galactosyltransferases ,Cell biology ,Complement system ,medicine.anatomical_structure ,Immunology ,biology.protein ,Cytokines ,Heterografts ,Antibody ,Chemokines - Abstract
Summary Objective Tissue-based xenografts such as cartilage are rejected within weeks by humoral and cellular mechanisms that preclude its clinical application in regenerative medicine. The problem could be overcome by identifying key molecules triggering rejection and the development of genetic-engineering strategies to counteract them. Accordingly, high expression of α1,2-fucosyltransferase (HT) in xenogeneic cartilage reduces the galactose α1,3-galactose (Gal) antigen and delays rejection. Yet, the role of complement activation in this setting is unknown. Design To determine its contribution, we assessed the effect of inhibiting C5 complement component in α1,3-galactosyltransferase-knockout (Gal KO) mice transplanted with porcine cartilage and studied the effect of human complement on porcine articular chondrocytes (PAC). Results Treatment with an anti-mouse C5 blocking antibody for 5 weeks enhanced graft survival by reducing cellular rejection. Moreover, PAC were highly resistant to complement-mediated lysis and primarily responded to human complement by releasing IL-6 and IL-8. This occurred even in the absence of anti-Gal antibody and was mediated by both C5a and C5b-9. Indeed, C5a directly triggered IL-6 and IL-8 secretion and up-regulated expression of swine leukocyte antigen I (SLA-I) and adhesion molecules on chondrocytes, all processes that enhance cellular rejection. Finally, the use of anti-human C5/C5a antibodies and/or recombinant expression of human complement regulatory molecule CD59 (hCD59) conferred protection in correspondence with their specific functions. Conclusions Our study demonstrates that complement activation contributes to rejection of xenogeneic cartilage and provides valuable information for selecting approaches for complement inhibition.
- Published
- 2013
- Full Text
- View/download PDF
30. Mutant p53 Reprograms TNF Signaling in Cancer Cells through Interaction with the Tumor Suppressor DAB2IP
- Author
-
Marco Dal Ferro, Antonio Rosato, Giulio Di Minin, Simona Nuzzo, Licio Collavin, Roberta Bulla, Arianna Bellazzo, Damiano Rami, Roberta Sommaggio, Silvano Piazza, Giulia Chiaruttini, Giannino Del Sal, Silvio Bicciato, Giulio, Di minin, Bellazzo, Arianna, Marco, Dal ferro, Giulia, Chiaruttini, Simona, Nuzzo, Silvio, Bicciato, Silvano, Piazza, Rami, Damiano, Bulla, Roberta, Roberta, Sommaggio, Antonio, Rosato, DEL SAL, Giannino, and Collavin, Licio
- Subjects
p53 ,Chemokine ,mutant p53 gain of function ,Mutant p53, cancer cell, tumor suppressor ,biology ,Cancer ,Inflammation ,Cell Biology ,medicine.disease ,DAB2IP ,Proinflammatory cytokine ,law.invention ,Tumor Necrosis Factor ,law ,Cancer cell ,Immunology ,medicine ,Cancer research ,biology.protein ,Suppressor ,Tumor necrosis factor alpha ,medicine.symptom ,Signal transduction ,Molecular Biology - Abstract
Inflammation is a significant factor in cancer development, and a molecular understanding of the parameters dictating the impact of inflammation on cancers could significantly improve treatment. The tumor suppressor p53 is frequently mutated in cancer, and p53 missense mutants (mutp53) can acquire oncogenic properties. We report that cancer cells with mutp53 respond to inflammatory cytokines increasing their invasive behavior. Notably, this action is coupled to expression of chemokines that can expose the tumor to host immunity, potentially affecting response to therapy. Mechanistically, mutp53 fuels NF-κB activation while it dampens activation of ASK1/JNK by TNFα, and this action depends on mutp53 binding and inhibiting the tumor suppressor DAB2IP in the cytoplasm. Interfering with such interaction reduced aggressiveness of cancer cells in xenografts. This novel interaction is an unexplored mechanism by which mutant p53 can influence tumor evolution, with implications for our understanding of the complex role of inflammation in cancer.
