86 results on '"Pupa SM"'
Search Results
2. Activated d16HER2 homodimers and src kinase mediate optimal efficacy for trastuzumab
- Author
-
Patrizia Gasparini, Giulia Marzano, Patrizia Nanni, Manuela Campiglio, Elda Tagliabue, Arianna Palladini, Pier Luigi Lollini, Claudia Chiodoni, Manuela Iezzi, Alessia Lamolinara, Lorenzo Castagnoli, Valentina Ciravolo, Tiziana Triulzi, Serenella M. Pupa, Gaia C. Ghedini, Augusto Amici, Roberta Zappasodi, Sylvie Ménard, Castagnoli, L, Iezzi, M, Ghedini, Gc, Ciravolo, V, Marzano, G, Lamolinara, A, Zappasodi, R, Gasparini, P, Campiglio, M, Amici, A, Chiodoni, C, Palladini, Arianna, Lollini, PIER LUIGI, Triulzi, T, Menard, S, Nanni, Patrizia, Tagliabue, E, and Pupa, Sm
- Subjects
Genetically modified mouse ,Cancer Research ,Receptor, ErbB-2 ,Transgene ,Mice, Transgenic ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Mice ,mammary cancer ,Trastuzumab ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Isoforms ,skin and connective tissue diseases ,Receptor ,neoplasms ,Effector ,business.industry ,Exons ,src-Family Kinases ,Oncology ,HER-2 ,Drug Resistance, Neoplasm ,Monoclonal ,Cancer research ,Female ,Signal transduction ,Neoplasm Recurrence, Local ,Protein Multimerization ,business ,Delta16-HER-2 ,medicine.drug ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
A splice isoform of the HER2 receptor that lacks exon 16 (d16HER2) is expressed in many HER2-positive breast tumors, where it has been linked with resistance to the HER2-targeting antibody trastuzumab, but the impact of d16HER2 on tumor pathobiology and therapeutic response remains uncertain. Here, we provide genetic evidence in transgenic mice that expression of d16HER2 is sufficient to accelerate mammary tumorigenesis and improve the response to trastuzumab. A comparative analysis of effector signaling pathways activated by d16HER2 and wild-type HER2 revealed that d16HER2 was optimally functional through a link to SRC activation (pSRC). Clinically, HER2-positive breast cancers from patients who received trastuzumab exhibited a positive correlation in d16HER2 and pSRC abundance, consistent with the mouse genetic results. Moreover, patients expressing high pSRC or an activated “d16HER2 metagene” were found to derive the greatest benefit from trastuzumab treatment. Overall, our results establish the d16HER2 signaling axis as a signature for decreased risk of relapse after trastuzumab treatment. Cancer Res; 74(21); 6248–59. ©2014 AACR.
- Published
- 2014
3. Serological identification of HSP105 as a novel non-Hodgkin lymphoma therapeutic target
- Author
-
Italia Bongarzone, Antonella Messina, Claudio Tripodo, Roberta Zappasodi, Carmelo Carlo-Stella, Monica Tortoreto, Michele Magni, Alessandro M. Gianni, Gaia C. Ghedini, Massimo Di Nicola, Lorenzo Castagnoli, Antonello Cabras, Serenella M. Pupa, Zappasodi, R, Bongarzone, I, Ghedini, GC, Castagnoli, L, Cabras, AD, Messina, A, Tortoreto, M, Tripodo, C, Magni, M, Carlo-Stella, C, Gianni, AM, Pupa, SM, and Di Nicola, M.
- Subjects
Immunology ,Mice, SCID ,Biochemistry ,Antibodies ,Flow cytometry ,Antigen-Antibody Reactions ,Cohort Studies ,HSP105 ,Mice ,Antigen ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Serologic Tests ,HSP110 Heat-Shock Proteins ,medicine.diagnostic_test ,biology ,business.industry ,Lymphoma, Non-Hodgkin ,non-Hodgkin lymphoma ,Cell Biology ,Hematology ,Cell cycle ,medicine.disease ,Immunohistochemistry ,Lymphoma ,Granzyme B ,Gene Expression Regulation, Neoplastic ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,biology.protein ,Antibody ,business ,Diffuse large B-cell lymphoma - Abstract
We reported that the clinical efficacy of dendritic cell–based vaccination is strongly associated with immunologic responses in relapsed B-cell non-Hodgkin lymphoma (B-NHL) patients. We have now investigated whether postvaccination antibodies from responders recognize novel shared NHL-restricted antigens. Immunohistochemistry and flow cytometry showed that they cross-react with allogeneic B-NHLs at significantly higher levels than their matched prevaccination samples or nonresponders' antibodies. Western blot analysis of DOHH-2 lymphoma proteome revealed a sharp band migrating at approximately 100 to 110 kDa only with postvaccine repertoires from responders. Mass spectrometry identified heat shock protein-105 (HSP105) in that molecular weight interval. Flow cytometry and immunohistochemistry disclosed HSP105 on the cell membrane and in the cytoplasm of B-NHL cell lines and 97 diagnostic specimens. A direct correlation between HSP105 expression and lymphoma aggressiveness was also apparent. Treatment of aggressive human B-NHL cell lines with an anti-HSP105 antibody had no direct effects on cell cycle or apoptosis but significantly reduced the tumor burden in xenotransplanted immunodeficient mice. In vivo antilymphoma activity of HSP105 engagement was associated with a significant local increase of Granzyme B+ killer cells that very likely contributed to the tumor-restricted necrosis. Our study adds HSP105 to the list of nononcogenes that can be exploited as antilymphoma targets.
- Published
- 2011
4. Breast cancer patient-derived organoids for the investigation of patient-specific tumour evolution.
- Author
-
Mazzucchelli S, Signati L, Messa L, Franceschini A, Bonizzi A, Castagnoli L, Gasparini P, Consolandi C, Mangano E, Pelucchi P, Cifola I, Camboni T, Severgnini M, Villani L, Tagliaferri B, Carelli S, Pupa SM, Cereda C, and Corsi F
- Abstract
Background: A reliable preclinical model of patient-derived organoids (PDOs) was developed in a case study of a 69-year-old woman diagnosed with breast cancer (BC) to investigate the tumour evolution before and after neoadjuvant chemotherapy and surgery. The results were achieved due to the development of PDOs from tissues collected before (O-PRE) and after (O-POST) treatment., Methods: PDO cultures were characterized by histology, immunohistochemistry (IHC), transmission electron microscopy (TEM), scanning electron microscopy (SEM), confocal microscopy, flow cytometry, real-time PCR, bulk RNA-seq, single-cell RNA sequencing (scRNA-seq) and drug screening., Results: Both PDO cultures recapitulated the histological and molecular profiles of the original tissues, and they showed typical mammary gland organization, confirming their reliability as a personalized in vitro model. Compared with O-PRE, O-POST had a greater proliferation rate with a significant increase in the Ki67 proliferation index. Moreover O-POST exhibited a more stem-like and aggressive phenotype, with increases in the CD24
low /CD44low and EPCAMlow /CD49fhigh cell populations characterized by increased tumour initiation potential and multipotency and metastatic potential in invasive lobular carcinoma. Analysis of ErbB receptor expression indicated a decrease in HER-2 expression coupled with an increase in EGFR expression in O-POST. In this context, deregulation of the PI3K/Akt signalling pathway was assessed by transcriptomic analysis, confirming the altered transcriptional profile. Finally, transcriptomic single-cell analysis identified 11 cell type clusters, highlighting the selection of the luminal component and the decrease in the number of Epithelial-mesenchymal transition cell types in O-POST., Conclusion: Neoadjuvant treatment contributed to the enrichment of cell populations with luminal phenotypes that were more resistant to chemotherapy in O-POST. PDOs represent an excellent 3D cell model for assessing disease evolution., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
5. Impact of in vitro SARS-CoV-2 infection on breast cancer cells.
- Author
-
Sommariva M, Dolci M, Triulzi T, Ambrogi F, Dugo M, De Cecco L, Le Noci V, Bernardo G, Anselmi M, Montanari E, Pupa SM, Signorini L, Gagliano N, Sfondrini L, Delbue S, and Tagliabue E
- Subjects
- Humans, Female, Cell Line, Tumor, MCF-7 Cells, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Breast Neoplasms virology, Breast Neoplasms pathology, COVID-19 virology, SARS-CoV-2 physiology, Virus Replication, Tamoxifen pharmacology
- Abstract
The pandemic of coronavirus disease 19 (COVID-19), caused by severe respiratory syndrome coronavirus 2 (SARS-CoV-2), had severe repercussions for breast cancer patients. Increasing evidence indicates that SARS-CoV-2 infection may directly impact breast cancer biology, but the effects of SARS-CoV-2 on breast tumor cells are still unknown. Here, we analyzed the molecular events occurring in the MCF7, MDA-MB-231 and HCC1937 breast cancer cell lines, representative of the luminal A, basal B/claudin-low and basal A subtypes, respectively, upon SARS-CoV-2 infection. Viral replication was monitored over time, and gene expression profiling was conducted. We found that MCF7 cells were the most permissive to viral replication. Treatment of MCF7 cells with Tamoxifen reduced the SARS-CoV-2 replication rate, suggesting an involvement of the estrogen receptor in sustaining virus replication in malignant cells. Interestingly, a metagene signature based on genes upregulated by SARS-CoV-2 infection in all three cell lines distinguished a subgroup of premenopausal luminal A breast cancer patients with a poor prognosis. As SARS-CoV-2 still spreads among the population, it is essential to understand the impact of SARS-CoV-2 infection on breast cancer, particularly in premenopausal patients diagnosed with the luminal A subtype, and to assess the long-term impact of COVID-19 on breast cancer outcomes., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. The Emerging Role of the Microbiota in Breast Cancer Progression.
- Author
-
Bernardo G, Le Noci V, Di Modica M, Montanari E, Triulzi T, Pupa SM, Tagliabue E, Sommariva M, and Sfondrini L
- Subjects
- Animals, Humans, Immunity, Symbiosis, Host Microbial Interactions, Breast microbiology, Breast Neoplasms microbiology, Breast Neoplasms pathology, Breast Neoplasms therapy, Gastrointestinal Microbiome
- Abstract
Emerging evidence suggests a profound association between the microbiota composition in the gastrointestinal tract and breast cancer progression. The gut microbiota plays a crucial role in modulating the immune response, releasing metabolites, and modulating estrogen levels, all of which have implications for breast cancer growth. However, recent research has unveiled a novel aspect of the relationship between the microbiota and breast cancer, focusing on microbes residing within the mammary tissue, which was once considered sterile. These localized microbial communities have been found to change in the presence of a tumor as compared to healthy mammary tissue, unraveling their potential contribution to tumor progression. Studies have identified specific bacterial species that are enriched within breast tumors and have highlighted the mechanisms by which even these microbes influence cancer progression through immune modulation, direct carcinogenic activity, and effects on cellular pathways involved in cell proliferation or apoptosis. This review aims to provide an overview of the current knowledge on the mechanisms of crosstalk between the gut/mammary microbiota and breast cancer. Understanding this intricate interplay holds promise for developing innovative therapeutic approaches.
- Published
- 2023
- Full Text
- View/download PDF
7. Fatty acid synthase as a new therapeutic target for HER2-positive gastric cancer.
- Author
-
Castagnoli L, Corso S, Franceschini A, Raimondi A, Bellomo SE, Dugo M, Morano F, Prisciandaro M, Brich S, Belfiore A, Vingiani A, Di Bartolomeo M, Pruneri G, Tagliabue E, Giordano S, Pietrantonio F, and Pupa SM
- Subjects
- Animals, Mice, Trastuzumab pharmacology, Fatty Acid Synthases metabolism, Fatty Acid Synthases therapeutic use, Cell Line, Tumor, Receptor, ErbB-2 metabolism, Stomach Neoplasms pathology
- Abstract
Purpose: Trastuzumab is an HER2-specific agent approved as the gold-standard therapy for advanced HER2-positive (HER2+) gastric cancer (GC), but the high rate and rapid appearance of resistance limit its clinical efficacy, resulting in the need to identify new vulnerabilities. Defining the drivers influencing HER2+ cancer stem cell (CSC) maintenance/survival could represent a clinically useful strategy to counteract tumor growth and therapy resistance. Accumulating evidence show that targeting crucial metabolic hubs, as the fatty acid synthase (FASN), may be clinically relevant., Methods: FASN protein and transcript expression were examined by WB and FACS and by qRT-PCR and GEP analyses, respectively, in trastuzumab-sensitive and trastuzumab-resistant HER2+ GC cell lines cultured in adherent (2D) or gastrosphere promoting (3D) conditions. Molecular data were analyzed in silico in public HER2+ GC datasets. The effectiveness of the FASN inhibitor TVB3166 to overcome anti-HER2 therapy resistance was tested in vitro in gastrospheres forming efficiency bioassays and in vivo in mice bearing trastuzumab-resistant GC cells., Results: We compared the transcriptome profiles of HER2+ GC cells cultured in 2D versus 3D conditions finding a significant enrichment of FASN in 3D cultures. FASN upregulation significantly correlated with high stemness score and poor prognosis in HER2+ GC cases. TVB3166 treatment significantly decreased GCSCs in all cell targets. HER2 and FASN cotargeting significantly decreased the capability to form gastrospheres versus monotherapy and reduced the in vivo growth of trastuzumab-resistant GC cells., Conclusion: Our findings indicate that cotargeting HER2 and FASN increase the benefit of anti-HER2 therapy representing a new opportunity for metabolically combating trastuzumab-resistant HER2+ GC., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. HER2 Copy Number and Resistance Mechanisms in Patients with HER2-positive Advanced Gastric Cancer Receiving Initial Trastuzumab-based Therapy in JACOB Trial.
