Network


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

Hotspot


Dive into the research topics where Venessa T. Chin is active.

Publication


Featured researches published by Venessa T. Chin.


Nature | 2015

Whole genomes redefine the mutational landscape of pancreatic cancer

Nicola Waddell; Marina Pajic; Ann-Marie Patch; David K. Chang; Karin S. Kassahn; Peter Bailey; Amber L. Johns; David Miller; Katia Nones; Kelly Quek; Michael Quinn; Alan Robertson; Muhammad Z.H. Fadlullah; Timothy J. C. Bruxner; Angelika N. Christ; Ivon Harliwong; Senel Idrisoglu; Suzanne Manning; Craig Nourse; Ehsan Nourbakhsh; Shivangi Wani; Peter J. Wilson; Emma Markham; Nicole Cloonan; Matthew J. Anderson; J. Lynn Fink; Oliver Holmes; Stephen Kazakoff; Conrad Leonard; Felicity Newell

Pancreatic cancer remains one of the most lethal of malignancies and a major health burden. We performed whole-genome sequencing and copy number variation (CNV) analysis of 100 pancreatic ductal adenocarcinomas (PDACs). Chromosomal rearrangements leading to gene disruption were prevalent, affecting genes known to be important in pancreatic cancer (TP53, SMAD4, CDKN2A, ARID1A and ROBO2) and new candidate drivers of pancreatic carcinogenesis (KDM6A and PREX2). Patterns of structural variation (variation in chromosomal structure) classified PDACs into 4 subtypes with potential clinical utility: the subtypes were termed stable, locally rearranged, scattered and unstable. A significant proportion harboured focal amplifications, many of which contained druggable oncogenes (ERBB2, MET, FGFR1, CDK6, PIK3R3 and PIK3CA), but at low individual patient prevalence. Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency. Of 8 patients who received platinum therapy, 4 of 5 individuals with these measures of defective DNA maintenance responded.


Annals of Oncology | 2012

The prognostic and predictive value of serum CA19.9 in pancreatic cancer

Jeremy L. Humphris; David K. Chang; Amber L. Johns; Christopher J. Scarlett; Marina Pajic; Marc D. Jones; Emily K. Colvin; Adnan Nagrial; Venessa T. Chin; Lorraine A. Chantrill; Jaswinder S. Samra; Anthony J. Gill; James G. Kench; Neil D. Merrett; Amitabha Das; Elizabeth A. Musgrove; Robert L. Sutherland; Andrew V. Biankin

Background Current staging methods for pancreatic cancer (PC) are inadequate, and biomarkers to aid clinical decision making are lacking. Despite the availability of the serum marker carbohydrate antigen 19.9 (CA19.9) for over two decades, its precise role in the management of PC is yet to be defined, and as a consequence, it is not widely used. Methods We assessed the relationship between perioperative serum CA19.9 levels, survival and adjuvant chemotherapeutic responsiveness in a cohort of 260 patients who underwent operative resection for PC. Results By specifically assessing the subgroup of patients with detectable CA19.9, we identified potential utility at key clinical decision points. Low postoperative CA19.9 at 3 months (median survival 25.6 vs 14.8 months, P = 0.0052) and before adjuvant chemotherapy were independent prognostic factors. Patients with postoperative CA 19.9 levels >90 U/ml did not benefit from adjuvant chemotherapy (P = 0.7194) compared with those with a CA19.9 of ≤90 U/ml (median 26.0 vs 16.7 months, P = 0.0108). Normalization of CA19.9 within 6 months of resection was also an independent favorable prognostic factor (median 29.9 vs 14.8 months, P = 0.0004) and normal perioperative CA19.9 levels identified a good prognostic group, which was associated with a 5-year survival of 42%. Conclusions Perioperative serum CA19.9 measurements are informative in patients with detectable CA19.9 (defined by serum levels of >5 U/ml) and have potential clinical utility in predicting outcome and response to adjuvant chemotherapy. Future clinical trials should prioritize incorporation of CA19.9 measurement at key decision points to prospectively validate these findings and facilitate implementation.