- Published
- 2014
31. Retargeting cytokine-induced killer cell activity by CD16 engagement with clinical-grade antibodies
- Author
-
Roberta Sommaggio, Antonio Rosato, Anna Tosi, Paola Zanovello, and Elisa Cappuzzello
- Subjects
0301 basic medicine ,medicine.drug_class ,medicine.medical_treatment ,Immunology ,Population ,Biology ,Monoclonal antibody ,cytokine induced killer (CIK) cells ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,education ,Original Research ,Antibody-dependent cell-mediated cytotoxicity (ADCC) ,Antibody-dependent cell-mediated cytotoxicity ,education.field_of_study ,Lymphokine-activated killer cell ,Cytokine-induced killer cell ,Immunotherapy ,030104 developmental biology ,Oncology ,immunotherapy ,monoclonal antibodies ,030220 oncology & carcinogenesis ,Ex vivo - Abstract
Cytokine-induced Killer (CIK) cells are a heterogeneous population of ex vivo expanded T lymphocytes capable of MHC-unrestricted antitumor activity, which share phenotypic and functional features with both NK and T cells. Preclinical data and initial clinical studies demonstrated their high tolerability in vivo, supporting CIK cells as a promising cell population for adoptive cell immunotherapy. In this study, we report for the first time that CIK cells display a donor-dependent expression of CD16, which can be engaged by trastuzumab or cetuximab to exert a potent antibody-dependent cell-mediated cytotoxicity (ADCC) against ovarian and breast cancer cell lines, leading to an increased lytic activity in vitro, and an enhanced therapeutic efficacy in vivo. Thus, an efficient tumor antigen-specific retargeting can be achieved by a combination therapy with clinical-grade monoclonal antibodies already widely used in cancer therapy, and CIK cell populations that are easily expandable in very large numbers, inexpensive, safe and do not require genetic manipulations. Overall, these data provide a new therapeutic strategy for the treatment of Her2 and EGFR expressing tumors by adoptive cell therapy, which could find wide implementation and application, and could also be expanded to the use of additional therapeutic antibodies.
- Published
- 2016
32. Cytokine-Induced Killer cells combined with anti EGFR monoclonal antibody abrogate triple negative breast cancer metastatization
- Author
-
Elisa Cappuzzello, Roberta Sommaggio, and Antonio Rosato
- Subjects
Cancer Research ,Oncology ,Cytokine-induced killer cell ,business.industry ,Cancer research ,Medicine ,Anti-EGFR Monoclonal Antibody ,business ,Triple-negative breast cancer - Published
- 2018
- Full Text
- View/download PDF
33. CD8+ T cells against multiple tumor-associated antigens in peripheral blood of midgut carcinoid patients
- Author
-
Magnus Essand, Roberta Sommaggio, Kjell Öberg, Sofia Vikman, Thomas H. Tötterman, and Valeria Giandomenico
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Amino Acid Motifs ,Molecular Sequence Data ,Immunology ,Epitopes, T-Lymphocyte ,Carcinoid Tumor ,CD8-Positive T-Lymphocytes ,Biology ,Epitope ,Cell Line ,Interferon-gamma ,Antigen ,Antigens, Neoplasm ,HLA-A2 Antigen ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Interferon gamma ,RNA, Messenger ,Pan-T antigens ,HLA-A Antigens ,Reverse Transcriptase Polymerase Chain Reaction ,Computational Biology ,Midgut ,Immunotherapy ,Flow Cytometry ,Oncology ,Peptides ,Algorithms ,Epitope Mapping ,CD8 ,medicine.drug - Abstract