- Author
-
Pietrantonio F, Manca P, Bellomo SE, Corso S, Raimondi A, Berrino E, Morano F, Migliore C, Niger M, Castagnoli L, Pupa SM, Marchiò C, Di Bartolomeo M, Restuccia E, Lambertini C, Tabernero J, and Giordano S
- Subjects
- Humans, Female, Trastuzumab therapeutic use, Trastuzumab metabolism, Receptor, ErbB-2 metabolism, DNA Copy Number Variations, Proto-Oncogene Proteins p21(ras) genetics, Antineoplastic Combined Chemotherapy Protocols adverse effects, Stomach Neoplasms drug therapy, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Breast Neoplasms drug therapy
- Abstract
Purpose: In JACOB trial, pertuzumab added to trastuzumab-chemotherapy did not significantly improve survival of patients with HER2-positive metastatic gastric cancer, despite 3.3 months increase versus placebo. HER2 copy-number variation (CNV) and AMNESIA panel encompassing primary resistance alterations (KRAS/PIK3CA/MET mutations, KRAS/EGFR/MET amplifications) may improve patients' selection for HER2 inhibition., Experimental Design: In a post hoc analysis of JACOB on 327 samples successfully sequenced by next-generation sequencing (NGS; Oncomine Focus DNA), HER2 CNV, HER2 expression by IHC, and AMNESIA were correlated with overall response rate (ORR), progression-free survival (PFS), and overall survival (OS) by univariable/multivariable models., Results: Median HER2 CNV was 4.7 (interquartile range, 2.2-16.9). HER2 CNV-high versus low using the median as cutoff was associated with longer median PFS (10.5 vs. 6.4 months; HR = 0.48; 95% confidence interval: 0.38-0.62; P < 0.001) and OS (20.3 vs. 13.0 months; HR = 0.54; 0.42-0.72; P < 0.001). Combining HER2 CNV and IHC improved discriminative ability, with better outcomes restricted to HER2-high/HER2 3+ subgroup. AMNESIA positivity was found in 51 (16%), with unadjusted HR = 1.35 (0.98-1.86) for PFS; 1.43 (1.00-2.03) for OS.In multivariable models, only HER2 CNV status remained significant for PFS (P < 0.001) and OS (P = 0.004). Higher ORR was significantly associated with IHC 3+ [61% vs. 34% in 2+; OR = 3.11 (1.89-5.17)] and HER2-high [59% vs. 43% in HER2-low; OR = 1.84 (1.16-2.94)], with highest OR in the top CNV quartile. These biomarkers were not associated with treatment effect of pertuzumab., Conclusions: HER2 CNV-high assessed by NGS may be associated with better ORR, PFS, and OS in a JACOB subgroup, especially if combined with HER2 3+. The negative prognostic role of AMNESIA requires further clinical validation., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
9. Predictive Role of CD36 Expression in HER2-Positive Breast Cancer Patients Receiving Neoadjuvant Trastuzumab.
- Author
-
Ligorio F, Di Cosimo S, Verderio P, Ciniselli CM, Pizzamiglio S, Castagnoli L, Dugo M, Galbardi B, Salgado R, Loi S, Michiels S, Triulzi T, Tagliabue E, El-Abed S, Izquierdo M, de Azambuja E, Nuciforo P, Huober J, Moscetti L, Janni W, Coccia-Portugal MA, Corsetto PA, Belfiore A, Lorenzini D, Daidone MG, Vingiani A, Gianni L, Pupa SM, Bianchini G, Pruneri G, and Vernieri C
- Subjects
- Humans, Female, Trastuzumab, Receptor, ErbB-2 metabolism, Chemotherapy, Adjuvant, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Lapatinib, Treatment Outcome, Neoadjuvant Therapy, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: Despite huge efforts to identify biomarkers associated with long-term clinical outcomes in patients with early-stage HER2-positive breast cancer (HER2+ BC) treated with (neo)adjuvant anti-HER2 therapy, no reliable predictors have been identified so far. Fatty acid uptake, a process mediated by the transmembrane transporter CD36, has recently emerged as a potential determinant of resistance to anti-HER2 treatments in preclinical HER2+ BC models., Methods: Here, we investigated the association between baseline intratumor CD36 gene expression and event-free survival in 180 patients enrolled in the phase III trial Neoadjuvant Lapatinib and/or Trastuzumab Treatment Optimization (NeoALTTO), which randomly assigned stage II-III HER2+ BC patients to receive neoadjuvant lapatinib, trastuzumab, or lapatinib-trastuzumab in combination with chemotherapy. To this aim, we selected NeoALTTO trial patients for whom pretreatment whole transcriptomic data were available. The main study results were validated in an independent cohort of patients enrolled in the neoadjuvant phase II trial NeoSphere., Results: In 180 NeoALTTO patients, high intratumor CD36 expression was independently associated with worse event-free survival in patients treated with trastuzumab-based therapy (hazard ratio [HR] = 1.72, 95% confidence interval [CI] = 1.20 to 2.46), but not with lapatinib-based (HR = 1.02, 95% CI = 0.68 to 1.53) or trastuzumab-lapatinib-based (HR = 1.08, 95% CI = 0.60 to 1.94) therapy. Among 331 NeoSphere patients evaluated, high CD36 expression was independently associated with worse patient disease-free survival in both the whole study cohort (HR = 1.197, 95% CI = 1.002 to 1.428) and patients receiving trastuzumab-based neoadjuvant therapy (HR = 1.282, 95% CI = 1.049 to 1.568)., Conclusions: High CD36 expression predicts worse clinical outcomes in early-stage HER2+ BC treated with trastuzumab-based neoadjuvant therapy., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2022
- Full Text
- View/download PDF
10. BCL6 and the Notch pathway: a signaling axis leading to a novel druggable biotarget in triple negative breast cancer.
- Author
-
De Santis F, Romero-Cordoba SL, Castagnoli L, Volpari T, Faraci S, Fucà G, Tagliabue E, De Braud F, Pupa SM, and Di Nicola M
- Subjects
- Cell Line, Tumor, Cell Proliferation, Humans, Neoplastic Stem Cells metabolism, Proto-Oncogene Proteins c-bcl-6 metabolism, Receptors, Notch, Signal Transduction genetics, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- Abstract
Background: The transcriptional repressor B-cell lymphoma 6 (BCL6) is dysregulated in several neoplasms, but its role in triple negative breast cancer (TNBC), a highly aggressive subtype which lacks effective treatment, is unclear. The presence of intratumoral cancer stem cells (CSCs) is a main cause of tumor relapse. The Notch signaling pathway is crucial for regulating CSC self-renewal and promoting breast cancer (BC) development and resistance to anticancer therapies. Here, we investigated signaling cascades of BCL6 in the CSC compartment of TNBCs, and the mechanisms that govern its activity, mainly through Notch signaling., Methods: Gene expression, somatic copy number alterations and clinical data from the Cancer Genome Atlas and METABRIC were accessed through the Xena and cbioportal browsers. Public transcriptome profiles from TNBC datasets were retrieved from the Gene Expression Omnibus. Mammosphere formation efficiency was calculated after BCL6 knockdown via transient siRNA transfection, stable silencing or pharmacological inhibition. The effects exhibited via BCL6 inhibition in putative TNBC stem-like cells were evaluated by immunofluorescence and qRT-PCR analyses. Chromatin immunoprecipitation experiments were performed to validate a putative BCL6 responsive element located in the first intron of the Numb gene and to define the circuit of corepressors engaged by BCL6 following its inhibition. Immunoprecipitation assays were carried out to investigate a novel interaction at the basis of BCL6 control of CSC activity in TNBC., Results: In silico analyses of benchmarked public datasets revealed a significant enrichment of BCL6 in cancer stemness related pathways, particularly of Notch signaling in TNBC. In vitro stable inhibition of BCL6 significantly reduced tumor cell growth and, accordingly, we found that the mammosphere formation efficiency of BCL6 silenced cells was significantly impaired by pharmacological inhibition of Notch signaling. BCL6 was found to be expressed at significantly higher levels in TNBC mammospheres than in their adherent counterparts, and loss of BCL6 function significantly decreased mammosphere formation with preferential targeting of CD44-positive versus ALDH-positive stem-like cells. Functional interplay between BCL6 and the chromatin remodeling factor EZH2 triggered the BCL6/Notch stemness signaling axis via inhibition of Numb transcription., Conclusions: Our results may be instrumental for the prospective design of combination treatment strategies that selectively target novel TNBC-associated biomarker(s) whose activity is implicated in the regulation of cancer stemness (such as BCL6) and molecules in developmentally conserved signaling pathways (such as Notch) to achieve long-lasting tumor control and improve patient outcomes., (© 2022. Springer Nature Switzerland AG.)
- Published
- 2022
- Full Text
- View/download PDF
11. HER2 Signaling and Breast Cancer Stem Cells: The Bridge behind HER2-Positive Breast Cancer Aggressiveness and Therapy Refractoriness.
- Author
-
Pupa SM, Ligorio F, Cancila V, Franceschini A, Tripodo C, Vernieri C, and Castagnoli L
- Abstract
HER2 overexpression/amplification occurs in 15-20% of breast cancers (BCs) and identifies a highly aggressive BC subtype. Recent clinical progress has increased the cure rates of limited-stage HER2-positive BC and significantly prolonged overall survival in patients with advanced disease; however, drug resistance and tumor recurrence remain major concerns. Therefore, there is an urgent need to increase knowledge regarding HER2 biology and implement available treatments. Cancer stem cells (CSCs) represent a subset of malignant cells capable of unlimited self-renewal and differentiation and are mainly considered to contribute to tumor onset, aggressiveness, metastasis, and treatment resistance. Seminal studies have highlighted the key role of altered HER2 signaling in the maintenance/enrichment of breast CSCs (BCSCs) and elucidated its bidirectional communication with stemness-related pathways, such as the Notch and Wingless/β-catenin cascades. d16HER2, a splice variant of full-length HER2 mRNA, has been identified as one of the most oncogenic HER2 isoform significantly implicated in tumorigenesis, epithelial-mesenchymal transition (EMT)/stemness and the response to targeted therapy. In addition, expression of a heterogeneous collection of HER2 truncated carboxy-terminal fragments (CTFs), collectively known as p95HER2, identifies a peculiar subgroup of HER2-positive BC with poor prognosis, with the p95HER2 variants being able to regulate CSC features. This review provides a comprehensive overview of the current evidence regarding HER2-/d16HER2-/p95HER2-positive BCSCs in the context of the signaling pathways governing their properties and describes the future prospects for targeting these components to achieve long-lasting tumor control.
- Published
- 2021
- Full Text
- View/download PDF
12. Targeting lipid metabolism is an emerging strategy to enhance the efficacy of anti-HER2 therapies in HER2-positive breast cancer.
- Author
-
Ligorio F, Pellegrini I, Castagnoli L, Vingiani A, Lobefaro R, Zattarin E, Santamaria M, Pupa SM, Pruneri G, de Braud F, and Vernieri C
- Subjects
- Breast Neoplasms pathology, Female, Humans, Protein Kinase Inhibitors pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Lipid Metabolism genetics, Protein Kinase Inhibitors therapeutic use, Receptor, ErbB-2 metabolism
- Abstract
De novo or acquired resistance of cancer cells to currently available Human Epidermal Growth Factor Receptor 2 (HER2) inhibitors represents a clinical challenge. Several resistance mechanisms have been identified in recent years, with lipid metabolism reprogramming, a well-established hallmark of cancer, representing the last frontier of preclinical and clinical research in this field. Fatty Acid Synthase (FASN), the key enzyme required for fatty acids (FAs) biosynthesis, is frequently overexpressed/activated in HER2-positive (HER2+) breast cancer (BC), and it crucially sustains HER2+ BC cell growth, proliferation and survival. After the synthesis of new, selective and well tolerated FASN inhibitors, clinical trials have been initiated to test if these compounds are able to re-sensitize cancer cells with acquired resistance to HER2 inhibition. More recently, the upregulation of FA uptake by cancer cells has emerged as a potentially new and targetable mechanism of resistance to anti-HER2 therapies in HER2+ BC, thus opening a new era in the field of targeting metabolic reprogramming in clinical setting. Here, we review the available preclinical and clinical evidence supporting the inhibition of FA biosynthesis and uptake in combination with anti-HER2 therapies in patients with HER2+ BC, and we discuss ongoing clinical trials that are investigating these combination approaches., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
13. T Cells Expressing Receptor Recombination/Revision Machinery Are Detected in the Tumor Microenvironment and Expanded in Genomically Over-unstable Models.
- Author
-
Morello G, Cancila V, La Rosa M, Germano G, Lecis D, Amodio V, Zanardi F, Iannelli F, Greco D, La Paglia L, Fiannaca A, Urso AM, Graziano G, Ferrari F, Pupa SM, Sangaletti S, Chiodoni C, Pruneri G, Bardelli A, Colombo MP, and Tripodo C
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Breast immunology, Breast pathology, Breast Neoplasms genetics, Breast Neoplasms pathology, CD8-Positive T-Lymphocytes metabolism, DNA Damage immunology, DNA Nucleotidylexotransferase genetics, DNA Nucleotidylexotransferase metabolism, DNA-Binding Proteins metabolism, Datasets as Topic, Disease Models, Animal, Female, Homeodomain Proteins metabolism, Humans, Lymphocytes, Tumor-Infiltrating metabolism, Mice, Mice, Knockout, Middle Aged, MutL Protein Homolog 1 genetics, MutL Protein Homolog 1 metabolism, Nuclear Proteins metabolism, RNA-Seq, Receptors, Antigen, T-Cell, Single-Cell Analysis, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Breast Neoplasms immunology, CD8-Positive T-Lymphocytes immunology, Lymphocytes, Tumor-Infiltrating immunology, Recombination, Genetic immunology, T-Cell Antigen Receptor Specificity genetics
- Abstract
Tumors undergo dynamic immunoediting as part of a process that balances immunologic sensing of emerging neoantigens and evasion from immune responses. Tumor-infiltrating lymphocytes (TIL) comprise heterogeneous subsets of peripheral T cells characterized by diverse functional differentiation states and dependence on T-cell receptor (TCR) specificity gained through recombination events during their development. We hypothesized that within the tumor microenvironment (TME), an antigenic milieu and immunologic interface, tumor-infiltrating peripheral T cells could reexpress key elements of the TCR recombination machinery, namely, Rag1 and Rag2 recombinases and Tdt polymerase, as a potential mechanism involved in the revision of TCR specificity. Using two syngeneic invasive breast cancer transplantable models, 4T1 and TS/A, we observed that Rag1, Rag2 , and Dntt in situ mRNA expression characterized rare tumor-infiltrating T cells. In situ expression of the transcripts was increased in coisogenic Mlh1 -deficient tumors, characterized by genomic overinstability, and was also modulated by PD-1 immune-checkpoint blockade. Through immunolocalization and mRNA hybridization analyses, we detected the presence of rare TDT
+ RAG1/2+ cells populating primary tumors and draining lymph nodes in human invasive breast cancer. Analysis of harmonized single-cell RNA-sequencing data sets of human cancers identified a very small fraction of tumor-associated T cells, characterized by the expression of recombination/revision machinery transcripts, which on pseudotemporal ordering corresponded to differentiated effector T cells. We offer thought-provoking evidence of a TIL microniche marked by rare transcripts involved in TCR shaping., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
14. Fifteen-year follow-up of relapsed indolent non-Hodgkin lymphoma patients vaccinated with tumor-loaded dendritic cells.
