Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Franco Caramia is active.

Publication


Featured researches published by Franco Caramia.


Genome Medicine | 2015

Mutational landscape of mucinous ovarian carcinoma and its neoplastic precursors.

Georgina L. Ryland; Sally M. Hunter; Maria A. Doyle; Franco Caramia; Jason Li; Simone M. Rowley; Michael Christie; Prue E. Allan; Andrew N. Stephens; David Bowtell; Ian G. Campbell; Kylie L. Gorringe

Mucinous ovarian tumors are an unusual group of rare neoplasms with an apparently clear progression from benign to borderline to carcinoma, yet with a controversial cell of origin in the ovarian surface epithelium. They are thought to be molecularly distinct from other ovarian tumors but there have been no exome-level sequencing studies performed to date. To understand the genetic etiology of mucinous ovarian tumors and assess the presence of novel therapeutic targets or pathways, we undertook exome sequencing of 24 tumors encompassing benign (5), borderline (8) and carcinoma (11) histologies and also assessed a validation cohort of 58 tumors for specific gene regions including exons 4–9 of TP53. The predominant mutational signature was of C>T transitions in a NpCpG context, indicative of deamination of methyl-cytosines. As well as mutations in known drivers (KRAS, BRAF and CDKN2A), we identified a high percentage of carcinomas with TP53 mutations (52 %), and recurrent mutations in RNF43, ELF3, GNAS, ERBB3 and KLF5. The diversity of mutational targets suggests multiple routes to tumorigenesis in this heterogeneous group of tumors that is generally distinct from other ovarian subtypes.BackgroundMucinous ovarian tumors are an unusual group of rare neoplasms with an apparently clear progression from benign to borderline to carcinoma, yet with a controversial cell of origin in the ovarian surface epithelium. They are thought to be molecularly distinct from other ovarian tumors but there have been no exome-level sequencing studies performed to date.MethodsTo understand the genetic etiology of mucinous ovarian tumors and assess the presence of novel therapeutic targets or pathways, we undertook exome sequencing of 24 tumors encompassing benign (5), borderline (8) and carcinoma (11) histologies and also assessed a validation cohort of 58 tumors for specific gene regions including exons 4–9 of TP53.ResultsThe predominant mutational signature was of C>T transitions in a NpCpG context, indicative of deamination of methyl-cytosines. As well as mutations in known drivers (KRAS, BRAF and CDKN2A), we identified a high percentage of carcinomas with TP53 mutations (52 %), and recurrent mutations in RNF43, ELF3, GNAS, ERBB3 and KLF5.ConclusionsThe diversity of mutational targets suggests multiple routes to tumorigenesis in this heterogeneous group of tumors that is generally distinct from other ovarian subtypes.


Cancer Research | 2016

Adenosine 2B Receptor Expression on Cancer Cells Promotes Metastasis

Deepak Mittal; Debottam Sinha; Deborah S. Barkauskas; Arabella Young; Murugan Kalimutho; Kimberley Stannard; Franco Caramia; Benjamin Haibe-Kains; John Stagg; Kum Kum Khanna; Sherene Loi; Mark J. Smyth

Adenosine plays an important role in inflammation and tumor development, progression, and responses to therapy. We show that an adenosine 2B receptor inhibitor (A2BRi) decreases both experimental and spontaneous metastasis and combines with chemotherapy or immune checkpoint inhibitors in mouse models of melanoma and triple-negative breast cancer (TNBC) metastasis. Decreased metastasis upon A2BR inhibition is independent of host A2BR and lymphocytes and myeloid cells. Knockdown of A2BR on mouse and human cancer cells reduces their metastasis in vivo and decreases their viability and colony-forming ability, while transiently delaying cell-cycle arrest in vitro The prometastatic activity of adenosine is partly tumor A2BR dependent and independent of host A2BR expression. In humans, TNBC cell lines express higher A2BR than luminal and Her2(+) breast cancer cell lines, and high expression of A2BR is associated with worse prognosis in TNBC. Collectively, high A2BR on mouse and human tumors promotes cancer metastasis and is an ideal candidate for therapeutic intervention. Cancer Res; 76(15); 4372-82. ©2016 AACR.