The aim of the study was to identify immunogenic HLA-A*0201-binding epitopes derived from a number of classical midgut carcinoid-associated proteins. CD8(+) T cells recognizing tumor-associated antigen (TAA) epitopes are of great interest for the establishment of immunotherapy as a novel treatment for this type of malignancy.Midgut carcinoid tumor specimens were microdissected and expression levels of potential TAAs were investigated by quantitative real time PCR. HLA-A*0201-binding motifs were selected using HLA peptide binding prediction algorithms and stabilization of HLA-A*0201 was verified using TAP-deficient T2 cells. Peripheral blood of midgut carcinoid patients was analyzed for peptide epitope recognition and the feasibility of generating peptide-reactive CD8(+) T cells in healthy blood donors was examined by an in vitro stimulation protocol using mature DCs. Activation of patient and healthy donor CD8(+) T cells was analyzed by intracellular flow cytometry staining of interferon gamma.Chromogranin A (CGA), tryptophan hydroxylase 1 (TPH-1), vesicular monoamine transporter 1 (VMAT-1), caudal type homeobox transcription factor 2 (CDX-2), and islet autoantigen 2 (IA-2) are properly expressed by midgut carcinoid tumor cells, with CGA mRNA expressed to greatest level. Midgut carcinoid patients have increased frequencies of peripheral blood CD8(+) T cells recognizing a pool of HLA-A*0201 peptides derived from these proteins compared to healthy age-matched individuals. Activated peptide-specific CD8(+) T cells could also be generated in healthy blood donors by in vitro stimulation.We have identified a number of immunogenic midgut carcinoid-associated peptide epitopes recognized by CD8(+) T cells. We show that midgut carcinoid patients display immune recognition of their tumors. Memory CD8(+) T cells in patient blood are of great interest when pursuing an immunotherapeutic treatment strategy.
- Published
- 2007
- Full Text
- View/download PDF
34. Genetic engineering strategies to prevent the effects of antibody and complement on xenogeneic chondrocytes
- Author
-
Magdiel Pérez-Cruz, D. Bello-Gil, Cristina Costa, Roberta Sommaggio, Rafael Mañez, and J.L. Brokaw
- Subjects
lcsh:Diseases of the musculoskeletal system ,Swine ,Transgene ,Transplantation, Heterologous ,lcsh:Surgery ,CD59 ,Biology ,anaphylatoxin ,Antibodies ,Animals, Genetically Modified ,Complement inhibitor ,Chondrocytes ,antibody ,medicine ,Animals ,complement ,Anaphylatoxin ,Cartilage ,complement regulatory proteins ,lcsh:RD1-811 ,Complement System Proteins ,Complement system ,Cell biology ,Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,cytokine release ,Immunology ,biology.protein ,Xenotransplantation ,lcsh:RC925-935 ,Antibody ,Genetic Engineering - Abstract
Advances in animal transgenesis may allow using xenogeneic chondrocytes in tissue-engineering applications for clinical cartilage repair. Porcine cartilage is rejected by humoral and cellular mechanisms that could be overcome by identifying key molecules triggering rejection and developing effective genetic-engineering strategies. Accordingly, high expression of α1,2-fucosyltransferase (HT) in xenogeneic cartilage protects from galactose α1,3-galactose (Gal)-mediated antibody responses. Now, we studied whether expression of a complement inhibitor provides further protection. First, porcine articular chondrocytes (PAC) were isolated from non-transgenic, single and double transgenic pigs expressing HT and moderate levels of human CD59 (hCD59) and their response to human serum was assessed. High recombinant expression of human complement regulatory molecules hCD59 and hDAF was also attained by retroviral transduction of PAC for further analyses. Complement activation on PAC after exposure to 20 % human serum for 24 hours mainly triggered the release of pro-inflammatory cytokines IL-6 and IL-8. Transgenic expression of HT and hCD59 did not suffice to fully counteract this effect. Nevertheless, the combination of blocking anti-Gal antibodies (or C5a) and high hCD59 levels conferred very high protection. On the contrary, high hDAF expression attained the most dramatic reduction in IL-6/IL-8 secretion by a single strategy, but the additional inhibition of anti-Gal antibodies or C5a did not provide further improvement. Notably, we demonstrate that both hCD59 and hDAF inhibit anaphylatoxin release in this setting. In conclusion, our study identifies genetic-engineering approaches to prevent humoral rejection of xenogeneic chondrocytes for use in cartilage repair.