- Author
-
Fucà G, Ambrosini M, Agnelli L, Brich S, Sgambelluri F, Mortarini R, Pupa SM, Magni M, Devizzi L, Matteucci P, Cabras A, Zappasodi R, De Santis F, Anichini A, De Braud F, Gianni AM, and Di Nicola M
- Subjects
- Cancer Vaccines pharmacology, Female, Follow-Up Studies, Humans, Male, Neoplasm Recurrence, Local, Recurrence, Time Factors, Cancer Vaccines therapeutic use, Dendritic Cells transplantation, Immunotherapy methods, Lymphoma, Non-Hodgkin therapy
- Abstract
We previously published the results of a pilot study showing that vaccination with tumor-loaded dendritic cells (DCs) induced both T and B cell response and produced clinical benefit in the absence of toxicity in patients with relapsed, indolent non-Hodgkin lymphoma (iNHL). The purpose of the present short report is to provide a 15-year follow-up of our study and to expand the biomarker analysis previously performed. The long-term follow-up highlighted the absence of particular or delayed toxicity and the benefit of active immunization with DCs loaded with autologous, heat-shocked and UV-C treated tumor cells in relapsed iNHL (5-year and 10-year progression-free survival (PFS) rates: 55.6% and 33.3%, respectively; 10-year overall survival (OS) rate: 83.3%). Female patients experienced a better PFS (p=0.016) and a trend towards a better OS (p=0.185) compared with male patients. Of note, we observed a non-negligible fraction of patients (22%) who experienced a long-lasting complete response. In a targeted gene expression profiling of pre-treatment tumor biopsies in 11 patients with available formalin-fixed, paraffin-embedded tissue, we observed that KIT , ATG12 , TNFRSF10C , PBK , ITGA2 , GATA3 , CLU , NCAM1 , SYT17 and LTK were differentially expressed in patients with responder versus non-responder tumors. The characterization of peripheral monocytic cells in a subgroup of 14 patients with available baseline blood samples showed a higher frequency of the subset of CD14
++ CD16+ cells (intermediate monocytes) in patients with responding tumors. Since in patients with relapsed iNHL the available therapeutic options are often incapable of inducing a long-lasting complete remission and can be sometimes characterized by intolerable toxicity, we think that the encouraging results of our long-term follow-up analysis represent a stimulus to further investigate the role of active vaccination in this specific setting and in earlier lines of therapy and to explore novel combinatorial strategies encompassing other innovative immunotherapy agents, such as immune-checkpoint inhibitors., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
15. A combination of extracellular matrix- and interferon-associated signatures identifies high-grade breast cancers with poor prognosis.
- Author
-
Lecchi M, Verderio P, Cappelletti V, De Santis F, Paolini B, Monica M, Sangaletti S, Pupa SM, Iorio MV, Bianchi G, Gennaro M, Fucà G, De Braud F, Tagliabue E, and Di Nicola M
- Subjects
- Adult, Aged, Biomarkers, Tumor genetics, Breast Neoplasms genetics, Breast Neoplasms pathology, Cohort Studies, Diagnosis, Differential, Extracellular Matrix metabolism, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Interferons metabolism, Middle Aged, Neoplasm Grading, Prognosis, Breast Neoplasms diagnosis, Extracellular Matrix genetics, Interferons genetics, Transcriptome
- Abstract
Breast cancer (BC) is a heterogeneous disease in which the tumor microenvironment (TME) seems to impact the clinical outcome. Here, we investigated whether a combination of gene expression signatures relating to both the structural and immune TME aspects can help predict prognosis in women with high-grade BC (HGBC). Thus, we focused on a combined molecular biomarker variable that involved extracellular matrix (ECM)-associated gene expression (ECM3 signature) and interferon (IFN)-associated metagene (IFN metagene) expression. In 97 chemo-naive HGBCs from the METABRIC dataset, the dichotomous ECM3/IFN (dECIF) variable identified a group of high-risk patients (ECM3
+ /IFN- vs other; hazard ratio = 3.2, 95% confidence interval: 1.5-6.7). Notably, ECM3+ /IFN- tumors showed low tumor-infiltrating lymphocytes, high levels of CD33-positive cells, absence of PD-1 expression, or low expression of PD-L1, as suggested by immune profiles and immune-histochemical analysis on an independent cohort of 131 HGBCs. To make our results transferable to clinical use, we refined the dECIF biomarker using reduced ECM3 and IFN signatures; notably, the prognostic value of this reduced dECIF was comparable to that of the original dECIF. After validation in a new BC cohort, reduced dECIF was translated into a robust qPCR classifier for real-world clinical use., (© 2021 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2021
- Full Text
- View/download PDF
16. Hormone receptor status influences the impact of body mass index and hyperglycemia on the risk of tumor relapse in early-stage HER2-positive breast cancer patients.
- Author
-
Ligorio F, Zambelli L, Bottiglieri A, Castagnoli L, Zattarin E, Lobefaro R, Ottini A, Vingiani A, Pupa SM, Bianchi GV, Capri G, Pruneri G, de Braud F, and Vernieri C
- Abstract
Background: High body mass index (BMI) has been associated with worse clinical outcomes in patients with early-stage breast cancer (BC), and its negative effects could be mediated by hyperglycemia/diabetes. However, the prognostic impact of high BMI in early-stage HER2-positive (HER2+) BC patients remains controversial., Methods: We conducted a retrospective study to investigate the impact of baseline BMI or glycemia on relapse-free survival (RFS) and overall survival (OS) in patients with surgically resected, stage I-III HER2+ BC treated with standard-of-care, trastuzumab-containing adjuvant biochemotherapy. The optimal BMI and glycemia cut-off values for RFS were identified through maximally selected rank statistics. Cox regression models were used to assess the impact of BMI, glycemia and other relevant variables on clinical outcomes., Results: Among 505 patients included in the study, a BMI cut-off of 27.77 kg/m
2 was identified as the best threshold to discriminate between patients with low BMI ( n = 390; 77.2%) or high BMI ( n = 115; 22.8%). At multivariable analysis, higher BMI was associated with significantly worse RFS [hazard ratio 2.26; 95% confidence interval (CI): 1.08-4.74, p = 0.031] and worse OS (hazard ratio 2.25, 95% CI 1.03-4.94, p = 0.043) in the whole patient population. The negative impact of high BMI was only observed in patients with hormone receptor (HR)-negative/HER2+ BC (hazard ratio 2.29; 95% CI: 1.01-5.20; p = 0.047), but not in patients with HR-positive (HR+)/HER2+ BC (hazard ratio 1.36; 95% CI: 0.61-3.07, p = 0.452). By contrast, hyperglycemia (⩾109 mg/dl) at baseline was associated with a trend toward significantly worse RFS at multivariable analysis only in patients with HR+/HER2+ BC (hazard ratio 2.52; 95% CI: 0.89-7.1; p = 0.080)., Conclusions: High BMI is associated with worse clinical outcomes in early-stage HR-/HER2+ BC patients treated with trastuzumab-containing adjuvant biochemotherapy, while baseline hyperglycemia could be a predictor of worse RFS in HR+/HER2+ BC patients. Prospective studies are needed to investigate if modifying patient BMI/glycemia during treatment can improve clinical outcomes., Competing Interests: Conflict of interest statement: The authors declare that there is no conflict of interest., (© The Author(s), 2021.)- Published
- 2021
- Full Text
- View/download PDF
17. Inhibition of the Wnt Signalling Pathway: An Avenue to Control Breast Cancer Aggressiveness.
- Author
-
Castagnoli L, Tagliabue E, and Pupa SM
- Subjects
- Breast Neoplasms genetics, Clinical Trials as Topic, Drug Resistance, Neoplasm genetics, Female, Humans, Models, Biological, Neoplasm Invasiveness, Breast Neoplasms metabolism, Breast Neoplasms pathology, Wnt Signaling Pathway genetics
- Abstract
Breast cancer (BC) is the most common tumour in women. Although the introduction of novel therapeutic approaches in clinical practice has dramatically improved the clinical outcome of BC patients, this malignant disease remains the second leading cause of cancer-related death worldwide. The wingless/integrated (Wnt) signalling pathway represents a crucial molecular node relevantly implicated in the regulation of normal somatic stem cells as well as cancer stem cell (CSC) traits and the epithelial-mesenchymal transition cell program. Accordingly, Wnt signalling is heavily dysregulated in BC, and the altered expression of different Wnt genes is significantly associated with cancer-related aggressive behaviours. For all these reasons, Wnt signalling represents a promising therapeutic target currently under clinical investigation to achieve cancer eradication by eliminating CSCs, considered by most to be responsible for tumour initiation, relapse, and drug resistance. In this review, we summarized the current knowledge on the Wnt signalling pathway in BC and have presented evidence implicating the suitability of Wnt targeting in an attempt to improve the outcome of patients without affecting the normal somatic stem cell population.
- Published
- 2020
- Full Text
- View/download PDF
18. Author Correction: The landscape of d16HER2 splice variant expression across HER2-positive cancers.
- Author
-
Volpi CC, Pietrantonio F, Gloghini A, Fucà G, Giordano S, Corso S, Pruneri G, Antista M, Cremolini C, Fasano E, Saggio S, Faraci S, Di Bartolomeo M, de Braud F, Di Nicola M, Tagliabue E, Pupa SM, and Castagnoli L
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
19. Cancer Stem Cells: Devil or Savior-Looking behind the Scenes of Immunotherapy Failure.
- Author
-
Castagnoli L, De Santis F, Volpari T, Vernieri C, Tagliabue E, Di Nicola M, and Pupa SM
- Subjects
- Epigenesis, Genetic, Humans, Immunomodulation, Immunosuppression Therapy, Treatment Failure, Immunotherapy, Neoplastic Stem Cells pathology
- Abstract
Although the introduction of immunotherapy has tremendously improved the prognosis of patients with metastatic cancers of different histological origins, some tumors fail to respond or develop resistance. Broadening the clinical efficacy of currently available immunotherapy strategies requires an improved understanding of the biological mechanisms underlying cancer immune escape. Globally, tumor cells evade immune attack using two main strategies: avoiding recognition by immune cells and instigating an immunosuppressive tumor microenvironment. Emerging data suggest that the clinical efficacy of chemotherapy or molecularly targeted therapy is related to the ability of these therapies to target cancer stem cells (CSCs). However, little is known about the role of CSCs in mediating tumor resistance to immunotherapy. Due to their immunomodulating features and plasticity, CSCs can be especially proficient at evading immune surveillance, thus potentially representing the most prominent malignant cell component implicated in primary or acquired resistance to immunotherapy. The identification of immunomodulatory properties of CSCs that include mechanisms that regulate their interactions with immune cells, such as bidirectional release of particular cytokines/chemokines, fusion of CSCs with fusogenic stromal cells, and cell-to-cell communication exerted by extracellular vesicles, may significantly improve the efficacy of current immunotherapy strategies. The purpose of this review is to discuss the current scientific evidence linking CSC biological, immunological, and epigenetic features to tumor resistance to immunotherapy., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2020
- Full Text
- View/download PDF
20. Anticancer innovative therapy: Highlights from the ninth annual meeting.
- Author
-
Volpari T, De Santis F, Bracken AP, Pupa SM, Buschbeck M, Wegner A, Di Cosimo S, Lisanti MP, Dotti G, Massaia M, Pruneri G, Anichini A, Fortunato O, De Braud F, Del Vecchio M, and Di Nicola M
- Subjects
- Animals, Biomarkers, Tumor, Congresses as Topic, Epigenesis, Genetic, Humans, Italy, Mice, Mutation, Neoplastic Stem Cells, Antineoplastic Agents therapeutic use, Immunotherapy, Neoplasms therapy, Therapies, Investigational
- Abstract
The Ninth Annual Conference of "Anticancer Innovative Therapy", organized by Fondazione IRCCS Istituto Nazionale dei Tumori di Milano (Fondazione IRCCS INT) and hosted by Hotel Michelangelo, was held in Milan on 25 January 2019. Cutting-edge science was presented in two main scientific sessions: i) pre-clinical evidences and new targets, and ii) clinical translation. The Keynote lecture entitled "Cancer stem cells (CSCs): metabolic strategies for their identification and eradication" presented by M. Lisanti, was one of the highlights of the conference. One key concept of the meeting was how the continuous advances in our knowledge about molecular mechanisms in various fields of research (cancer metabolism reprogramming, epigenetic regulation, transformation/invasiveness, and immunology, among others) are driving cancer research towards more effective personalized antineoplastic strategies. Specifically, recent preclinical data on the following topics were discussed: 1. Polycomb group proteins in cancer; 2. A d16HER2 splice variant is a flag of HER2 addiction across HER2-positive cancers; 3. Studying chromatin as a nexus between translational and basic research; 4. Metabolomic analysis in cancer patients; 5. CDK4-6 cyclin inhibitors: clinical activity and future perspectives as immunotherapy adjuvant; and 6. Cancer stem cells (CSCs): metabolic strategies for their identification and eradication. In terms of clinical translation, several novel approaches were presented: 1. Developing CAR-T cell therapies: an update of preclinical and clinical development at University of North Carolina; 2. Vγ9Vδ2 T-cell activation and immune suppression in multiple myeloma; 3. Predictive biomarkers for real-world immunotherapy: the cancer immunogram model in the clinical arena; and 4. Mechanisms of resistance to immune checkpoint blockade in solid tumors. Overall, the pre-clinical and clinical findings presented could pave the way to identify novel actionable therapeutic targets to significantly enhance the care of persons with cancer., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
21. Phenethyl isothiocyanate hampers growth and progression of HER2-positive breast and ovarian carcinoma by targeting their stem cell compartment.