Clinical Cancer Research | 2015

Precision Medicine for Advanced Pancreas Cancer: The Individualized Molecular Pancreatic Cancer Therapy (IMPaCT) Trial

Lorraine A. Chantrill; Adnan Nagrial; Clare Watson; Amber L. Johns; Mona Martyn-Smith; Skye Simpson; Scott Mead; Marc D. Jones; Jaswinder S. Samra; Anthony J. Gill; Nicole Watson; Venessa T. Chin; Jeremy L. Humphris; Angela Chou; Belinda Brown; Adrienne Morey; Marina Pajic; Sean M. Grimmond; David K. Chang; David Thomas; Lucille Sebastian; Katrin Marie Sjoquist; Sonia Yip; Nick Pavlakis; Ray Asghari; Sandra Harvey; Peter Grimison; John Simes; Andrew V. Biankin

Purpose: Personalized medicine strategies using genomic profiling are particularly pertinent for pancreas cancer. The Individualized Molecular Pancreatic Cancer Therapy (IMPaCT) trial was initially designed to exploit results from genome sequencing of pancreatic cancer under the auspices of the International Cancer Genome Consortium (ICGC) in Australia. Sequencing revealed small subsets of patients with aberrations in their tumor genome that could be targeted with currently available therapies. Experimental Design: The pilot stage of the IMPaCT trial assessed the feasibility of acquiring suitable tumor specimens for molecular analysis and returning high-quality actionable genomic data within a clinically acceptable timeframe. We screened for three molecular targets: HER2 amplification; KRAS wild-type; and mutations in DNA damage repair pathways (BRCA1, BRCA2, PALB2, ATM). Results: Tumor biopsy and archived tumor samples were collected from 93 patients and 76 were screened. To date 22 candidate cases have been identified: 14 KRAS wild-type, 5 cases of HER2 amplification, 2 mutations in BRCA2, and 1 ATM mutation. Median time from consent to the return of validated results was 21.5 days. An inability to obtain a biopsy or insufficient tumor content in the available specimen were common reasons for patient exclusion from molecular analysis while deteriorating performance status prohibited a number of patients from proceeding in the study. Conclusions: Documenting the feasibility of acquiring and screening biospecimens for actionable molecular targets in real time will aid other groups embarking on similar trials. Key elements include the need to better prescreen patients, screen more patients, and offer more attractive clinical trial options. Clin Cancer Res; 21(9); 2029–37. ©2015 AACR.


Journal of Clinical Oncology | 2013

Histomolecular Phenotypes and Outcome in Adenocarcinoma of the Ampulla of Vater

David K. Chang; Nigel B. Jamieson; Amber L. Johns; Christopher J. Scarlett; Marina Pajic; Angela Chou; Mark Pinese; Jeremy L. Humphris; Marc D. Jones; Christopher W. Toon; Adnan Nagrial; Lorraine A. Chantrill; Venessa T. Chin; Andreia V. Pinho; Ilse Rooman; Mark J. Cowley; Jianmin Wu; R. Scott Mead; Emily K. Colvin; Jaswinder S. Samra; Vincenzo Corbo; Claudio Bassi; Massimo Falconi; Rita T. Lawlor; Stefano Crippa; Nicola Sperandio; Samantha Bersani; Euan J. Dickson; Mohamed Mohamed; Karin A. Oien