- Published
- 2015
35. HMGA1 promotes metastatic processes in basal-like breast cancer regulating EMT and stemness
- Author
-
Riccardo Sgarra, Roberta Sommaggio, Yari Ciani, Gloria Ros, Silvia Pegoraro, Antonio Rosato, Giannino Del Sal, Guidalberto Manfioletti, Silvano Piazza, Pegoraro, Silvia, Ros, Gloria, Piazza, S., Sommaggio, R., Ciani, Yari, Rosato, A., Sgarra, Riccardo, DEL SAL, Giannino, and Manfioletti, Guidalberto
- Subjects
Oncology ,medicine.medical_specialty ,HMGA1 ,Epithelial-Mesenchymal Transition ,Breast Neoplasms ,Cell Growth Processes ,Mice, SCID ,Biology ,HMGA1 Gene ,Metastasis ,Mice ,stemness ,Breast cancer ,Epithelial to mesenchymal transition ,breast cancer ,invasion ,metastasis ,gene-signature ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,HMGA1a Protein ,Neoplasm Metastasis ,Mesenchymal stem cell ,Wnt signaling pathway ,Gene signature ,medicine.disease ,Prognosis ,stemne ,Carcinoma, Basal Cell ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Heterografts ,metastasi ,Female ,Transcriptome ,Signal Transduction ,Research Paper - Abstract
// Silvia Pegoraro 1 , Gloria Ros 1 , Silvano Piazza 2 , Roberta Sommaggio 3 , Yari Ciani 1,2 , Antonio Rosato 3,4 , Riccardo Sgarra 1 , Giannino Del Sal 1,2 , and Guidalberto Manfioletti 1,* 1 Dipartimento di Scienze della Vita, Universita degli Studi di Trieste, Trieste, Italy 2 Laboratorio Nazionale CIB, (LNCIB), Area Science Park, Trieste, Italy 3 Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy 4 Istituto Oncologico Veneto IRCCS, Padova, Italy Correspondence: Guidalberto Manfioletti, email: // Keywords : Epithelial to mesenchymal transition, stemness, HMGA1, breast cancer, invasion, metastasis, gene-signature Received : June 28, 2013 Accepted : July 7, 2013 Published : July 9, 2013 Abstract Breast cancer is a heterogeneous disease that progresses to the critical hallmark of metastasis. In the present study, we show that the High Mobility Group A1 (HMGA1) protein plays a fundamental role in this process in basal-like breast cancer subtype. HMGA1 knockdown induces the mesenchymal to epithelial transition and dramatically decreases stemness and self-renewal. Notably, HMGA1 depletion in basal-like breast cancer cell lines reduced migration and invasion in vitro and the formation of metastases in vivo . Mechanistically, HMGA1 activated stemness and key migration-associated genes which were linked to the Wnt/beta-catenin, Notch and Pin1/mutant p53 signalling pathways. Moreover, we identified a specific HMGA1 gene expression signature that was activated in a large subset of human primary breast tumours and was associated with poor prognosis. Taken together, these data provide new insights into the role of HMGA1 in the acquisition of aggressive features in breast cancer.
- Published
- 2013
36. Cellular studies for in vitro modeling of xenogeneic immune responses
- Author
-
Roberta, Sommaggio, Magdiel, Pérez-Cruz, and Cristina, Costa
- Subjects
Cytotoxicity, Immunologic ,Killer Cells, Natural ,Swine ,Primary Cell Culture ,Transplantation, Heterologous ,Cell Adhesion ,Animals ,Cytokines ,Humans ,Coculture Techniques ,Cell Line - Abstract
Cellular studies are essential in the xenotransplantation field in order to investigate the cellular immune responses triggered by xenogeneic cells and identify the key molecules involved. A series of functional studies can be conducted with this purpose that include treatment with proinflammatory cytokines and xenogeneic cell-based assays that put together pig cells and human leukocytes such as monocytes, NK cells, and T cells. The choice of the pig cell type is critical to appropriately model the transplant setting of interest. Thus, pig endothelial cells are commonly used for studying the rejection process of vascularized organs. Treatment with cytokines allows studying the regulation of adhesion, costimulatory molecules, and receptors involved in triggering the immune response in an attempt to reproduce the more complex in vivo situation. The adhesion assays are used to determine the capacity of human leukocytes to adhere to porcine cells under various conditions. Furthermore, we describe coculture, costimulatory, and cytotoxicity assays for investigating the cellular and molecular mechanisms that take place during the xenogeneic immune response.