- Author
-
Koschorke A, Faraci S, Giani D, Chiodoni C, Iorio E, Canese R, Colombo MP, Lamolinara A, Iezzi M, Ladomery M, Vernieri C, de Braud F, Di Nicola M, Tagliabue E, Castagnoli L, and Pupa SM
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Female, Humans, Mice, Transgenic, Neoplasm Proteins metabolism, Neoplastic Stem Cells drug effects, Ovarian Neoplasms metabolism, Signal Transduction drug effects, Trastuzumab pharmacology, Breast Neoplasms pathology, Isothiocyanates pharmacology, Neoplastic Stem Cells pathology, Ovarian Neoplasms pathology, Receptor, ErbB-2 metabolism
- Abstract
Purpose: Isothiocyanates elicit anticancer effects by targeting cancer stem cells (CSCs). Here, we tested the antitumor activity of phenethyl-isothiocyanate (PEITC), either alone or in combination with trastuzumab, in HER2-positive tumor models., Methods: We assessed the in vitro anticancer activity of PEITC, alone or combined with trastuzumab, in HER2-positive BT474, SKBR3, HCC1954 and SKOV3 cancer cells by measuring their sphere forming efficiency (SFE). The expression of the human/rodent CSC biomarkers aldehyde-dehydrogenase (ALDH) and CD29
High /CD24+ /Sca1Low was evaluated by cytofluorimetric analysis. The expression of wild type HER2 (WTHER2), its splice variant d16HER2 and NOTCH was analysed by quantitative RT-PCR and Western blotting. The in vivo activity of PEITC and trastuzumab was evaluated in mice orthotopically implanted with MI6 tumor cells transgenic for the human d16HER2 splice isoform. Magnetic resonance imaging/spectroscopy and immunohistochemistry were used to assess morpho-functional and metabolic profiles of treated versus untreated mice., Results: We found that PEITC significantly impaired the SFE of HER2-positive human cancer cells by decreasing their ALDH-positive compartments. The anti-CSC activity of PEITC was demonstrated by a reduced expression/activation of established cancer-stemness biomarkers. Similar results were obtained with MI6 cells, where PEITC, alone or in combination with trastuzumab, significantly inhibited their SFE. We also found that PEITC hampered the in vivo growth of MI6 nodules by inducing hemorrhagic and necrotic intra-tumor areas and, in combination with trastuzumab, by significantly reducing spontaneous tumor development in d16HER2 transgenic mice., Conclusions: Our results indicate that PEITC targets HER2-positive CSCs and that its combination with trastuzumab may pave the way for a novel therapeutic strategy for HER2-positive tumors.- Published
- 2019
- Full Text
- View/download PDF
22. The d16HER2 Splice Variant: A Friend or Foe of HER2-Positive Cancers?
- Author
-
Castagnoli L, Ladomery M, Tagliabue E, and Pupa SM
- Abstract
Human epidermal growth factor receptor 2 ( ERBB2 or HER2) amplification/overexpression is associated with a particularly aggressive molecular subtype of breast cancer (BC), characterized by a poor prognosis, increased metastatic potential, and disease recurrence. As only approximately 50% of HER2-positive patients respond to HER2-targeted therapies, greater knowledge of the biology of HER2 and the mechanisms that underlie drug susceptibility is needed to improve cure rates. Evidence suggests that the coexistence of full-length, wild-type HER2 (wtHER2) and altered forms of HER2-such as carboxy-terminus-truncated fragments, activating mutations, and splice variants-significantly increases the heterogeneity of HER2-positive disease, affecting its biology, clinical course, and treatment response. In particular, expression of the d16HER2 splice variant in human HER2-positive BC has a crucial pathobiological function, wherein the absence of sixteen amino acids from the extracellular domain induces the formation of stable and constitutively active HER2 homodimers on the tumor cell surface. Notably, the d16HER2 variant significantly influences the initiation and aggressiveness of tumors, cancer stem cell properties, epithelial-mesenchymal transition (EMT), and the susceptibility of HER2-positive BC cells to trastuzumab compared with its wtHER2 counterpart, thus constituting a novel and potentially clinically useful biomarker. The aims of this review are to summarize the existing evidence regarding the pathobiological functions of the d16HER2 variant and discuss its current and future value with regard to risk assessment and treatment choices in HER2-positive disease.
- Published
- 2019
- Full Text
- View/download PDF
23. WNT signaling modulates PD-L1 expression in the stem cell compartment of triple-negative breast cancer.
- Author
-
Castagnoli L, Cancila V, Cordoba-Romero SL, Faraci S, Talarico G, Belmonte B, Iorio MV, Milani M, Volpari T, Chiodoni C, Hidalgo-Miranda A, Tagliabue E, Tripodo C, Sangaletti S, Di Nicola M, and Pupa SM
- Subjects
- Aldehyde Dehydrogenase metabolism, Animals, Biomarkers, Tumor metabolism, Cell Line, Tumor, Down-Regulation physiology, Female, Humans, Hyaluronan Receptors metabolism, Mice, Inbred BALB C, Up-Regulation physiology, B7-H1 Antigen metabolism, Stem Cells metabolism, Triple Negative Breast Neoplasms metabolism, Wnt Signaling Pathway physiology
- Abstract
Triple-negative breast cancers (TNBCs) are characterized by a poor prognosis and lack of targeted treatments, and thus, new therapeutic strategies are urgently needed. Inhibitors against programmed death-1 (PD-1)/PD-1 ligand (PD-L1) have shown significant efficacy in various solid cancers, but their activity against TNBCs remains limited. Here, we report that human TNBCs molecularly stratified for high levels of PD-L1 (PD-L1
High ) showed significantly enriched expression of immune and cancer stemness pathways compared with those with low PD-L1 expression (PD-L1Low ). In addition, the PD-L1High cases were significantly associated with a high stemness score (SSHigh ) signature. TNBC cell lines gated for aldehyde dehydrogenase (ALDH) and CD44 stemness markers exhibited increased levels of PD-L1 versus their ALDH-negative and CD44Low counterparts, and PD-L1High cells generated significantly more mammospheres than PD-L1Low cells. Murine mammary SCA-1-positive tumor cells with PD-L1High expression generated tumors in vivo with higher efficacy than PD-L1Low cells. Furthermore, treatment of TNBC cells with selective WNT inhibitors or activators downregulated or upregulated PD-L1 expression, respectively, implying a functional cross-talk between WNT activity and PD-L1 expression. Remarkably, human TNBC samples contained tumor elements co-expressing PD-L1 with ALDH1A1 and/or CD44v6. Additionally, both PD-L1-/SCA1-positive and ALDH1A1-positive tumor elements were found in close contact with CD3-, and PD-1-positive T cells in murine and human tumor samples. Overall, our study suggests that PD-L1-positive tumor elements with a stemness phenotype may participate in the complex dynamics of TNBC-related immune evasion, which might be targeted through WNT signaling inhibition.- Published
- 2019
- Full Text
- View/download PDF
24. The landscape of d16HER2 splice variant expression across HER2-positive cancers.
- Author
-
Volpi CC, Pietrantonio F, Gloghini A, Fucà G, Giordano S, Corso S, Pruneri G, Antista M, Cremolini C, Fasano E, Saggio S, Faraci S, Di Bartolomeo M, de Braud F, Di Nicola M, Tagliabue E, Pupa SM, and Castagnoli L
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Transformation, Neoplastic, Humans, MCF-7 Cells, Mice, Protein Isoforms genetics, Protein Isoforms metabolism, Breast Neoplasms pathology, Exons genetics, Gene Expression Regulation, Neoplastic, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism
- Abstract
The HER2 splice variant characterized by the deletion of exon 16 and denominated as d16HER2, is associated with HER2-positive breast cancer (BC) aggressiveness, stemness, and trastuzumab susceptibility and is considered to be a "flag" of HER2 dependence. However, with the exception of quantitative real-time PCR analysis, easily reproducible assays are still lacking to clinically detect and quantify the d16HER2 expression. Further, no data on d16HER2 expression and its potential role are available in HER2-positive gastrointestinal malignancies. Here, we used a novel RNA in situ hybridization technique (BaseScope) to discriminate d16HER2 variant expression from the wild type isoform (WTHER2) and to assess their levels across different HER2-positive histological samples. Our results demonstrate the existence of outliers, with d16HER2 mRNA high scores restricted to HER2-positive gastric cancer (GC) and colorectal cancer (CRC) coupled with increased d16HER2 expression compared with BC. Consistent with previously reported data on BC, experiments performed in HER2-positive GC patient-derived xenografts suggest that increased d16HER2 expression is associated with a clinical benefit/response to single-agent trastuzumab. Therefore, d16HER2 may be considered as a "flag" of HER2 dependence in GC and can be clinically investigated as a marker of trastuzumab susceptibility in several other HER2-driven cancers, including CRC. As a clinical proof-of-concept, we indicate that high d16HER2 mRNA scores are exclusively found in patients with a long-term benefit from trastuzumab exceeding 12 months (clinical "outliers"), and that d16HER2 expression is also increased in circulating tumor-released exosomes obtained from baseline plasma samples of long-term responders.
- Published
- 2019
- Full Text
- View/download PDF
25. Intratumor lactate levels reflect HER2 addiction status in HER2-positive breast cancer.
- Author
-
Castagnoli L, Iorio E, Dugo M, Koschorke A, Faraci S, Canese R, Casalini P, Nanni P, Vernieri C, Di Nicola M, Morelli D, Tagliabue E, and Pupa SM
- Subjects
- Animals, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor antagonists & inhibitors, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cell Line, Tumor, Chemotherapy, Adjuvant, Female, Gene Expression Regulation, Neoplastic, Humans, Italy, Lapatinib pharmacology, Magnetic Resonance Spectroscopy, Mice, Patient Selection, Precision Medicine, Protein Kinase Inhibitors therapeutic use, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism, Retrospective Studies, Trastuzumab therapeutic use, Treatment Outcome, Up-Regulation, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, Glycolysis, Lactic Acid metabolism, Oncogene Addiction, Receptor, ErbB-2 genetics
- Abstract
Despite different molecular tumor profiles indicate that human epidermal growth factor receptor 2 (HER2) messenger RNA (mRNA) levels mirror HER2 addiction and trastuzumab benefit in HER2-positive breast cancer (BC), the identification of noninvasive clinical predictors of trastuzumab sensitivity remains an unmet clinical need. In the current study, we investigated whether intratumor lactate levels reflect HER2 addiction and, in turn, trastuzumab susceptibility. Accordingly, the gene expression profiles of transgenic murine BC cell lines expressing the human d16HER2 variant (HER2-addicted) or human full-length HER2 (WTHER2; HER2-nonaddicted) revealed a significant enrichment of glycolysis-related gene pathways in HER2-addicted cells. We studied the metabolic content of 22 human HER2-positive BC by quantitative nuclear magnetic resonance spectroscopy and found that those cases with higher lactate levels were characterized by higher HER2 transcript levels. Moreover, gene expression analyses of HER2-positive BC samples from a TCGA data set revealed a significant enrichment in glycolysis-related pathways in high/HER2-addicted tumors. These data were confirmed by metabolic analyses of human HER2-positive BC cell lines with high or low HER2 transcript levels, which revealed significantly more active glycolytic metabolism in high HER2 transcript than in low HER2 transcript cells. Overall, our results provide evidence for noninvasive intratumor lactate detection as a potential metabolic biomarker of HER2 addiction and trastuzumab response suggesting the possibility to use in vivo imaging to assess lactate levels and, in turn, select HER2-positive BC patients who are more likely to benefit from anti-HER2 treatments., (© 2018 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
26. Innovative therapy, monoclonal antibodies, and beyond: Highlights from the eighth annual meeting.
- Author
-
De Santis F, Del Vecchio M, Castagnoli L, De Braud F, Di Cosimo S, Franceschini D, Fucà G, Hiscott J, Malmberg KJ, McGranahan N, Pietrantonio F, Rivoltini L, Sangaletti S, Tagliabue E, Tripodo C, Vernieri C, Zitvogel L, Pupa SM, and Di Nicola M
- Subjects
- Animals, Gastrointestinal Microbiome, Humans, Neoplasms immunology, Neoplasms microbiology, Antibodies, Monoclonal therapeutic use, Immunotherapy, Neoplasms therapy
- Abstract
The eighth annual conference of "Innovative therapy, monoclonal antibodies, and beyond" was held in Milan on Jan. 26, 2018, and hosted by Fondazione IRCCS-Istituto Nazionale dei Tumori (Fondazione IRCCS INT). The conference was divided into two main scientific sessions, of i) pre-clinical assays and novel biotargets, and ii) clinical translation, as well as a third session of presentations from young investigators, which focused on recent achievements within Fondazione IRCCS INT on immunotherapy and targeted therapies. Presentations in the first session addressed the issue of cancer immunotherapy activity with respect to tumor heterogeneity, with key topics addressing: 1) tumor heterogeneity and targeted therapy, with the definition of the evolutionary Index as an indicator of tumor heterogeneity in both space and time; 2) the analysis of cancer evolution, with the introduction of the TRACERx Consortium-a multi-million pound UK research project focused on non-small cell lung cancer (NSCLC); 3) the use of anti-estrogen agents to boost immune recognition of breast cancer cells; and 4) the high degree of functional plasticity within the NK cell repertoire, including the expansion of adaptive NK cells following viral challenges. The second session addressed: 1) the effectiveness of radiotherapy to enhance the proportion of patients responsive to immune-checkpoint blockers (ICBs); 2) the use of MDSC scores in selecting melanoma patients with high probability to be responsive to ICBs; and 3) the relevance of the gut microbiome as a predictive factor, and the potential of its perturbation in increasing the immune response rate to ICBs. Overall, a picture emerged of tumor heterogeneity as the main limitation that impairs the effectiveness of anti-cancer therapies. Thus, the choice of a specific therapy based on reproducible and selective predictive biomarkers is an urgent unmet clinical need that should be addressed in order to increase the proportion of long-term responding patients and to improve the sustainability of novel drugs., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. RET fusions in a small subset of advanced colorectal cancers at risk of being neglected.
