Luca Malorni
Baylor College of Medicine
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Luca Malorni.
Cancer Research | 2008
Cj Creighton; Suleiman Massarweh; Shixia Huang; Anna Tsimelzon; Sg Hilsenbeck; Ck Osborne; Jiang Shou; Luca Malorni; Rachel Schiff
The effectiveness of therapies targeting specific pathways in breast cancer, such as the estrogen receptor or HER2, is limited because many tumors manifest resistance, either de novo or acquired, during the course of treatment. To investigate molecular mechanisms of resistance, we used two xenograft models of estrogen receptor-positive (ER+) breast cancer, one with and one without HER2 overexpression (MCF7/HER2-18 and MCF7 wt, respectively). Mice with established tumors were assigned to the following treatment groups: estrogen supplementation (E2), estrogen deprivation (ED), ED plus tamoxifen (Tam), all with or without the epidermal growth factor receptor tyrosine kinase inhibitor gefitinib (G). Another group received ED plus the antiestrogen fulvestrant (MCF7 wt only). Tumors with acquired or de novo resistance to these endocrine therapies were profiled for gene expression and compared with tumors in the E2 control group. One class of genes underexpressed in endocrine-resistant tumors (relative to E2-treated tumors) were estrogen inducible in vitro and associated with ER+ human breast cancers (luminal subtype). Another class of genes overexpressed in tumors with acquired resistance in both models represented transcriptional targets of HER2 signaling and was associated with ER-/HER2+ human cancers (ERBB2+ subtype). A third class of genes overexpressed in MCF7/HER2-18 tumors exhibiting de novo resistance to tamoxifen was associated with ER+ human cancers but not with estrogen-regulated genes. Thus, in response to various endocrine therapy regimens, these xenograft breast tumors shut down classic estrogen signaling and activate alternative pathways such as HER2 that contribute to treatment resistance. Over time, the molecular phenotype of breast cancer can change.
Molecular Oncology | 2015
Marta Pestrin; Francesca Salvianti; Francesca Galardi; Francesca De Luca; Natalie Turner; Luca Malorni; Mario Pazzagli; Angelo Di Leo; Pamela Pinzani
Circulating Tumor Cells (CTCs) represent a “liquid biopsy of the tumor” which might allow real‐time monitoring of cancer biology and therapies in individual patients. CTCs are extremely rare in the blood stream and their analysis is technically challenging.
Nature Reviews Clinical Oncology | 2015
Christopher D. Hart; Ilenia Migliaccio; Luca Malorni; Cristina Guarducci; Laura Biganzoli; Angelo Di Leo
Hormone-receptor-positive breast cancer accounts for the majority—up to 80%—of all breast cancers. The evolution of breast cancer from early stage to the metastatic setting leads to increased heterogeneity, the occurrence of new mutations, and the development of treatment resistance representing a great challenge for management decisions. Unfortunately, little data exist to offer guidance in this context, and a reliance on traditional clinical parameters remains when deciding on optimal treatment. In advanced-stage oestrogen receptor-positive (ER+) disease, ongoing issues include the choice between endocrine therapy and chemotherapy, the appropriate sequence of treatment agents, and the incorporation of biological agents, such as everolimus, into the treatment armamentarium. In metastatic disease, repeated biopsies can help to reassess the receptor or genetic mutational status; however, the evidence to support this approach is limited. In this Review, we examine the current evidence that can guide treatment decisions in patients with advanced-stage ER+ breast cancer, discuss how to tackle these therapeutic challenges and provide suggestions for the optimal management of this patient population.
Oncotarget | 2016
Luca Malorni; Silvano Piazza; Yari Ciani; Cristina Guarducci; Martina Bonechi; Chiara Biagioni; Christopher D. Hart; Roberto Verardo; Angelo Di Leo; Ilenia Migliaccio
Palbociclib is a CDK4/6 inhibitor that received FDA approval for treatment of hormone receptor positive (HR+) HER2 negative (HER2neg) advanced breast cancer. To better personalize patients treatment it is critical to identify subgroups that would mostly benefit from it. We hypothesize that complex alterations of the Retinoblastoma (Rb) pathway might be implicated in resistance to CDK4/6 inhibitors and aim to investigate whether signatures of Rb loss-of-function would identify breast cancer cell lines resistant to palbociclib. We established a gene expression signature of Rb loss-of-function (RBsig) by identifying genes correlated with E2F1 and E2F2 expression in breast cancers within The Cancer Genome Atlas. We assessed the RBsig prognostic role in the METABRIC and in a comprehensive breast cancer meta-dataset. Finally, we analyzed whether RBsig would discriminate palbociclib-sensitive and -resistant breast cancer cells in a large RNA sequencing-based dataset. The RBsig was associated with RB1 genetic status in all tumors (p <7e-32) and in luminal or basal subtypes (p < 7e-11 and p < 0.002, respectively). The RBsig was prognostic in the METABRIC dataset (discovery: HR = 1.93 [1.5-2.4] p = 1.4e-08; validation: HR = 2.01 [1.6-2.5] p = 1.3e-09). Untreated and endocrine treated patients with estrogen receptor positive breast cancer expressing high RBsig had significantly worse recurrence free survival compared to those with low RBsig (HR = 2.37 [1.8 − 3.2] p = 1.87e−08 and HR = 2.62 [1.9− 3.5] p = 8.6e−11, respectively). The RBsig was able to identify palbociclib resistant and sensitive breast cancer cells (ROC AUC = 0,7778). Signatures of RB loss might be helpful in personalizing treatment of patients with HR+/HER2neg breast cancer. Further validation in patients receiving palbociclib is warranted.