- Published
- 2012
37. Cellular Studies for In Vitro Modeling of Xenogeneic Immune Responses
- Author
-
Magdiel Pérez-Cruz, Cristina Costa, and Roberta Sommaggio
- Subjects
Cell type ,Immune system ,medicine.anatomical_structure ,Xenotransplantation ,medicine.medical_treatment ,Cell ,medicine ,Biology ,Cytotoxicity ,Receptor ,In vitro ,Proinflammatory cytokine ,Cell biology - Abstract
Cellular studies are essential in the xenotransplantation field in order to investigate the cellular immune responses triggered by xenogeneic cells and identify the key molecules involved. A series of functional studies can be conducted with this purpose that include treatment with proinflammatory cytokines and xenogeneic cell-based assays that put together pig cells and human leukocytes such as monocytes, NK cells, and T cells. The choice of the pig cell type is critical to appropriately model the transplant setting of interest. Thus, pig endothelial cells are commonly used for studying the rejection process of vascularized organs. Treatment with cytokines allows studying the regulation of adhesion, costimulatory molecules, and receptors involved in triggering the immune response in an attempt to reproduce the more complex in vivo situation. The adhesion assays are used to determine the capacity of human leukocytes to adhere to porcine cells under various conditions. Furthermore, we describe coculture, costimulatory, and cytotoxicity assays for investigating the cellular and molecular mechanisms that take place during the xenogeneic immune response.
- Published
- 2012
- Full Text
- View/download PDF
38. Multiple receptors trigger human NK cell-mediated cytotoxicity against porcine chondrocytes
- Author
-
Cristina Costa, André Cohnen, Carsten Watzl, and Roberta Sommaggio
- Subjects
Swine ,Xenotransplantation ,medicine.medical_treatment ,Immunology ,Biology ,Ligands ,Microbiology ,Proinflammatory cytokine ,Chondrocytes ,Cell Adhesion ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxicity ,Cell adhesion ,Receptor ,Cells, Cultured ,Cell Death ,Cartilage ,Antibody-Dependent Cell Cytotoxicity ,Cytotoxicity Tests, Immunologic ,NKG2D ,Coculture Techniques ,Cell biology ,Killer Cells, Natural ,Cytolysis ,medicine.anatomical_structure ,Receptors, Natural Killer Cell - Abstract
Xenotransplantation of genetically engineered porcine chondrocytes may provide a therapeutic solution for the repair of cartilage defects of various types. However, the mechanisms underlying the humoral and cellular responses that lead to rejection of xenogeneic cartilage are not well understood. In this study, we investigated the interaction between human NK cells and isolated porcine costal chondrocytes (PCC). Our data show that freshly isolated NK cells adhere weakly to PCC. Consequently, PCC were highly resistant to cytolysis mediated by freshly isolated NK cells. However, the presence of human natural Abs in the coculture was often sufficient to trigger cytotoxicity against PCC. Furthermore, IL-2 stimulation of NK cells or activation of PCC with the proinflammatory cytokines TNF-α or IL-1α resulted in increased adhesion, which was paralleled by increased NK cell-mediated lysis of PCC. NK cell adhesion to PCC could be blocked by Abs against human LFA-1 and porcine VCAM-1. NKG2D and NKp44 were involved in triggering cytotoxicity against PCC, which expressed ligands for these activating NK cell receptors. Our data further suggest that NKp30 and NKp46 may contribute to the activation of NK cells by PCC under certain conditions. Finally, comparative studies confirmed that PCC are more resistant than porcine aortic endothelial cells to human NK cell-mediated lysis. Thus, the data demonstrate that human NK cells can kill pig chondrocytes and may therefore contribute to rejection of xenogeneic cartilage. In addition, we identify potential targets for intervention to prevent the NK cell response against pig xenografts.