- Author
-
Pietrantonio F, Di Nicolantonio F, Schrock AB, Lee J, Morano F, Fucà G, Nikolinakos P, Drilon A, Hechtman JF, Christiansen J, Gowen K, Frampton GM, Gasparini P, Rossini D, Gigliotti C, Kim ST, Prisciandaro M, Hodgson J, Zaniboni A, Chiu VK, Milione M, Patel R, Miller V, Bardelli A, Novara L, Wang L, Pupa SM, Sozzi G, Ross J, Di Bartolomeo M, Bertotti A, Ali S, Trusolino L, Falcone A, de Braud F, and Cremolini C
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Metastasis, Prognosis, Survival Rate, Young Adult, Biomarkers, Tumor genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Gene Rearrangement, Oncogene Proteins, Fusion genetics, Proto-Oncogene Proteins c-ret genetics
- Abstract
Background: Recognition of rare molecular subgroups is a challenge for precision oncology and may lead to tissue-agnostic approval of targeted agents. Here we aimed to comprehensively characterize the clinical, pathological and molecular landscape of RET rearranged metastatic colorectal cancer (mCRC)., Patients and Methods: In this case series, we compared clinical, pathological and molecular characteristics of 24 RET rearranged mCRC patients with those of a control group of 291 patients with RET negative tumors. RET rearranged and RET negative mCRCs were retrieved by systematic literature review and by taking advantage of three screening sources: (i) Ignyta's phase 1/1b study on RXDX-105 (NCT01877811), (ii) cohorts screened at two Italian and one South Korean Institutions and (iii) Foundation Medicine Inc. database. Next-generation sequencing data were analyzed for RET rearranged cases., Results: RET fusions were more frequent in older patients (median age of 66 versus 60 years, P = 0.052), with ECOG PS 1-2 (90% versus 50%, P = 0.02), right-sided (55% versus 32%, P = 0.013), previously unresected primary tumors (58% versus 21%, P < 0.001), RAS and BRAF wild-type (100% versus 40%, P < 0.001) and MSI-high (48% versus 7%, P < 0.001). Notably, 11 (26%) out of 43 patients with right-sided, RAS and BRAF wild-type tumors harbored a RET rearrangement. At a median follow-up of 45.8 months, patients with RET fusion-positive tumors showed a significantly worse OS when compared with RET-negative ones (median OS 14.0 versus 38.0 months, HR: 4.59; 95% CI, 3.64-32.66; P < 0.001). In the multivariable model, RET rearrangements were still associated with shorter OS (HR: 2.97; 95% CI, 1.25-7.07; P = 0.014), while primary tumor location, RAS and BRAF mutations and MSI status were not., Conclusions: Though very rare, RET rearrangements define a new subtype of mCRC that shows poor prognosis with conventional treatments and is therefore worth of a specific management.
- Published
- 2018
- Full Text
- View/download PDF
28. Biomarkers of Primary Resistance to Trastuzumab in HER2-Positive Metastatic Gastric Cancer Patients: the AMNESIA Case-Control Study.
- Author
-
Pietrantonio F, Fucà G, Morano F, Gloghini A, Corso S, Aprile G, Perrone F, De Vita F, Tamborini E, Tomasello G, Gualeni AV, Ongaro E, Busico A, Giommoni E, Volpi CC, Laterza MM, Corallo S, Prisciandaro M, Antista M, Pellegrinelli A, Castagnoli L, Pupa SM, Pruneri G, de Braud F, Giordano S, Cremolini C, and Di Bartolomeo M
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents, Immunological therapeutic use, Case-Control Studies, Female, Humans, Male, Middle Aged, Mutation, Patient Selection, Precision Medicine methods, Progression-Free Survival, Prospective Studies, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism, Response Evaluation Criteria in Solid Tumors, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Survival Analysis, Trastuzumab pharmacology, Trastuzumab therapeutic use, Antineoplastic Agents, Immunological pharmacology, Biomarkers, Tumor genetics, Drug Resistance, Neoplasm genetics, Stomach Neoplasms drug therapy, Trastuzumab pharmacokinetics
- Abstract
Purpose: Refining the selection of HER2-positive metastatic gastric cancer patient candidates for trastuzumab is a challenge of precision oncology. Preclinical studies have suggested several genomic mechanisms of primary resistance, leading to activation of tyrosine kinase receptors other than HER2 or downstream signaling pathways. Experimental Design: We carried out this multicenter, prospective, case-control study to demonstrate the negative predictive impact of a panel of candidate genomic alterations (AMNESIA panel), including EGFR/MET/KRAS/PI3K/PTEN mutations and EGFR/MET/KRAS amplifications. Hypothesizing a prevalence of candidate alterations of 30% and 0% in resistant and sensitive HER2-positive patients, respectively, 20 patients per group were needed. Results: AMNESIA panel alterations were significantly more frequent in resistant (11 of 20, 55%) as compared with sensitive (0% of 17) patients ( P < 0.001), and in HER2 IHC 2
+ (7 of 13, 53.8%) than 3+ (4 of 24, 16.7%) tumors ( P = 0.028). Patients with tumors bearing no candidate alterations had a significantly longer median progression-free [5.2 vs. 2.6 months; HR, 0.34; 95% confidence interval (CI), 0.07-0.48; P = 0.001] and overall survival (16.1 vs. 7.6 months; HR, 0.38; 95% CI, 0.09-0.75; P = 0.015). The predictive accuracy of the AMNESIA panel and HER2 IHC was 76% and 65%, respectively. The predictive accuracy of the combined evaluation of the AMNESIA panel and HER2 IHC was 84%. Conclusions: Our panel of candidate genomic alterations may be clinically useful to predict primary resistance to trastuzumab in patients with HER2-positive metastatic gastric cancer and should be further validated with the aim of molecularly stratifying HER2-addicted cancers for the development of novel treatment strategies. Clin Cancer Res; 24(5); 1082-9. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
29. Innovative Therapy, Monoclonal Antibodies and Beyond.
- Author
-
Di Nicola M, Apetoh L, Bellone M, Colombo MP, Dotti G, Ferrone S, Muscolini M, Hiscott J, Anichini A, Pupa SM, Braud F, and Del Vecchio M
- Subjects
- Animals, Humans, Neoplasms drug therapy, Antibodies, Monoclonal therapeutic use, Immunotherapy, Neoplasms therapy
- Abstract
The seventh Edition of "Innovative Therapy, Monoclonal Antibodies and Beyond" Meeting took place in Milan, Italy, on January 27, 2017. The two sessions of the meeting were focused on: 1) Preclinical assays and novel biotargets; and 2) monoclonal antibodies, cell therapies and targeted molecules. Between these two sessions, a lecture entitled "HLA-antigens modulation and response to immune checkpoint inhibitor immunotherapy" was also presented. Despite the impressive successes in cancer immunotherapy in recent years, the response to immune based interventions occurs only in a minority of patients (∼20%). Several basic and translational mechanisms of resistance to immune checkpoint blockers (ICBs) were discussed during the meeting: 1. the impact of tumor microenvironment on the activity of immune system; 2. strategies to inhibit the cross-talk between extracellular matrix and myeloid-derived suppressor cells (MDSC) in the preclinical setting; 3. microRNA expression as a biomarker and as a target of therapy in non-small cell lung cancer (NSCLC); 4. the significance of complement activation pathways in response to immune checkpoint inhibitors; 5. the immunosuppressive activity of the microbiota by inducing IL-17 producing cells; and 6. modulation of HLA antigens as possible markers of response to ICB therapy. In order to overcome the deficiency in active anti-tumor T cells, several clinically applicable combination strategies were also discussed: 1. strategies to enhance the anticancer effects of immunogenic cell death inducing-chemotherapy; 2. the use of CAR T-cells in solid tumors; 3. the use of combination strategies involving oncolytic viruses and ICBs; 4. combinations of new ICBs with anti-PD-1/CTLA-4 therapy; and 4. combinations of targeted therapies and ICBs in melanoma. Overall, this conference emphasized the many novel strategies that are being investigated to improve the overall patient response to cancer immunotherapy. Optimization of biomarkers to accurately select patients who will respond to immunotherapy, coupled with combination strategies to improve long term patient survival remain critical challenges in the immuno-oncology field., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. HER2 isoforms co-expression differently tunes mammary tumor phenotypes affecting onset, vasculature and therapeutic response.
- Author
-
Palladini A, Nicoletti G, Lamolinara A, Dall'Ora M, Balboni T, Ianzano ML, Laranga R, Landuzzi L, Giusti V, Ceccarelli C, Santini D, Taffurelli M, Di Oto E, Asioli S, Amici A, Pupa SM, De Giovanni C, Tagliabue E, Iezzi M, Nanni P, and Lollini PL
- Abstract
Full-length HER2 oncoprotein and splice variant Delta16 are co-expressed in human breast cancer. We studied their interaction in hybrid transgenic mice bearing human full-length HER2 and Delta16 (F1 HER2/Delta16) in comparison to parental HER2 and Delta16 transgenic mice. Mammary carcinomas onset was faster in F1 HER2/Delta16 and Delta16 than in HER2 mice, however tumor growth was slower, and metastatic spread was comparable in all transgenic mice. Full-length HER2 tumors contained few large vessels or vascular lacunae , whereas Delta16 tumors presented a more regular vascularization with numerous endothelium-lined small vessels. Delta16-expressing tumors showed a higher accumulation of i.v . injected doxorubicin than tumors expressing full-length HER2. F1 HER2/Delta16 tumors with high full-length HER2 expression made few large vessels, whereas tumors with low full-length HER2 and high Delta16 contained numerous small vessels and expressed higher levels of VEGF and VEGFR2. Trastuzumab strongly inhibited tumor onset in F1 HER2/Delta16 and Delta16 mice, but not in full-length HER2 mice. Addiction of F1 tumors to Delta16 was also shown by long-term stability of Delta16 levels during serial transplants, in contrast full-length HER2 levels underwent wide fluctuations. In conclusion, full-length HER2 leads to a faster tumor growth and to an irregular vascularization, whereas Delta16 leads to a faster tumor onset, with more regular vessels, which in turn could better transport cytotoxic drugs within the tumor, and to a higher sensitivity to targeted therapeutic agents. F1 HER2/Delta16 mice are a new immunocompetent mouse model, complementary to patient-derived xenografts, for studies of mammary carcinoma onset, prevention and therapy., Competing Interests: CONFLICTS OF INTEREST The authors disclose no potential conflicts of interest
- Published
- 2017
- Full Text
- View/download PDF
31. Pathobiological implications of the d16HER2 splice variant for stemness and aggressiveness of HER2-positive breast cancer.
- Author
-
Castagnoli L, Ghedini GC, Koschorke A, Triulzi T, Dugo M, Gasparini P, Casalini P, Palladini A, Iezzi M, Lamolinara A, Lollini PL, Nanni P, Chiodoni C, Tagliabue E, and Pupa SM
- Subjects
- Animals, Apoptosis genetics, Biomarkers, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cluster Analysis, Disease Progression, Epithelial-Mesenchymal Transition genetics, Female, Gene Expression, Gene Expression Profiling, Humans, Mice, Mutation, Receptor, ErbB-2 metabolism, Receptors, Notch metabolism, Signal Transduction, Alternative Splicing, Breast Neoplasms genetics, Breast Neoplasms pathology, Neoplastic Stem Cells metabolism, Receptor, ErbB-2 genetics
- Abstract
We have previously shown that the d16HER2 splice variant is linked to HER2-positive breast cancer (BC) tumorigenesis, progression and response to Trastuzumab. However, the mechanisms by which d16HER2 contributes to HER2-driven aggressiveness and targeted therapy susceptibility remain uncertain. Here, we report that the d16HER2-positive mammary tumor cell lines MI6 and MI7, derived from spontaneous lesions of d16HER2 transgenic (tg) mice and resembling the aggressive features of primary lesions, are enriched in the expression of Wnt, Notch and epithelial-mesenchymal transition pathways related genes compared with full-length wild-type (WT) HER2-positive cells (WTHER2_1 and WTHER2_2) derived from spontaneous tumors arising in WTHER2 tg mice. MI6 cells exhibited increased resistance to anoikis and significantly higher mammosphere-forming efficiency (MFE) and self-renewal capability than the WTHER2-positive counterpart. Furthermore, d16HER2-positive tumor cells expressed a higher fraction of CD29
High /CD24+ /SCA1Low cells and displayed greater in vivo tumor engraftment in serial dilution conditions than WTHER2_1 cells. Accordingly, NOTCH inhibitors impaired mammosphere formation only in MI6 cells. A comparative analysis of stemness-related features driven by d16HER2 and WTHER2 in ad hoc engineered human BC cells (MCF7 and T47D) revealed a higher MFE and aldehyde dehydrogenase-positive staining in d16HER2- vs WTHER2-infected cells, sustaining consistent BC-initiating cell enrichment in the human setting. Moreover, marked CD44 expression was found in MCF7_d16 and T47D_d16 cells vs their WTHER2 and Mock counterparts. Clinically, BC cases from two distinct HER2-positive cohorts characterized by high levels of expression of the activated-d16HER2 metagene were significantly enriched in the Notch family and signal transducer genes vs those with low levels of the metagene.- Published
- 2017
- Full Text
- View/download PDF
32. Synergistic Activation upon MET and ALK Coamplification Sustains Targeted Therapy in Sarcomatoid Carcinoma, a Deadly Subtype of Lung Cancer.