PLOS Medicine | 2016

The Subclonal Architecture of Metastatic Breast Cancer: Results from a Prospective Community-Based Rapid Autopsy Program "CASCADE"

Peter Savas; Zhi Ling Teo; Christophe Lefevre; Christoffer Flensburg; Franco Caramia; Kathryn Alsop; Mariam Mansour; Prudence A. Francis; Heather A. Thorne; Maria Joao Silva; Nnennaya Kanu; Michelle Dietzen; Andrew Rowan; Maik Kschischo; Stephen B. Fox; David Bowtell; Sarah-Jane Dawson; Terence P. Speed; Charles Swanton; Sherene Loi

Background Understanding the cancer genome is seen as a key step in improving outcomes for cancer patients. Genomic assays are emerging as a possible avenue to personalised medicine in breast cancer. However, evolution of the cancer genome during the natural history of breast cancer is largely unknown, as is the profile of disease at death. We sought to study in detail these aspects of advanced breast cancers that have resulted in lethal disease. Methods and Findings Three patients with oestrogen-receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer and one patient with triple negative breast cancer underwent rapid autopsy as part of an institutional prospective community-based rapid autopsy program (CASCADE). Cases represented a range of management problems in breast cancer, including late relapse after early stage disease, de novo metastatic disease, discordant disease response, and disease refractory to treatment. Between 5 and 12 metastatic sites were collected at autopsy together with available primary tumours and longitudinal metastatic biopsies taken during life. Samples underwent paired tumour-normal whole exome sequencing and single nucleotide polymorphism (SNP) arrays. Subclonal architectures were inferred by jointly analysing all samples from each patient. Mutations were validated using high depth amplicon sequencing. Between cases, there were significant differences in mutational burden, driver mutations, mutational processes, and copy number variation. Within each case, we found dramatic heterogeneity in subclonal structure from primary to metastatic disease and between metastatic sites, such that no single lesion captured the breadth of disease. Metastatic cross-seeding was found in each case, and treatment drove subclonal diversification. Subclones displayed parallel evolution of treatment resistance in some cases and apparent augmentation of key oncogenic drivers as an alternative resistance mechanism. We also observed the role of mutational processes in subclonal evolution. Limitations of this study include the potential for bias introduced by joint analysis of formalin-fixed archival specimens with fresh specimens and the difficulties in resolving subclones with whole exome sequencing. Other alterations that could define subclones such as structural variants or epigenetic modifications were not assessed. Conclusions This study highlights various mechanisms that shape the genome of metastatic breast cancer and the value of studying advanced disease in detail. Treatment drives significant genomic heterogeneity in breast cancers which has implications for disease monitoring and treatment selection in the personalised medicine paradigm.


PLOS ONE | 2014

Bioinformatics pipelines for targeted resequencing and whole-exome sequencing of human and mouse genomes: a virtual appliance approach for instant deployment.

Jason Li; Maria A. Doyle; Isaam Saeed; Stephen Q. Wong; Victoria Mar; David L. Goode; Franco Caramia; Ken Doig; Georgina L. Ryland; Ella R. Thompson; Sally M. Hunter; Saman K. Halgamuge; Jason Ellul; Alexander Dobrovic; Ian G. Campbell; Anthony T. Papenfuss; Grant A. McArthur; Richard W. Tothill

Targeted resequencing by massively parallel sequencing has become an effective and affordable way to survey small to large portions of the genome for genetic variation. Despite the rapid development in open source software for analysis of such data, the practical implementation of these tools through construction of sequencing analysis pipelines still remains a challenging and laborious activity, and a major hurdle for many small research and clinical laboratories. We developed TREVA (Targeted REsequencing Virtual Appliance), making pre-built pipelines immediately available as a virtual appliance. Based on virtual machine technologies, TREVA is a solution for rapid and efficient deployment of complex bioinformatics pipelines to laboratories of all sizes, enabling reproducible results. The analyses that are supported in TREVA include: somatic and germline single-nucleotide and insertion/deletion variant calling, copy number analysis, and cohort-based analyses such as pathway and significantly mutated genes analyses. TREVA is flexible and easy to use, and can be customised by Linux-based extensions if required. TREVA can also be deployed on the cloud (cloud computing), enabling instant access without investment overheads for additional hardware. TREVA is available at http://bioinformatics.petermac.org/treva/.