PURPOSE Individuals with adenocarcinoma of the ampulla of Vater demonstrate a broad range of outcomes, presumably because these cancers may arise from any one of the three epithelia that converge at that location. This variability poses challenges for clinical decision making and the development of novel therapeutic strategies. PATIENTS AND METHODS We assessed the potential clinical utility of histomolecular phenotypes defined using a combination of histopathology and protein expression (CDX2 and MUC1) in 208 patients from three independent cohorts who underwent surgical resection for adenocarcinoma of the ampulla of Vater. RESULTS Histologic subtype and CDX2 and MUC1 expression were significant prognostic variables. Patients with a histomolecular pancreaticobiliary phenotype (CDX2 negative, MUC1 positive) segregated into a poor prognostic group in the training (hazard ratio [HR], 3.34; 95% CI, 1.69 to 6.62; P < .001) and both validation cohorts (HR, 5.65; 95% CI, 2.77 to 11.5; P < .001 and HR, 2.78; 95% CI, 1.25 to 7.17; P = .0119) compared with histomolecular nonpancreaticobiliary carcinomas. Further stratification by lymph node (LN) status defined three clinically relevant subgroups: one, patients with histomolecular nonpancreaticobiliary (intestinal) carcinoma without LN metastases who had an excellent prognosis; two, those with histomolecular pancreaticobiliary carcinoma with LN metastases who had a poor outcome; and three, the remainder of patients (nonpancreaticobiliary, LN positive or pancreaticobiliary, LN negative) who had an intermediate outcome. CONCLUSION Histopathologic and molecular criteria combine to define clinically relevant histomolecular phenotypes of adenocarcinoma of the ampulla of Vater and potentially represent distinct diseases with significant implications for current therapeutic strategies, the ability to interpret past clinical trials, and future trial design.


Genome Medicine | 2013

Clinical and molecular characterization of HER2 amplified-pancreatic cancer

Angela Chou; Nicola Waddell; Mark J. Cowley; Anthony J. Gill; David K. Chang; Ann-Marie Patch; Katia Nones; Jianmin Wu; Mark Pinese; Amber L. Johns; David Miller; Karin S. Kassahn; Adnan Nagrial; Harpreet Wasan; David Goldstein; Christopher W. Toon; Venessa T. Chin; Lorraine A. Chantrill; Jeremy L. Humphris; R. Scott Mead; Ilse Rooman; Jaswinder S. Samra; Marina Pajic; Elizabeth A. Musgrove; John V. Pearson; Adrienne Morey; Sean M. Grimmond; Andrew V. Biankin

BackgroundPancreatic cancer is one of the most lethal and molecularly diverse malignancies. Repurposing of therapeutics that target specific molecular mechanisms in different disease types offers potential for rapid improvements in outcome. Although HER2 amplification occurs in pancreatic cancer, it is inadequately characterized to exploit the potential of anti-HER2 therapies.MethodsHER2 amplification was detected and further analyzed using multiple genomic sequencing approaches. Standardized reference laboratory assays defined HER2 amplification in a large cohort of patients (n = 469) with pancreatic ductal adenocarcinoma (PDAC).ResultsAn amplified inversion event (1 MB) was identified at the HER2 locus in a patient with PDAC. Using standardized laboratory assays, we established diagnostic criteria for HER2 amplification in PDAC, and observed a prevalence of 2%. Clinically, HER2- amplified PDAC was characterized by a lack of liver metastases, and a preponderance of lung and brain metastases. Excluding breast and gastric cancer, the incidence of HER2-amplified cancers in the USA is >22,000 per annum.ConclusionsHER2 amplification occurs in 2% of PDAC, and has distinct features with implications for clinical practice. The molecular heterogeneity of PDAC implies that even an incidence of 2% represents an attractive target for anti-HER2 therapies, as options for PDAC are limited. Recruiting patients based on HER2 amplification, rather than organ of origin, could make trials of anti-HER2 therapies feasible in less common cancer types.


Science Translational Medicine | 2017

Transient tissue priming via ROCK inhibition uncouples pancreatic cancer progression, sensitivity to chemotherapy, and metastasis

Claire Vennin; Venessa T. Chin; Sean C. Warren; Morghan C. Lucas; David Herrmann; Astrid Magenau; Pauline Mélénec; Stacey N. Walters; Gonzalo del Monte-Nieto; James R.W. Conway; Max Nobis; Amr H. Allam; Rachael A. McCloy; Nicola Currey; Mark Pinese; Alice Boulghourjian; Anaiis Zaratzian; Arne A. S. Adam; Celine Heu; Adnan Nagrial; Angela Chou; Angela Steinmann; Alison Drury; Danielle Froio; Marc Giry-Laterriere; Nathanial L. E. Harris; Tri Giang Phan; Rohit Jain; Wolfgang Weninger; Ewan J. McGhee