- Author
-
Pelosi G, Gasparini P, Conte D, Fabbri A, Perrone F, Tamborini E, Pupa SM, Ciravolo V, Caserini R, Rossi G, Cavazza A, Papotti M, Nakatani Y, Maisonneuve P, Pastorino U, and Sozzi G
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma secondary, Adult, Aged, Aged, 80 and over, Anaplastic Lymphoma Kinase, Biomarkers, Tumor, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell secondary, Carcinosarcoma genetics, Carcinosarcoma metabolism, Carcinosarcoma secondary, Female, Follow-Up Studies, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Lymphatic Metastasis, Male, Middle Aged, Molecular Targeted Therapy, Mutation genetics, Neoplasm Invasiveness, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local metabolism, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Prognosis, Proto-Oncogene Mas, Proto-Oncogene Proteins c-met genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinosarcoma drug therapy, Gene Amplification, Lung Neoplasms drug therapy, Proto-Oncogene Proteins c-met metabolism, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Introduction: Genetic alterations suitable for targeted therapy are poorly known issues in pulmonary sarcomatoid carcinoma (PSC), an uncommon and life-threatening family of non-small cell lung cancers., Methods: Ninety-eight PSCs were assessed for MNNG HOS Transforming gene (MET) and anaplastic lymphoma receptor tyrosine kinase gene (ALK) status by fluorescence in situ hybridization (FISH) and for relevant protein expression by immunohistochemical analysis, also taking advantage of phosphorylated (p-) antibodies. Moreover, levels of ALK and MET mRNA were also determined by real-time polymerase chain reaction and Western blot analysis for downstream activation pathways involving p-MET, p-protein kinase B, p-mitogen-activated protein kinase, p-SRC proto-oncogene tyrosine-protein kinase, and p-focal adhesion kinase (p-FAK)., Results: MET amplification was detected by FISH in 25 of 98 PSCs (25.6%) and ALK amplification (but not the relevant rearrangement) was found in 16 of 98 (16.3%), with all ALK-amplified tumors also showing MET amplification (p < 0.0001). Nine PSCs, however, showed MET amplification without any ALK gene alteration. ALK protein expression was always lacking, whereas MET and p-MET were confined to the relevant amplified tumors only. Increased levels of ALK and MET mRNA were detectable in tumors with no direct relationship between mRNA content, protein expression, or alterations detected by FISH. Western blot assays showed complete activation of downstream signal pathways up to p-SRC proto-oncogene tyrosine-protein kinase, and p-focal adhesion kinase recruitment in MET and ALK-coamplified tumors only, whereas isolated MET amplification, MET and ALK borderline amplification (5%-10% of tumor cells with ≥15 copies of the relevant gene), or negative tumors showing eusomy or chromosome polysomy were confined to p-mitogen-activated protein kinase, p-protein kinase B, and/or p-MET activation. Multivariate survival analysis pushed a higher percentage of MET altered cells or a higher value of MET copy gain per cell to marginally emerge for overall survival (p = 0.140) and disease-free survival (p = 0.060), respectively., Conclusions: ALK and MET seemed to act as synergistic, nonrandom coactivators of downstream signal when coamplified in a subset of patients with PSC, thus likely suggesting a combined mechanism of oncogene addiction. These alterations could be a suitable target for therapy based on specific inhibitors., (Copyright © 2016 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. HSPH1 inhibition downregulates Bcl-6 and c-Myc and hampers the growth of human aggressive B-cell non-Hodgkin lymphoma.
- Author
-
Zappasodi R, Ruggiero G, Guarnotta C, Tortoreto M, Tringali C, Cavanè A, Cabras AD, Castagnoli L, Venerando B, Zaffaroni N, Gianni AM, De Braud F, Tripodo C, Pupa SM, and Di Nicola M
- Subjects
- Animals, Cell Line, Tumor, DNA-Binding Proteins genetics, Down-Regulation, Gene Knockdown Techniques, HSP110 Heat-Shock Proteins genetics, Humans, Lymphoma, B-Cell pathology, Mice, Mice, SCID, Proto-Oncogene Proteins c-bcl-6, Proto-Oncogene Proteins c-myc genetics, Xenograft Model Antitumor Assays, DNA-Binding Proteins metabolism, HSP110 Heat-Shock Proteins antagonists & inhibitors, Lymphoma, B-Cell drug therapy, Lymphoma, B-Cell metabolism, Proto-Oncogene Proteins c-myc metabolism
- Abstract
We have shown that human B-cell non-Hodgkin lymphomas (B-NHLs) express heat shock protein (HSP)H1/105 in function of their aggressiveness. Here, we now clarify its role as a functional B-NHL target by testing the hypothesis that it promotes the stabilization of key lymphoma oncoproteins. HSPH1 silencing in 4 models of aggressive B-NHLs was paralleled by Bcl-6 and c-Myc downregulation. In vitro and in vivo analysis of HSPH1-silenced Namalwa cells showed that this effect was associated with a significant growth delay and the loss of tumorigenicity when 10(4) cells were injected into mice. Interestingly, we found that HSPH1 physically interacts with c-Myc and Bcl-6 in both Namalwa cells and primary aggressive B-NHLs. Accordingly, expression of HSPH1 and either c-Myc or Bcl-6 positively correlated in these diseases. Our study indicates that HSPH1 concurrently favors the expression of 2 key lymphoma oncoproteins, thus confirming its candidacy as a valuable therapeutic target of aggressive B-NHLs., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
34. Activated d16HER2 homodimers and SRC kinase mediate optimal efficacy for trastuzumab.
- Author
-
Castagnoli L, Iezzi M, Ghedini GC, Ciravolo V, Marzano G, Lamolinara A, Zappasodi R, Gasparini P, Campiglio M, Amici A, Chiodoni C, Palladini A, Lollini PL, Triulzi T, Menard S, Nanni P, Tagliabue E, and Pupa SM
- Subjects
- Animals, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Exons genetics, Female, Humans, Mice, Mice, Transgenic, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Protein Multimerization genetics, Signal Transduction genetics, Trastuzumab, Antibodies, Monoclonal, Humanized administration & dosage, Neoplasm Recurrence, Local drug therapy, Protein Isoforms genetics, Receptor, ErbB-2 genetics, src-Family Kinases genetics
- Abstract
A splice isoform of the HER2 receptor that lacks exon 16 (d16HER2) is expressed in many HER2-positive breast tumors, where it has been linked with resistance to the HER2-targeting antibody trastuzumab, but the impact of d16HER2 on tumor pathobiology and therapeutic response remains uncertain. Here, we provide genetic evidence in transgenic mice that expression of d16HER2 is sufficient to accelerate mammary tumorigenesis and improve the response to trastuzumab. A comparative analysis of effector signaling pathways activated by d16HER2 and wild-type HER2 revealed that d16HER2 was optimally functional through a link to SRC activation (pSRC). Clinically, HER2-positive breast cancers from patients who received trastuzumab exhibited a positive correlation in d16HER2 and pSRC abundance, consistent with the mouse genetic results. Moreover, patients expressing high pSRC or an activated "d16HER2 metagene" were found to derive the greatest benefit from trastuzumab treatment. Overall, our results establish the d16HER2 signaling axis as a signature for decreased risk of relapse after trastuzumab treatment., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
35. Identification of relevant conformational epitopes on the HER2 oncoprotein by using Large Fragment Phage Display (LFPD).
- Author
-
Gabrielli F, Salvi R, Garulli C, Kalogris C, Arima S, Tardella L, Monaci P, Pupa SM, Tagliabue E, Montani M, Quaglino E, Stramucci L, Curcio C, Marchini C, and Amici A
- Subjects
- Animals, BALB 3T3 Cells, Breast Neoplasms diagnosis, Breast Neoplasms genetics, Breast Neoplasms immunology, Breast Neoplasms pathology, Female, Humans, Mice, Neoplasm Metastasis, Protein Structure, Tertiary, Rats, Epitopes, B-Lymphocyte chemistry, Epitopes, B-Lymphocyte genetics, Epitopes, B-Lymphocyte immunology, Peptide Library, Receptor, ErbB-2 chemistry, Receptor, ErbB-2 genetics, Receptor, ErbB-2 immunology
- Abstract
We developed a new phage-display based approach, the Large Fragment Phage Display (LFPD), that can be used for mapping conformational epitopes on target molecules of immunological interest. LFPD uses a simplified and more effective phage-display approach in which only a limited set of larger fragments (about 100 aa in length) are expressed on the phage surface. Using the human HER2 oncoprotein as a target, we identified novel B-cell conformational epitopes. The same homologous epitopes were also detected in rat HER2 and all corresponded to the epitopes predicted by computational analysis (PEPITO software), showing that LFPD gives reproducible and accurate results. Interestingly, these newly identified HER2 epitopes seem to be crucial for an effective immune response against HER2-overexpressing breast cancers and might help discriminating between metastatic breast cancer and early breast cancer patients. Overall, the results obtained in this study demonstrated the utility of LFPD and its potential application to the detection of conformational epitopes on many other molecules of interest, as well as, the development of new and potentially more effective B-cell conformational epitopes based vaccines.
- Published
- 2013
- Full Text
- View/download PDF
36. Activity and resistance of trastuzumab according to different clinical settings.
- Author
-
Tagliabue E, Campiglio M, Pupa SM, Ménard S, and Balsari A
- Subjects
- Antibody-Dependent Cell Cytotoxicity drug effects, Antineoplastic Agents pharmacology, Breast Neoplasms pathology, Cell Proliferation drug effects, DNA Repair drug effects, Drug Resistance, Neoplasm, Female, Humans, Neoadjuvant Therapy, Neoplastic Stem Cells drug effects, Receptor, ErbB-2 metabolism, Trastuzumab, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy
- Abstract
Trastuzumab, a humanized monoclonal antibody directed against HER2, has shown efficacy in breast cancers; however many patients do not respond to this reagent. Here, we discuss the potential mechanisms of trastuzumab efficacy and resistance in different clinical settings as a step toward optimizing the appropriate application of this antibody. The three major antitumor mechanisms of trastuzumab, i.e., inhibition of proliferation, antibody-dependent cell cytotoxicity (ADCC) and inhibition of DNA repair, appear to be differentially operative in different clinical settings. ADCC appears to be the prevalent mechanism in trastuzumab neoadjuvant monotherapy, whereas in neoadjuvant, adjuvant or metastatic settings in which trastuzumab is combined with chemotherapy, the relative role of ADCC is probably small, considering the compromising effects of chemotherapy on the immune cells that mediate this mechanism. In neoadjuvant and adjuvant settings involving concomitant use of trastuzumab and chemotherapy, the primary mechanism at play is presumably inhibition of DNA repair by the antibody, while in sequential protocols, the antibody acts mostly by exerting cytostatic activity through inhibition of HER2-mediated tumor cell proliferation. According to the ability of the antibody to induce cytotoxic or cytostatic antitumor effects depending on the clinical setting, different criteria, i.e., RECIST for cytotoxic effect, OS, and DFS for cytostatic, must be considered in accurately estimating antibody efficacy. Moreover, since trastuzumab resistance likely depends directly on the mechanisms responsible for its antitumor activity, resistance mechanisms must also be considered with respect to the different clinical settings., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
37. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy.
- Author
-
Ciravolo V, Huber V, Ghedini GC, Venturelli E, Bianchi F, Campiglio M, Morelli D, Villa A, Della Mina P, Menard S, Filipazzi P, Rivoltini L, Tagliabue E, and Pupa SM
- Subjects
- Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Exosomes metabolism, Female, Humans, Neoplasm Invasiveness, Receptor, ErbB-2 genetics, Trastuzumab, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Drug Resistance, Neoplasm physiology, Gene Expression Regulation, Neoplastic drug effects, Receptor, ErbB-2 metabolism
- Abstract
Exosomes are endosome-derived nanovesicles actively released into the extracellular environment and biological fluids, both under physiological and pathological conditions, by different cell types. We characterized exosomes constitutively secreted by HER2-overexpressing breast carcinoma cell lines and analyzed in vitro and in vivo their potential role in interfering with the therapeutic activity of the humanized antibody Trastuzumab and the dual tyrosine kinase inhibitor (TKI) Lapatinib anti-HER2 biodrugs. We show that exosomes released by the HER2-overexpressing tumor cell lines SKBR3 and BT474 express a full-length HER2 molecule that is also activated, although to a lesser extent than in the originating cells. Release of these exosomes was significantly modulated by the growth factors EGF and heregulin, two of the known HER2 receptor-activating ligands and naturally present in the surrounding tumor microenvironment. Exosomes secreted either in HER2-positive tumor cell-conditioned supernatants or in breast cancer patients' serum bound to Trastuzumab. Functional assays revealed that both xenogeneic and autologous HER2-positive nanovesicles, but not HER2-negative ones, inhibited Trastuzumab activity on SKBR3 cell proliferation. By contrast, Lapatinib activity on SKBR3 cell proliferation was unaffected by the presence of autologous exosomes. Together, these findings point to the role of HER2-positive exosomes in modulating sensitivity to Trastuzumab, and, consequently, to HER2-driven tumor aggressiveness., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
38. Promise and failure of targeted therapy in breast cancer.
- Author
-
Giordano A, Tagliabue E, and Pupa SM
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms pathology, Humans, Molecular Targeted Therapy, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms metabolism
- Abstract
The current molecular targets in breast cancer (BC) clinical trials were identified before the advent of the genomic era and their relevance was confirmed and validated by the introduction of gene profiling. Pioneering molecular analyses and repeated data validations on different gene platforms have thus far served to define 5 subtypes of BC based on their gene signature: luminal A, luminal B, normal-like, HER2-positive, and basal. Luminal A and B tumors are estrogen receptor (ER)-positive, while basal-like are mostly negative for ER, progesterone receptor, and HER2, i.e., triple-negative. Normal-like tumors resemble normal breast tissue and the HER2 subtype is characterized by HER2 overexpression. Here, we summarize current targeted therapeutic options for the luminal, HER2-positive, and basal-like BC subtypes with respect to results observed in clinical trials as a step toward optimizing their appropriate application in the different clinical settings. We give particular consideration to the ER- and HER2-targeted therapies approved for clinical practice with respect to their merits and shortcomings in early and advanced disease, and mention the therapeutic options currently available and potentially promising for the basal-like subtype.