- Published
- 2012
39. Midgut carcinoid patients display increased numbers of regulatory T cells in peripheral blood with infiltration into tumor tissue
- Author
-
Thomas H. Tötterman, Valeria Giandomenico, Magnus Essand, Manuel de la Torre, Roberta Sommaggio, Kjell Öberg, Sofia Vikman, and Angelica Loskog
- Subjects
Medicin och hälsovetenskap ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,T cell ,Fluorescent Antibody Technique ,Enzyme-Linked Immunosorbent Assay ,Carcinoid Tumor ,Lymphocyte Activation ,Immunofluorescence ,Medical and Health Sciences ,T-Lymphocytes, Regulatory ,Flow cytometry ,Immunoenzyme Techniques ,Lymphocytes, Tumor-Infiltrating ,Intestinal Neoplasms ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,IL-2 receptor ,medicine.diagnostic_test ,business.industry ,Liver Neoplasms ,FOXP3 ,Midgut ,Hematology ,General Medicine ,Flow Cytometry ,Cytokine ,medicine.anatomical_structure ,Oncology ,Case-Control Studies ,CD4 Antigens ,Cytokines ,Immunohistochemistry ,business - Abstract
INTRODUCTION: Our aim was to investigate the immune status of midgut carcinoid patients. Cancer patients generally display suppressed Th1-type immunity that disables mounting of an efficient anti-tumor response. However, little is known about patients with neuroendocrine midgut carcinoids. MATERIAL AND METHODS: Circulating regulatory T cells were determined in patient blood by staining for CD4, CD25 and FoxP3 in flow cytometric analysis. T cell proliferation was measured by Alamar Blue in response to polyclonal activation and the regulatory phenotype of patient CD25+ cells was validated by allogeneic stimulation of CFSE labelled responders. Cytokine levels in patient peripheral blood were measured by ELISA and CBA. Tumor infiltrating T cells were analyzed by immunohistochemistry and immunofluorescence. RESULTS: The results demonstrate that midgut carcinoid patients exhibit increased frequencies of circulating Tregs and patient T cells have a decreased proliferative capacity compared to healthy donors. Systemic Th1-promoting cytokines are reduced. Midgut carcinoid tumors display CD4+ and CD8+ T cell infiltration, always in the presence of regulatory CD4+FoxP3+ cells. DISCUSSION: Midgut carcinoid patients display elevated T regulatory cell numbers and T cell dysfunction. Therapeutic strategies to overcome tumor-induced Th1 immunosuppression are required in combination with anti-tumor vaccinations.
- Published
- 2009
40. TNF, pig CD86, and VCAM-1 identified as potential targets for intervention in xenotransplantation of pig chondrocytes
- Author
-
Rafael Mañez, Roberta Sommaggio, and Cristina Costa
- Subjects
Swine ,Xenotransplantation ,medicine.medical_treatment ,Interleukin-1beta ,Transplantation, Heterologous ,Biomedical Engineering ,Vascular Cell Adhesion Molecule-1 ,lcsh:Medicine ,Apoptosis ,Jurkat cells ,Cell Line ,Jurkat Cells ,Chondrocytes ,Downregulation and upregulation ,Interleukin-1alpha ,Cell Adhesion ,medicine ,Animals ,Humans ,Cell adhesion ,CD86 ,Transplantation ,CD40 ,biology ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Chemistry ,Cartilage ,Interleukin-8 ,lcsh:R ,hemic and immune systems ,Cell Biology ,Intercellular Adhesion Molecule-1 ,Up-Regulation ,Cell biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,B7-2 Antigen ,CD80 - Abstract
Xenotransplantation of genetically engineered porcine chondrocytes may benefit many patients who suffer cartilage defects. In this work, we sought to elucidate the molecular bases of the cellular response to xenogeneic cartilage. To this end, we isolated pig costal chondrocytes (PCC) and conducted a series of functional studies. First, we determined by flow cytometry the cell surface expression of multiple immunoregulatory proteins in resting conditions or after treatment with human TNF-alpha, IL-1alpha, or IL-1beta, which did not induce apoptosis. TNF-alpha and to a lesser extent IL-1alpha led to a marked upregulation of SLA I, VCAM-1, and ICAM-1 on PCC. SLA II and E-selectin remained undetectable in all the conditions assayed. Notably, CD86 was constitutively expressed at moderate levels, whereas CD80 and CD40 were barely detected. To assess their function, we next studied the interaction of PCC with human monoblastic U937 and Jurkat T cells. U937 cells adhered to resting and in a greater proportion to cytokine-stimulated PCC. Consistent with its expression pattern, pig VCAM-1 was key, mediating the increased adhesion after cytokine stimulation. We also conducted coculture experiments with U937 and PCC and measured the release of pig and human cytokines. Stimulated PCC secreted IL-6 and IL-8, whereas U937 secreted IL-8 in response to PCC. Finally, coculture of PCC with Jurkat in the presence of PHA led to a marked Jurkat activation as determined by the increase in IL-2 secretion. This process was dramatically reduced by blocking pig CD86. In summary, CD86 and VCAM-1 on pig chondrocytes may be important triggers of the xenogeneic cellular immune response. These molecules together with TNF could be considered potential targets for intervention in order to develop xenogeneic therapies for cartilage repair.