Cancers | 2014
Natalie Turner; Marta Pestrin; Francesca Galardi; Francesca De Luca; Luca Malorni; Angelo Di Leo
Circulating tumor cell (CTC) count has prognostic significance in metastatic breast cancer, but the predictive utility of CTCs is uncertain. Molecular studies on CTCs have often been limited by a low number of CTCs isolated from a high background of leukocytes. Improved enrichment techniques are now allowing molecular characterisation of single CTCs, whereby molecular markers on single CTCs may provide a real-time assessment of tumor biomarker status from a blood test or “liquid biopsy”, potentially negating the need for a more invasive tissue biopsy. The predictive ability of CTC biomarker analysis has predominantly been assessed in relation to HER2, with variable and inconclusive results. Limited data exist for other biomarkers, such as the estrogen receptor. In addition to the need to define and validate the most accurate and reproducible method for CTC molecular analysis, the clinical relevance of biomarkers, including gain of HER2 on CTC after HER2 negative primary breast cancer, remains uncertain. This review summarises the currently available data relating to biomarker evaluation on CTCs and its role in directing management in metastatic breast cancer, discusses limitations, and outlines measures that may enable future development of this approach.
Journal of Experimental & Clinical Cancer Research | 2009
Maddalena Barba; Li Yang; Holger J. Schünemann; Francesca Sperati; Sara Grioni; Saverio Stranges; Kim C. Westerlind; Giovanni Blandino; Michele Gallucci; Rossella Lauria; Luca Malorni; Paola Muti
ObjectiveTo investigate prostate cancer (Pca) risk in relation to estrogen metabolism, expressed as urinary 2-hydroxyestrone (2-OHE1), 16α-hydroxyestrone (16α-OHE1) and 2-OHE1 to 16α-OHE1 ratio.MethodsWe conducted a case-control study within the Western New York Health Cohort Study (WNYHCS) from 1996 to 2001. From January 2003 through September 2004, we completed the re-call and follow-up of 1092 cohort participants. Cases (n = 26) and controls (n = 110) were matched on age, race and recruitment period according to a 1:4 ratio. We used the unconditional logistic regression to compute crude and adjusted odds ratios (OR) and 95% confident interval (CI) of Pca in relation to 2-OHE1, 16αOHE1 and 2-OHE1 to 16α-OHE1 by tertiles of urine concentrations (stored in a biorepository for an average of 4 years). We identified age, race, education and body mass index as covariates. We also conducted a systematic review of the literature which revealed no additional studies, but we pooled the results from this study with those from a previously conducted case-control study using the DerSimonian-Laird random effects method.ResultsWe observed a non-significant risk reduction in the highest tertile of 2-OHE1 (OR 0.72, 95% CI 0.25-2.10). Conversely, the odds in the highest tertile of 16α-OHE1 showed a non-significant risk increase (OR 1.76 95% CI 0.62-4.98). There was a suggestion of reduced Pca risk for men in the highest tertile of 2-OHE1 to 16α-OHE1 ratio (OR 0.56, 95% CI 0.19-1.68). The pooled estimates confirmed the association between an increased Pca risk and higher urinary levels of 16α-OHE1 (third vs. first tertile: OR 1.82, 95% CI 1.09-3.05) and the protective effect of a higher 2-OHE 1 to 16α-OHE1 ratio (third vs. first tertile: OR 0.53, 95% CI 0.31-0.90).ConclusionOur study and the pooled results provide evidence for a differential role of the estrogen hydroxylation pathway in Pca development and encourage further study.
BMC Medicine | 2015
Ilenia Migliaccio; Luca Malorni; Christopher D. Hart; Cristina Guarducci; Angelo Di Leo
The standard of care for patients with hormone receptor positive, human epidermal growth factor receptor type 2 negative advanced breast cancer is endocrine therapy. Endocrine agents, including aromatase inhibitors, tamoxifen, and fulvestrant, are often administered alone as first line treatment and demonstrate durable responses with limited side effects. Endocrine resistance represents a major clinical problem. In the future, poly-endocrine therapy and combination therapies with biological agents might become valuable options for the first line treatment of hormone receptor-positive advanced breast cancer. However, it will be critical to develop clinical tools that can reliably identify the subgroup of patients most likely to benefit from endocrine therapy alone, and those who might benefit from alternative approaches.Herein, we will review and discuss current issues in the endocrine treatment of postmenopausal patients with hormone receptor positive, human epidermal growth factor receptor type 2 negative advanced breast cancer.