Cancer Research | 2017

Combined CDK4/6 and PI3Kα inhibition is synergistic and immunogenic in triple negative breast cancer

Zhi Ling Teo; Stephanie Versaci; Sathana Dushyanthen; Franco Caramia; Peter Savas; Chris P. Mintoff; Magnus Zethoven; Balaji Virassamy; Stephen J. Luen; Grant A. McArthur; Wayne A. Phillips; Phillip K. Darcy; Sherene Loi

New treatments for triple-negative breast cancer (TNBC) are urgently needed. Despite there being little evidence of clinical activity as single-agent therapies, we show that dual blockade of PI3Kα and CDK4/6 is synergistically effective against multiple RB1-wild-type TNBC models. Combined PI3Kα and CDK4/6 inhibition significantly increased apoptosis, cell-cycle arrest, and tumor immunogenicity and generated immunogenic cell death in human TNBC cell lines. Combination treatment also significantly improved disease control in human xenograft models compared with either monotherapy. Combined PI3Kα and CDK4/6 inhibition significantly increased tumor-infiltrating T-cell activation and cytotoxicity and decreased the frequency of immunosuppressive myeloid-derived suppressor cells in a syngeneic TNBC mouse model. Notably, combined PI3Kα and CDK4/6 inhibition, along with inhibition of immune checkpoints PD-1 and CTLA-4, induced complete and durable regressions (>1 year) of established TNBC tumors in vivo Overall, our results illustrate convergent mechanisms of PI3Kα and CDK4/6 blockade on cell-cycle progression, DNA damage response, and immune-modulation and may provide a novel therapeutic approach for TNBC. Cancer Res; 77(22); 6340-52. ©2017 AACR.


Genome Biology | 2014

Deciphering clonality in aneuploid breast tumors using SNP array and sequencing data

Ingrid Lönnstedt; Franco Caramia; Jason Li; Debora Fumagalli; Roberto Salgado; Andrew Rowan; Max Salm; Nnennaya Kanu; Peter Savas; Stuart Horswell; Stephan Gade; Sibylle Loibl; Patrick Neven; Christos Sotiriou; Charles Swanton; Sherene Loi; Terence P. Speed

Intra-tumor heterogeneity concerns the existence of genetically different subclones within the same tumor. Single sample quantification of heterogeneity relies on precise determination of chromosomal copy numbers throughout the genome, and an assessment of whether identified mutation variant allele fractions match clonal or subclonal copy numbers. We discuss these issues using data from SNP arrays, whole exome sequencing and pathologist purity estimates on several breast cancers characterized by ERBB2 amplification. We show that chromosomal copy numbers can only be estimated from SNP array signals or sequencing depths for subclonal tumor samples with simple subclonal architectures under certain assumptions.


Nature Medicine | 2018

Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis

Peter Savas; Balaji Virassamy; Chengzhong Ye; Agus Salim; Christopher P. Mintoff; Franco Caramia; Roberto Salgado; David J. Byrne; Zhi L. Teo; Sathana Dushyanthen; Ann Byrne; Lironne Wein; Stephen J. Luen; Catherine Poliness; Sophie S. Nightingale; Anita S. Skandarajah; David E. Gyorki; Chantel M. Thornton; Paul A. Beavis; Stephen B. Fox; Phillip K. Darcy; Terence P. Speed; Laura K. Mackay; Paul Neeson; Sherene Loi

The quantity of tumor-infiltrating lymphocytes (TILs) in breast cancer (BC) is a robust prognostic factor for improved patient survival, particularly in triple-negative and HER2-overexpressing BC subtypes1. Although T cells are the predominant TIL population2, the relationship between quantitative and qualitative differences in T cell subpopulations and patient prognosis remains unknown. We performed single-cell RNA sequencing (scRNA-seq) of 6,311 T cells isolated from human BCs and show that significant heterogeneity exists in the infiltrating T cell population. We demonstrate that BCs with a high number of TILs contained CD8+ T cells with features of tissue-resident memory T (TRM) cell differentiation and that these CD8+ TRM cells expressed high levels of immune checkpoint molecules and effector proteins. A CD8+ TRM gene signature developed from the scRNA-seq data was significantly associated with improved patient survival in early-stage triple-negative breast cancer (TNBC) and provided better prognostication than CD8 expression alone. Our data suggest that CD8+ TRM cells contribute to BC immunosurveillance and are the key targets of modulation by immune checkpoint inhibition. Further understanding of the development, maintenance and regulation of TRM cells will be crucial for successful immunotherapeutic development in BC.Extensive, high-dimensional characterization of T cells in breast cancer reveals activated TRM population and a gene signature associated with improved prognosis.