- Published
- 2012
- Full Text
- View/download PDF
39. Serological identification of HSP105 as a novel non-Hodgkin lymphoma therapeutic target.
- Author
-
Zappasodi R, Bongarzone I, Ghedini GC, Castagnoli L, Cabras AD, Messina A, Tortoreto M, Tripodo C, Magni M, Carlo-Stella C, Gianni AM, Pupa SM, and Di Nicola M
- Subjects
- Animals, Antigen-Antibody Reactions, Cell Line, Tumor, Cohort Studies, Gene Expression Regulation, Neoplastic, HSP110 Heat-Shock Proteins genetics, Humans, Immunohistochemistry, Lymphoma, Non-Hodgkin genetics, Mice, Mice, SCID, Serologic Tests, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Antibodies therapeutic use, HSP110 Heat-Shock Proteins immunology, Lymphoma, Non-Hodgkin immunology, Lymphoma, Non-Hodgkin therapy
- Abstract
We reported that the clinical efficacy of dendritic cell-based vaccination is strongly associated with immunologic responses in relapsed B-cell non-Hodgkin lymphoma (B-NHL) patients. We have now investigated whether postvaccination antibodies from responders recognize novel shared NHL-restricted antigens. Immunohistochemistry and flow cytometry showed that they cross-react with allogeneic B-NHLs at significantly higher levels than their matched prevaccination samples or nonresponders' antibodies. Western blot analysis of DOHH-2 lymphoma proteome revealed a sharp band migrating at approximately 100 to 110 kDa only with postvaccine repertoires from responders. Mass spectrometry identified heat shock protein-105 (HSP105) in that molecular weight interval. Flow cytometry and immunohistochemistry disclosed HSP105 on the cell membrane and in the cytoplasm of B-NHL cell lines and 97 diagnostic specimens. A direct correlation between HSP105 expression and lymphoma aggressiveness was also apparent. Treatment of aggressive human B-NHL cell lines with an anti-HSP105 antibody had no direct effects on cell cycle or apoptosis but significantly reduced the tumor burden in xenotransplanted immunodeficient mice. In vivo antilymphoma activity of HSP105 engagement was associated with a significant local increase of Granzyme B(+) killer cells that very likely contributed to the tumor-restricted necrosis. Our study adds HSP105 to the list of nononcogenes that can be exploited as antilymphoma targets.
- Published
- 2011
- Full Text
- View/download PDF
40. Increased overall survival independent of RECIST response in metastatic breast cancer patients continuing trastuzumab treatment: evidence from a retrospective study.
- Author
-
Campiglio M, Bufalino R, Sandri M, Ferri E, Aiello RA, De Matteis A, Mottolese M, De Placido S, Querzoli P, Jirillo A, Bottini A, Fantini M, Bonetti A, Pedani F, Mauri M, Molino A, Ferro A, Pupa SM, Sasso M, Ménard S, Balsari A, and Tagliabue E
- Subjects
- Adult, Breast Neoplasms mortality, Female, Humans, Kaplan-Meier Estimate, Middle Aged, Neoplasm Metastasis, Retrospective Studies, Trastuzumab, Treatment Outcome, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms pathology
- Abstract
Recent studies have reported the potential clinical utility for metastatic breast cancer (MBC) patients of continuing trastuzumab beyond progression. Based on those results, here the authors have examined the benefits of trastuzumab-continuation by specifically evaluating RECIST responses upon first line trastuzumab-treatment as a potential predictive marker for therapeutic effect of trastuzumab-continuation beyond metastatic disease progression. The authors carried out a retrospective analysis of 272 HER2 positive MBC patients under trastuzumab treatment at 22 different oncology Italian centers during the years of 2000 and 2001 who progressed under first line trastuzumab-treatment. The primary end point of the study was the survival from the date of first documented progression upon first line trastuzumab treatment of disease. Data analysis involved the use of matching on propensity score to balance variables between treated and untreated subjects and to reduce bias. Of the 272 HER2-positive MBC patients, 154 (56.6%) continued treatment. 79 (51.3%) of those 154 patients showed responses based on RECIST criteria during first-line trastuzumab-treatment. Of the 118 patients that suspended trastuzumab, RECIST responses had been observed in 44 (37.3%). Cox proportional hazards analysis of progressed patients, matched using propensity score, showed that discontinuation of trastuzumab at metastatic disease progression was a risk factor for significantly reduced overall survival in both responder (HR = 2.23; 95% CI = 1.03-4.82) and non-responder groups (HR = 3.53, 95% CI = 1.73-7.21), with no significant differences in the two estimated HRs (P-value of the likelihood-ratio test = 0.690). Continued trastuzumab treatment after disease progression has clinically and statistically significant effects in both RECIST responder and non-responder MBC patients.
- Published
- 2011
- Full Text
- View/download PDF
41. The human splice variant Δ16HER2 induces rapid tumor onset in a reporter transgenic mouse.
- Author
-
Marchini C, Gabrielli F, Iezzi M, Zenobi S, Montani M, Pietrella L, Kalogris C, Rossini A, Ciravolo V, Castagnoli L, Tagliabue E, Pupa SM, Musiani P, Monaci P, Menard S, and Amici A
- Subjects
- Animals, Cell Line, Tumor, Dimerization, Disulfides, Female, Genes, Reporter, Humans, Mammary Neoplasms, Animal, Mice, Mice, Transgenic, Oncogenes, Protein Isoforms, Alternative Splicing, Mutation, Promoter Regions, Genetic, Receptor, ErbB-2 genetics
- Abstract
Several transgenic mice models solidly support the hypothesis that HER2 (ERBB2) overexpression or mutation promotes tumorigenesis. Recently, a HER2 splice variant lacking exon-16 (Δ16HER2) has been detected in human breast carcinomas. This alternative protein, a normal byproduct of HER2, has an increased transforming potency compared to wild-type (wt) HER2 receptors. To examine the ability of Δ16HER2 to transform mammary epithelium in vivo and to monitor Δ16HER2-driven tumorigenesis in live mice, we generated and characterized a mouse line that transgenically expresses both human Δ16HER2 and firefly luciferase under the transcriptional control of the MMTV promoter. All the transgenic females developed multifocal mammary tumors with a rapid onset and an average latency of 15.11 weeks. Immunohistochemical analysis revealed the concurrent expression of luciferase and the human Δ16HER2 oncogene only in the mammary gland and in strict correlation with tumor development. Transgenic Δ16HER2 expressed on the tumor cell plasma membrane from spontaneous mammary adenocarcinomas formed constitutively active homodimers able to activate the oncogenic signal transduction pathway mediated through Src kinase. These new transgenic animals demonstrate the ability of the human Δ16HER2 isoform to transform "per se" mammary epithelium in vivo. The high tumor incidence as well as the short latency strongly suggests that the Δ16HER2 splice variant represents the transforming form of the HER2 oncoprotein.
- Published
- 2011
- Full Text
- View/download PDF
42. Do pre-diagnostic drinking habits influence breast cancer survival?
- Author
-
Allemani C, Berrino F, Krogh V, Sieri S, Pupa SM, Tagliabue E, Tagliabue G, and Sant M
- Subjects
- Adult, Aged, Aged, 80 and over, Breast Neoplasms etiology, Female, Humans, Italy epidemiology, Life Style, Likelihood Functions, Middle Aged, Predictive Value of Tests, Prognosis, Risk Assessment, Risk Factors, Severity of Illness Index, Survival Analysis, Alcohol Drinking adverse effects, Breast Neoplasms diagnosis, Breast Neoplasms mortality
- Abstract
Aims and Background: Alcohol consumption increases the risk of developing breast cancer and may also be associated with late diagnosis, recurrence, distant metastases and death. Many studies have examined the role of alcohol as a risk factor for the development of breast cancer, but very few studies have addressed the role of alcohol as a prognostic factor for survival among women diagnosed with breast cancer. The aim of this study was to investigate the survival of women with breast cancer in relation to pre-diagnostic alcohol intake and other factors known to influence prognosis., Methods: We analyzed data for 264 women in the EUROCARE and ORDET studies who were diagnosed with breast cancer from 1987 up to 31 December 2001 and for whom information was available on follow-up, stage at diagnosis, HER-2 and hormone receptor status, and pre-diagnostic dietary alcohol intake, categorized as zero (0 g/day, non-drinkers), moderate (up to 13 g/day, about 1 serving) and high (>13 g/day). Ten-year relative survival was estimated using the maximum-likelihood approach. The excess risk of death within 10 years of diagnosis was modeled by level of alcohol intake, adjusting separately for age, stage, body mass index and tumor subtype., Results: Ten-year relative survival was lower in women who drank more than 13 g/day (65%; 95% CI, 47-78) than in non-drinkers (88%; 95% CI, 75-95). The excess risk of death within 10 years was significantly higher in women who drank more than 13 g/day than non-drinkers (relative excess risk, 4.13; 95% CI, 1.69-10.10) and was not altered by adjustment for other prognostic factors. The excess risk within 10 years was higher for women with a body mass index of 25 kg/m2 or higher (relative excess risk, 2.20; 95% CI, 1.01-4.70) and higher for those with more advanced disease., Conclusions: Women who drank more than 13 g alcohol per day had lower survival than non-drinkers. The excess risk of death within 10 years of diagnosis was unaffected by other known risk factors. High alcohol consumption may be an adverse prognostic factor for breast cancer.
- Published
- 2011
- Full Text
- View/download PDF
43. The HER2 World: Better Treatment Selection for Better Outcome.
- Author
-
Tagliabue E, Campiglio M, Pupa SM, Balsari A, and Ménard S
- Subjects
- Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Proliferation drug effects, Clinical Trials as Topic, DNA Repair drug effects, Disease Progression, Drug Synergism, Female, Humans, Neoadjuvant Therapy methods, Patient Selection, Trastuzumab, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Agents pharmacology, Biomarkers, Tumor metabolism, Breast Neoplasms metabolism, Molecular Targeted Therapy methods, Receptor, ErbB-2 metabolism
- Abstract
Understanding the mechanisms of trastuzumab efficacy and resistance is a step toward optimizing treatment outcome in HER2-positive breast carcinoma patients. Preclinical studies have indicated different trastuzumab antitumor mechanisms, that is, cytostatic inhibition of tumor proliferation, antibody-dependent cell cytotoxicity, and inhibition of HER2-mediated DNA repair. Clinical studies point to the clinical setting dependence of these mechanisms, with antibody-dependent cell cytotoxicity predominating when trastuzumab is used as monotherapy in neoadjuvant and metastatic settings, whereas inhibition of DNA repair predominates in neoadjuvant and adjuvant settings involving concomitant trastuzumab and chemotherapy; in sequential protocols, the antibody appears to act primarily through cytostatic activity by inhibiting HER2-mediated cell proliferation. Because the mechanisms of resistance to trastuzumab likely depend directly on those of its antitumor activity, resistance mechanisms must also be considered with respect to the different clinical settings. Moreover, the response to this reagent should be assessed according to its ability to induce tumor cytotoxic or cytostatic activity.
- Published
- 2011
- Full Text
- View/download PDF
44. Improved clinical outcome in indolent B-cell lymphoma patients vaccinated with autologous tumor cells experiencing immunogenic death.