Molecular Cancer Research | 2016
Luca Malorni; Mario Giuliano; Ilenia Migliaccio; Tao Wang; Chad J. Creighton; Mathieu Lupien; Xiaoyong Fu; Susan G. Hilsenbeck; Nuala Healy; Carmine De Angelis; Abhijit Mazumdar; Meghana V. Trivedi; Suleiman Massarweh; Carolina Gutierrez; Sabino De Placido; Rinath Jeselsohn; Myles Brown; Powel H. Brown; C. Kent Osborne; Rachel Schiff
The transcription factor AP-1 is downstream of growth factor (GF) receptors (GFRs) and stress-related kinases, both of which are implicated in breast cancer endocrine resistance. Previously, we have suggested that acquired endocrine resistance is associated with increased activity of AP-1 in an in vivo model. In this report, we provide direct evidence for the role of AP-1 in endocrine resistance. First, significant overlap was found between genes modulated in tamoxifen resistance and a gene signature associated with GF-induced estrogen receptor (ER) cistrome. Interestingly, these overlapping genes were enriched for key signaling components of GFRs and stress-related kinases and had AP-1 motifs in their promoters/enhancers. Second, to determine a more definitive role of AP-1 in endocrine resistance, AP-1 was inhibited using an inducible dominant-negative (DN) cJun expressed in MCF7 breast cancer cells in vitro and in vivo. AP-1 blockade enhanced the antiproliferative effect of endocrine treatments in vitro, accelerated xenograft tumor response to tamoxifen and estrogen deprivation in vivo, promoted complete regression of tumors, and delayed the onset of tamoxifen resistance. Induction of DN-cJun after the development of tamoxifen resistance resulted in dramatic tumor shrinkage, accompanied by reduced proliferation and increased apoptosis. These data suggest that AP-1 is a key determinant of endocrine resistance by mediating a global shift in the ER transcriptional program. Implications: AP-1 represents a viable therapeutic target to overcome endocrine resistance. Mol Cancer Res; 14(5); 470–81. ©2016 AACR.
Clinical Cancer Research | 2016
Rinath Jeselsohn; William T. Barry; Ilenia Migliaccio; Chiara Biagioni; Jin Zhao; Jonas de Tribolet-Hardy; Christina Guarducci; Martina Bonechi; Naomi Laing; Myles Brown; Angelo Di Leo; Luca Malorni
Purpose: Fulvestrant is an estrogen receptor (ER) antagonist and an approved treatment for metastatic estrogen receptor–positive (ER+) breast cancer. With the exception of ER levels, there are no established predictive biomarkers of response to single-agent fulvestrant. We attempted to identify a gene signature of response to fulvestrant in advanced breast cancer. Experimental Design: Primary tumor samples from 134 patients enrolled in the phase III CONFIRM study of patients with metastatic ER+ breast cancer comparing treatment with either 250 mg or 500 mg fulvestrant were collected for genome-wide transcriptomic analysis. Gene expression profiling was performed using Affymetrix microarrays. An exploratory analysis was performed to identify biologic pathways and new signatures associated with response to fulvestrant. Results: Pathway analysis demonstrated that increased EGF pathway and FOXA1 transcriptional signaling is associated with decreased response to fulvestrant. Using a multivariate Cox model, we identified a novel set of 37 genes with an expression that is independently associated with progression-free survival (PFS). TFAP2C, a known regulator of ER activity, was ranked second in this gene set, and high expression was associated with a decreased response to fulvestrant. The negative predictive value of TFAP2C expression at the protein level was confirmed by IHC. Conclusions: We identified biologic pathways and a novel gene signature in primary ER+ breast cancers that predicts for response to treatment in the CONFIRM study. These results suggest potential new therapeutic targets and warrant further validation as predictive biomarkers of fulvestrant treatment in metastatic breast cancer. Clin Cancer Res; 22(23); 5755–64. ©2016 AACR.
The Breast | 2013
Natalie Turner; Laura Biganzoli; Luca Malorni; Ilenia Migliaccio; Erica Moretti; Marta Pestrin; Giuseppina Sanna; Olimpia Siclari; Angelo Di Leo
The Oxford Overview process has provided us with extremely high-powered meta-analyses assessing the role of adjuvant chemotherapy in early breast cancer. From the most recent publication, the proportional benefits from chemotherapy are relatively equivalent across all patient subgroups, a finding contradictory to our growing understanding of the role of tumour biology in dictating chemosensitivity. Several factors, including heterogeneity of patient groups and chemotherapy regimens, lack of data on underlying tumour biological subtypes, and confounding effect of chemotherapy-induced ovarian suppression in premenopausal women with hormone receptor positive breast cancer, impact on the applicability and clinical utility of the Overview in current and future oncological practice. With these considerations, the Overview has become less clinically relevant as a tool for guiding adjuvant chemotherapy treatment decisions, and a new direction is required.
Collaboration
Dive into the Luca Malorni's collaboration.
European Organisation for Research and Treatment of Cancer
View shared research outputs