- Published
- 2009
41. Prolyl-isomerase Pin1 controls normal and cancer stem cells of the breast
- Author
-
Simona Nuzzo, Federica Benvenuti, Alessandra Rustighi, Vincenzo Eterno, Antonio Rosato, Alessandro Zannini, Luca Tiberi, Roberta Sommaggio, Giannino Del Sal, Iannis Aifantis, Antonella Tuscano, Silvio Bicciato, Giovanni Sorrentino, Silvano Piazza, Alberto Zambelli, Libero Santarpia, Rustighi, Alessandra, Zannini, Alessandro, Tiberi, Luca, Sommaggio, Roberta, Piazza, Silvano, Sorrentino, Giovanni, Nuzzo, Simona, Tuscano, Antonella, Eterno, Vincenzo, Benvenuti, Federica, Santarpia, Libero, Aifantis, Ianni, Rosato, Antonio, Bicciato, Silvio, Zambelli, Alberto, and DEL SAL, Giannino
- Subjects
Ubiquitin-Protein Ligase ,F-Box-WD Repeat-Containing Protein 7 ,Cell Cycle Proteins ,Triple Negative Breast Neoplasms ,Stem cells ,Mice, SCID ,Metastasis ,Antineoplastic Agent ,Mice ,Breast cancer ,Receptors ,Cell Cycle Protein ,Transplantation, Heterologou ,Receptor, Notch1 ,Receptor, Notch4 ,Research Articles ,cancer cell ,Mice, Knockout ,Heterologous ,Proto-Oncogene Protein ,Tumor ,Receptors, Notch ,Peptidylprolyl Isomerase ,Mammary Glands ,3. Good health ,stem cells ,Neoplastic Stem Cells ,Fbxw7 E3 ubiquitin-ligase ,Molecular Medicine ,Female ,Stem cell ,Corrigendum ,Breast Neoplasm ,Receptor ,Human ,Signal Transduction ,Notch ,Prolyl-isomerase Pin1 ,Animals ,Antineoplastic Agents ,Breast Neoplasms ,Cell Line, Tumor ,F-Box Proteins ,Humans ,Mammary Glands, Human ,Proto-Oncogene Proteins ,Stem Cells ,Transplantation, Heterologous ,Ubiquitin-Protein Ligases ,Knockout ,Triple Negative Breast Neoplasm ,Notch signaling pathway ,Biology ,SCID ,Cell Line ,Cancer stem cell ,Stem Cell ,medicine ,F-Box Protein ,Peptidylprolyl isomerase ,Notch1 ,Transplantation ,Animal ,Biologie moléculaire ,medicine.disease ,NIMA-Interacting Peptidylprolyl Isomerase ,Immunology ,Cancer cell ,Cancer research ,Neoplastic Stem Cell - Abstract
Mammary epithelial stem cells are fundamental to maintain tissue integrity. Cancer stem cells (CSCs) are implicated in both treatment resistance and disease relapse, and the molecular bases of their malignant properties are still poorly understood. Here we show that both normal stem cells and CSCs of the breast are controlled by the prolyl-isomerase Pin1. Mechanistically, following interaction with Pin1, Notch1 and Notch4, key regulators of cell fate, escape from proteasomal degradation by their major ubiquitin-ligase Fbxw7α. Functionally, we show that Fbxw7α acts as an essential negative regulator of breast CSCs' expansion by restraining Notch activity, but the establishment of a Notch/Pin1 active circuitry opposes this effect, thus promoting breast CSCs self-renewal, tumor growth and metastasis in vivo. In human breast cancers, despite Fbxw7α expression, high levels of Pin1 sustain Notch signaling, which correlates with poor prognosis. Suppression of Pin1 holds promise in reverting aggressive phenotypes, through CSC exhaustion as well as recovered drug sensitivity carrying relevant implications for therapy of breast cancers. © 2013 The Authors., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2014
42. 287 An HMGA1 Specific Transcriptional Program Promotes Metastasis in Breast Cancer