Cancer Research | 2016

Improved treatment of breast cancer with anti-HER2 therapy requires interleukin-21 signaling in CD8+ T cells

Deepak Mittal; Franco Caramia; Stefan Michiels; Heikki Joensuu; Pirkko-Liisa Kellokumpu-Lehtinen; Christos Sotiriou; Sherene Loi; Mark J. Smyth

The HER2/ErbB2 monoclonal antibody (mAb) trastuzumab is combined with chemotherapy as a standard-of-care for newly diagnosed HER2(+) breast cancer patients, but some patients treated with this combination therapy experience early relapse. Our analysis of data from a clinical trial evaluating the efficacy of chemotherapy plus/minus trastuzumab suggested that the magnitude of trastuzumab benefit on distant disease-free survival was higher for increasing expression of the IL21 receptor (IL21R). Therefore, we investigated a possible role for IL21 signaling in promoting HER2 mAb therapeutic efficacy. We found that IL21R-deficient mice and wild-type mice treated with a neutralizing anti-IL21 mAb were less susceptible to trastuzumab-like anti-ErbB2 therapy. Furthermore, IL21R expression on CD8(+) T cells, but not on natural killer cells, was required for optimal anti-ErbB2 mAb efficacy, and IL21 expression was enhanced in tumor-infiltrating CD4(+) T lymphocytes after anti-ErbB2 therapy. Finally, we found that administering recombinant IL21 in combination with anti-ErbB2 therapy was therapeutic against primary tumors and experimental metastases in mice. Collectively, our findings suggest that elevating IL21 signaling may enhance trastuzumab efficacy, thus constituting a novel candidate strategy to overcome trastuzumab resistance and improve patient survival. Cancer


Current Opinion in Oncology | 2014

Oncogene addiction and immunity: clinical implications of tumour infiltrating lymphocytes in breast cancers overexpressing the HER2/neu oncogene.

Peter Savas; Franco Caramia; Zhi Ling Teo; Sherene Loi

Purpose of review To review the evidence that correlates tumour infiltrating lymphocytes, a surrogate biomarker of pre-existing host antitumour immunity, and survival in HER2-overexpressing breast cancers. This is of particular relevance to developing immune biomarkers and harnessing new immunotherapeutics in this breast cancer subtype. Recent findings Oncogene addiction, in which cancer cells become reliant on a single oncogenic pathway for tumour growth and progression, has traditionally been thought of as a cell intrinsic characteristic. However, increasing evidence from multiple studies exploring the relationship between markers of an antitumour immune response and clinical outcome in HER2-overexpressing breast cancer points to the importance of a permissive microenvironment in oncogene-addicted tumours. Summary Characterizing the immune microenvironment in HER2-overexpressing breast cancer has the potential to furnish predictive and prognostic biomarkers that may be useful in routine clinical decision-making. The host-tumour immune interface is emerging as a key aspect of breast cancer biology that is likely to yield novel therapies in the near future.


Nature Communications | 2017

Agonist immunotherapy restores T cell function following MEK inhibition improving efficacy in breast cancer

Sathana Dushyanthen; Zhi Ling Teo; Franco Caramia; Peter Savas; Christopher P. Mintoff; Balaji Virassamy; Melissa A. Henderson; Stephen J. Luen; Mariam Mansour; Michael H. Kershaw; Joseph A. Trapani; Paul Neeson; Roberto Salgado; Grant A. McArthur; Justin M. Balko; Paul A. Beavis; Phillip K. Darcy; Sherene Loi

The presence of tumor-infiltrating lymphocytes in triple-negative breast cancers is correlated with improved outcomes. Ras/MAPK pathway activation is associated with significantly lower levels of tumor-infiltrating lymphocytes in triple-negative breast cancers and while MEK inhibition can promote recruitment of tumor-infiltrating lymphocytes to the tumor, here we show that MEK inhibition adversely affects early onset T-cell effector function. We show that α-4-1BB and α-OX-40 T-cell agonist antibodies can rescue the adverse effects of MEK inhibition on T cells in both mouse and human T cells, which results in augmented anti-tumor effects in vivo. This effect is dependent upon increased downstream p38/JNK pathway activation. Taken together, our data suggest that although Ras/MAPK pathway inhibition can increase tumor immunogenicity, the negative impact on T-cell activity is functionally important. This undesirable impact is effectively prevented by combination with T-cell immune agonist immunotherapies resulting in superior therapeutic efficacy.MEK inhibition in breast cancer is associated with increased tumour infiltrating lymphocytes (TILs), however, MAPK activity is required for T cells function. Here the authors show that TILs activity following MEK inhibition can be enhanced by agonist immunotherapy resulting in synergic therapeutic effects.

Collaboration


Dive into the Franco Caramia's collaboration.

Top Co-Authors

Avatar

Sherene Loi

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Peter Savas

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Paul Neeson

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Terence P. Speed

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ygal Haupt

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Jason Li

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Phillip K. Darcy

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Stephen B. Fox

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Christos Sotiriou

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Roberto Salgado

Université libre de Bruxelles

View shared research outputs
Researchain Logo
Decentralizing Knowledge