- Author
-
Zappasodi R, Pupa SM, Ghedini GC, Bongarzone I, Magni M, Cabras AD, Colombo MP, Carlo-Stella C, Gianni AM, and Di Nicola M
- Subjects
- Adenosine Triphosphate metabolism, Apoptosis radiation effects, Blotting, Western, Calreticulin metabolism, Cancer Vaccines administration & dosage, Cell Line, Tumor, Cell Survival immunology, Cell Survival radiation effects, Cytotoxicity, Immunologic immunology, Dendritic Cells immunology, Dendritic Cells metabolism, HSP90 Heat-Shock Proteins metabolism, Hot Temperature, Humans, Lymphoma, B-Cell pathology, Lymphoma, B-Cell prevention & control, Necrosis immunology, Protein Transport immunology, Time Factors, Treatment Outcome, Antigens, Neoplasm immunology, Apoptosis immunology, Cancer Vaccines immunology, Lymphoma, B-Cell immunology
- Abstract
Increasing evidence argues that the success of an anticancer treatment may rely on immunoadjuvant side effects including the induction of immunogenic tumor cell death. Based on the assumption that this death mechanism is a similar prerequisite for the efficacy of an active immunotherapy using killed tumor cells, we examined a vaccination strategy using dendritic cells (DC) loaded with apoptotic and necrotic cell bodies derived from autologous tumors. Using this approach, clinical and immunologic responses were achieved in 6 of 18 patients with relapsed indolent non-Hodgkin's lymphoma (NHL). The present report illustrates an impaired ability of the neoplastic cells used to vaccinate nonresponders to undergo immunogenic death on exposure to a cell death protocol based on heat shock, γ-ray, and UVC ray. Interestingly, when compared with doxorubicin, this treatment increased surface translocation of calreticulin and cellular release of high-mobility group box 1 and ATP in histologically distinct NHL cell lines. In contrast, treated lymphoma cells from responders displayed higher amounts of calreticulin and heat shock protein 90 (HSP90) compared with those from nonresponders and boosted the production of specific antibodies when loaded into DCs for vaccination. Accordingly, the extent of calreticulin and HSP90 surface expression in the DC antigenic cargo was significantly associated with the clinical and immunologic responses achieved. Our results indicate that a positive clinical effect is obtained when immunogenically killed autologous neoplastic cells are used for the generation of a DC-based vaccine. Therapeutic improvements may thus be accomplished by circumventing the tumor-impaired ability to undergo immunogenic death and prime the antitumor immune response., (Copyright © 2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
45. Shed HER2 extracellular domain in HER2-mediated tumor growth and in trastuzumab susceptibility.
- Author
-
Ghedini GC, Ciravolo V, Tortoreto M, Giuffrè S, Bianchi F, Campiglio M, Mortarino M, Figini M, Coliva A, Carcangiu ML, Zambetti M, Piazza T, Ferrini S, Ménard S, Tagliabue E, and Pupa SM
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Breast Neoplasms genetics, Cell Line, Tumor, Cell Proliferation, Female, Humans, Mice, Mice, Nude, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Structure, Tertiary, Receptor, ErbB-2 genetics, Signal Transduction physiology, Trastuzumab, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Receptor, ErbB-2 chemistry, Receptor, ErbB-2 metabolism
- Abstract
The question of the serum HER2 extracellular domain (HER2/ECD) measurement for prediction of response to the anti-HER2 antibody Trastuzumab is still an open and current matter of clinical debate. To elucidate the involvement of shed HER2/ECD in HER2-driven tumor progression and in guiding therapy of individual patients, we examined biological effects exerted by elevated HER2/ECD in cancer growth and in response to Trastuzumab. To this purpose SKOV3 tumor cells were stably transfected to release a recombinant HER2/ECD molecule (rECD). Transfectants releasing high levels of 110-kDa rECD, identical in size to native HER2/ECD (nECD), grew significantly slower than did controls, which constitutively released only basal levels of nECD. While transmembrane HER2 and HER1 were expressed at equal levels by both controls and transfected cells, activation of these molecules and of downstream ERK2 and Akt was significantly reduced only in rECD transfectants. Surface plasmon resonance analysis revealed heterodimerization of the rECD with HER1, -2, and -3. In cell growth bioassays in vitro, shed HER2 significantly blocked HER2-driven tumor cell proliferation. In mice, high levels of circulating rECD significantly impaired HER2-driven SKOV3 tumor growth but not that of HER2-negative tumor cells. In vitro and in mice, Trastuzumab significantly inhibited tumor growth due to the rECD-facilitated accumulation of the antibody on tumor cells. Globally our findings sustain the biological relevance of elevated HER2/ECD levels in the outcome of HER2-disease and in the susceptibility to Trastuzumab-based therapy., ((c) 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
46. HER2 as a target for breast cancer therapy.
- Author
-
Tagliabue E, Balsari A, Campiglio M, and Pupa SM
- Subjects
- Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized, Antineoplastic Agents adverse effects, Breast Neoplasms enzymology, Breast Neoplasms immunology, Cancer Vaccines adverse effects, Drug Resistance, Neoplasm, Female, Humans, Lapatinib, Protein Kinase Inhibitors adverse effects, Quinazolines adverse effects, Receptor, ErbB-2 immunology, Receptor, ErbB-2 metabolism, Trastuzumab, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Cancer Vaccines therapeutic use, Protein Kinase Inhibitors therapeutic use, Quinazolines therapeutic use, Receptor, ErbB-2 antagonists & inhibitors
- Abstract
Importance of the Field: Differential levels of HER2 expression in normal versus HER2-overexpressing breast carcinomas, together with the demonstration of a key role for HER2 in tumor progression, make HER2 an ideal target for specific therapeutic approaches., Areas Covered in This Review: This review considers the clinical value of trastuzumab and lapatinib, the two HER2-targeted therapies approved for clinical practice. References were chosen by searching the PubMed and MEDLINE datasets using as search term: 'HER2', in association with 'prognosis', 'response', 'trastuzumab', 'lapatinib' and 'resistance'., What the Reader Will Gain: This review deals with HER2 as a target for breast carcinoma treatment, focusing on anti-HER2 therapies used in clinical practice, their merits and shortcomings., Take Home Message: The benefit of anti-HER2 therapies demonstrated in clinical trials indicates that HER2 is, to date, one of the most promising molecules for targeted therapy. Nevertheless, since tumor cells utilizing alternative growth signaling pathways through transmembrane receptors as well as intracellular signaling transduction molecules can bypass HER2 blockade, a future ambitious aim is the successful combination of anti-HER2 strategies with drugs directed to molecules that contribute to anti-HER2 resistance.
- Published
- 2010
- Full Text
- View/download PDF
47. Vaccination with autologous tumor-loaded dendritic cells induces clinical and immunologic responses in indolent B-cell lymphoma patients with relapsed and measurable disease: a pilot study.
- Author
-
Di Nicola M, Zappasodi R, Carlo-Stella C, Mortarini R, Pupa SM, Magni M, Devizzi L, Matteucci P, Baldassari P, Ravagnani F, Cabras A, Anichini A, and Gianni AM
- Subjects
- Aged, Cancer Vaccines adverse effects, Cancer Vaccines standards, Female, Follow-Up Studies, Humans, Immunophenotyping, Killer Cells, Natural immunology, Lymphoma, B-Cell diagnostic imaging, Male, Middle Aged, Pilot Projects, Quality Control, Recurrence, T-Lymphocytes, Regulatory immunology, Tomography, X-Ray Computed, Treatment Outcome, Cancer Vaccines administration & dosage, Dendritic Cells immunology, Immunotherapy, Adoptive, Lymphoma, B-Cell immunology, Lymphoma, B-Cell therapy
- Abstract
Eighteen relapsed patients with measurable indolent non-Hodgkin lymphoma (NHL) were vaccinated with dendritic cells (DCs) loaded with killed autologous tumor cells. Six patients had objective clinical responses including 3 continuous complete responses (CRs) and 3 partial responses (PRs), with a median follow up of 50.5 months. Eight patients had stable disease, whereas 4 had progressive disease. Clinical responses were significantly associated with a reduction in CD4(+)CD25(+)FOXP3(+) regulatory T cells, an increase in CD3(-)CD56(dim)CD16(+) natural killer (NK) cells, and maturation of lymphocytes to the effector memory stage in either postvaccination peripheral blood or tumor specimen samples. In partial responding patients, vaccination significantly boosted the IFN-gamma-producing T-cell response to autologous tumor challenge. In one HLA-A*0201(+) patient who achieved CR, IL-4 release by circulating T cells in response to tumor-specific IgH-encoded peptides was also documented. Immunohistochemical analysis of tumor biopsies using biotin-conjugated autologous serum samples revealed a tumor-restricted humoral response only in the postvaccination serum from responding patients. Collectively these results demonstrate that vaccination with tumor-loaded DCs may induce both T- and B-cell responses and produces clinical benefits in indolent NHL patients with measurable disease. This study is registered with the Istituto Superiore di Sanità: http://www.iss.it with protocol number 7578-PRE 21-801.
- Published
- 2009
- Full Text
- View/download PDF
48. Regulation of breast cancer response to chemotherapy by fibulin-1.
- Author
-
Pupa SM, Giuffré S, Castiglioni F, Bertola L, Cantú M, Bongarzone I, Baldassari P, Mortarini R, Argraves WS, Anichini A, Menard S, and Tagliabue E
- Subjects
- Animals, Breast Neoplasms pathology, Calcium-Binding Proteins antagonists & inhibitors, Calcium-Binding Proteins genetics, Cell Line, Tumor, Cell Survival drug effects, Extracellular Matrix drug effects, Extracellular Matrix metabolism, Extracellular Matrix Proteins metabolism, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, RNA, Small Interfering genetics, Transfection, Antibiotics, Antineoplastic pharmacology, Breast Neoplasms drug therapy, Calcium-Binding Proteins physiology, Doxorubicin pharmacology
- Abstract
Doxorubicin treatment was found to augment the expression of the extracellular matrix (ECM) protein fibulin-1 in cultured human breast cancer cell lines and in MDA-MB-361 tumors grown in athymic mice. Doxorubicin was also found to augment tumor expression of the fibulin-1-binding proteins fibronectin and laminin-1. Growth of breast cancer cell lines on Matrigel, an ECM extract containing fibulin-1 and laminin-1, resulted in lower levels of doxorubicin-induced apoptosis as compared with controls. Moreover, tumors formed by injection of athymic mice with MDA-MB-361 cells mixed with Matrigel were significantly more doxorubicin resistant and displayed lower levels of apoptosis compared with those that formed in the absence of Matrigel. Monoclonal antibodies against fibulin-1 reversed Matrigel-dependent doxorubicin resistance. Furthermore, small interfering RNA-mediated suppression of fibulin-1 expression in breast cancer cells resulted in a 10-fold increase in doxorubicin sensitivity as compared with control cells. Together, these findings point to a role for fibulin-1 in breast cancer chemoresistance.
- Published
- 2007
- Full Text
- View/download PDF
49. SEL1L a multifaceted protein playing a role in tumor progression.
- Author
-
Biunno I, Cattaneo M, Orlandi R, Canton C, Biagiotti L, Ferrero S, Barberis M, Pupa SM, Scarpa A, and Ménard S
- Subjects
- Amino Acid Sequence, Animals, Cell Proliferation, Cell Transformation, Neoplastic genetics, Chromosome Deletion, Chromosomes, Human, Pair 14, DNA Mutational Analysis, DNA, Neoplasm genetics, Disease Progression, Exons genetics, Fetus chemistry, Gene Expression Regulation, Neoplastic genetics, Humans, Molecular Sequence Data, Neoplasm Metastasis, Neoplasms genetics, Polymorphism, Genetic genetics, Protein Isoforms analysis, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms physiology, Proteins analysis, Proteins chemistry, Proteins genetics, Receptors, Notch genetics, Receptors, Notch physiology, Signal Transduction genetics, Signal Transduction physiology, Transforming Growth Factor beta genetics, Transforming Growth Factor beta physiology, Cell Transformation, Neoplastic pathology, Gene Expression Regulation, Neoplastic physiology, Neoplasms pathology, Neoplasms physiopathology, Proteins physiology
- Abstract
Since the cloning in 1997 of SEL1L, the human ortholog of the sel-1 gene of C. elegans, most studies have focused on its role in cancer progression and have provided significant evidences to link its increased expression to a decrease in tumor aggressiveness. SEL1L resides on a "Genome Desert area" on chromosome 14q24.3-31 and is highly conserved in evolution. The function of the SEL1L encoded protein is still very elusive although, several evidences from lower organisms indicate that it plays a major role in protein degradation using the ubiquitin-proteosome system. SEL1L has a very complex structure made up of modules: genomically it consists of 21 exons featuring several alternative transcripts encoding for putative protein isoforms. This structural complexity ensures protein flexibility and specificity, indeed the protein was found in different sub-cellular compartments and may turn on a particular transcript in response to specific stimuli. The overall architecture of SEL1L guarantees an exquisite regulation in the expression of the gene., (Copyright 2005 Wiley-Liss, Inc.)
- Published
- 2006
- Full Text
- View/download PDF
50. Role of exon-16-deleted HER2 in breast carcinomas.
- Author
-
Castiglioni F, Tagliabue E, Campiglio M, Pupa SM, Balsari A, and Ménard S
- Subjects
- 3T3 Cells, Animals, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Humanized, Antineoplastic Agents therapeutic use, Apoptosis, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cell Proliferation, Cells, Cultured, Drug Tolerance, Emodin pharmacology, Enzyme Inhibitors pharmacology, ErbB Receptors antagonists & inhibitors, Flow Cytometry, Gefitinib, Gene Expression Regulation, Neoplastic, Humans, Kidney drug effects, Kidney metabolism, Mice, Mice, Inbred BALB C, Mice, Nude, Polymerase Chain Reaction, Quinazolines therapeutic use, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism, Trastuzumab, Breast Neoplasms genetics, Exons genetics, RNA Splicing, Receptor, ErbB-2 genetics
- Abstract
A splice variant of the human gene HER2, lacking exon-16 (DeltaHER2) which encodes a small extracellular region, has been described. This altered receptor forms disulfide bond-stabilized homodimers. We report here that the DeltaHER2 splice variant represents about 9% of the HER2 mRNA obtained from most of the 46 breast carcinoma samples with HER2 expression levels ranging from 3+ to 0 by HercepTest. Analysis of human cells transfected with DeltaHER2 or wild-type (WT) cDNA revealed no growth of WT cells in nude mice, whereas clones expressing 10-fold less DeltaHER2 were tumorigenic. Unlike WT transfectants, DeltaHER2-expressing cells showed low sensitivity to two new therapeutic drugs targeting receptors of the HER family (ZD1839 and Trastuzumab), whereas an inhibitor of the HER2 tyrosine kinase domain (Emodin) blocked activation of both DeltaHER2 and WT transfectants. Taken together, our findings indicate that the DeltaHER2 transcript encodes the transforming form of the oncoprotein. It is plausible that malignant transformation arises when a critical threshold of DeltaHER2 is reached in HER2-overexpressing tumors. Specific inhibitors of HER2 catalytic activity represent a promising approach to therapy of HER2-overexpressing tumors.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.