- Author
-
Antonio Rosato, Stefano Gustincich, Yari Ciani, Silvano Piazza, Gloria Ros, Guidalberto Manfioletti, G Del Sal, Silvia Pegoraro, Roberta Sommaggio, and Riccardo Sgarra
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,medicine.disease ,HMGA1 ,Metastasis ,Breast cancer ,Internal medicine ,Cancer research ,biology.protein ,medicine ,business - Published
- 2012
- Full Text
- View/download PDF
43. Xenotransplantation of Pig chondrocytes: Therapeutic potential and barriers for cartilage repair
- Author
-
M Marquina, Roberta Sommaggio, M Uribe-Herranz, and Cristina Costa
- Subjects
Cartilage, Articular ,0301 basic medicine ,Chemokine ,lcsh:Diseases of the musculoskeletal system ,Xenotransplantation ,medicine.medical_treatment ,Sus scrofa ,Transplantation, Heterologous ,T cells ,lcsh:Surgery ,NK cells ,Models, Biological ,03 medical and health sciences ,Chondrocytes ,0302 clinical medicine ,Immune system ,Antigen ,antibody ,medicine ,Animals ,complement ,cartilage ,Immunity, Cellular ,Wound Healing ,030222 orthopedics ,Innate immune system ,biology ,Cell adhesion molecule ,Cartilage ,lcsh:RD1-811 ,Cell biology ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,cytokine release ,biology.protein ,lcsh:RC925-935 ,monocytes - Abstract
Transplantation may be the best option for the repair of many cartilage lesions including early osteoarthritis. Currently, autologous and allogeneic chondrocytes are grafted into cartilage defects to treat selected patients with moderate clinical success. However, their limited use justifies exploring novel therapies for cartilage repair. Xenotransplantation could become a solution by offering high cell availability, quality and genetic engineering capabilities. The rejection process of xenogeneic cartilage is thus being elucidated in order to develop counteractive strategies. Initial studies determined that pig cartilage xenografts are rejected by a slow process comprising humoral and cellular responses in which the galactose α1,3-galactose antigen participates. Since then, our group has identified key mechanisms of the human response to pig chondrocytes (PCs). In particular, human antibody and complement contribute to PC rejection by inducing a pro-inflammatory milieu. Furthermore, PCs express and up-regulate molecules which are functionally relevant for a variety of cellular immune responses (SLA-I, the potent co-stimulatory molecule CD86, and adhesion molecules VCAM-1 and ICAM-1). These participate by triggering a T cell response, as well as supporting a prominent role of the innate immune responses led by natural killer (NK) cells and monocytes/macrophages. Human NK cells lyse PCs by using selected NK activating receptors, whereas human monocytes are activated by PCs to secrete cytokines and chemokines. All this knowledge sets the bases for the development of genetic engineering approaches designed to avert rejection of xenogeneic chondrocytes and leads the way to developing new clinical applications for cartilage repair.
44. Human NK cells kill porcine chondrocytes using multiple receptors
- Author
-
Roberta Sommaggio, Watzl, Carsten, and Costa, Cristina
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.