Fine-tuned manipulation of tumor tension and vasculature enhances response to chemotherapy and impairs metastatic spread in pancreatic cancer. ROCK-ing pancreatic cancer to the core Pancreatic cancer, one of the most deadly and difficult-to-treat tumor types in patients, usually has a dense stroma that can be difficult for drugs to penetrate. Stromal characteristics can also affect multiple other aspects of tumor biology, including metastatic spread, vascular supply, and immune response. Vennin et al. used Fasudil, a drug that inhibits a protein called ROCK and is already used for some conditions in people, to demonstrate the feasibility including short-term tumor stroma remodeling as part of cancer treatment. In genetically engineered and patient-derived mouse models of pancreatic cancer, priming with Fasudil disrupted the tumors’ extracellular matrix and improved the effectiveness of subsequent treatment with standard-of-care chemotherapy for this disease. The emerging standard of care for patients with inoperable pancreatic cancer is a combination of cytotoxic drugs gemcitabine and Abraxane, but patient response remains moderate. Pancreatic cancer development and metastasis occur in complex settings, with reciprocal feedback from microenvironmental cues influencing both disease progression and drug response. Little is known about how sequential dual targeting of tumor tissue tension and vasculature before chemotherapy can affect tumor response. We used intravital imaging to assess how transient manipulation of the tumor tissue, or “priming,” using the pharmaceutical Rho kinase inhibitor Fasudil affects response to chemotherapy. Intravital Förster resonance energy transfer imaging of a cyclin-dependent kinase 1 biosensor to monitor the efficacy of cytotoxic drugs revealed that priming improves pancreatic cancer response to gemcitabine/Abraxane at both primary and secondary sites. Transient priming also sensitized cells to shear stress and impaired colonization efficiency and fibrotic niche remodeling within the liver, three important features of cancer spread. Last, we demonstrate a graded response to priming in stratified patient-derived tumors, indicating that fine-tuned tissue manipulation before chemotherapy may offer opportunities in both primary and metastatic targeting of pancreatic cancer.


Gastroenterology | 2017

Hypermutation In Pancreatic Cancer

Jeremy L. Humphris; Ann-Marie Patch; Katia Nones; Peter Bailey; Amber L. Johns; Skye McKay; David K. Chang; David Miller; Marina Pajic; Karin S. Kassahn; Michael Quinn; Timothy J. C. Bruxner; Angelika N. Christ; Ivon Harliwong; Senel Idrisoglu; Suzanne Manning; Craig Nourse; Ehsan Nourbakhsh; Andrew Stone; Peter J. Wilson; Matthew Anderson; J. Lynn Fink; Oliver Holmes; Stephen Kazakoff; Conrad Leonard; Felicity Newell; Nick Waddell; Scott Wood; Ronald S. Mead; Qinying Xu

Pancreatic cancer is molecularly diverse, with few effective therapies. Increased mutation burden and defective DNA repair are associated with response to immune checkpoint inhibitors in several other cancer types. We interrogated 385 pancreatic cancer genomes to define hypermutation and its causes. Mutational signatures inferring defects in DNA repair were enriched in those with the highest mutation burdens. Mismatch repair deficiency was identified in 1% of tumors harboring different mechanisms of somatic inactivation of MLH1 and MSH2. Defining mutation load in individual pancreatic cancers and the optimal assay for patient selection may inform clinical trial design for immunotherapy in pancreatic cancer.


British Journal of Cancer | 2014

Adjuvant chemotherapy in elderly patients with pancreatic cancer

Adnan Nagrial; David K. Chang; Nam Q. Nguyen; Amber L. Johns; Lorraine A. Chantrill; Jeremy L. Humphris; Venessa T. Chin; Jaswinder S. Samra; Anthony J. Gill; Marina Pajic; Mark Pinese; Emily K. Colvin; Christopher J. Scarlett; Angela Chou; James G. Kench; Robert L. Sutherland; Lisa G. Horvath; Andrew V. Biankin

Background:Adjuvant chemotherapy improves survival for patients with resected pancreatic cancer. Elderly patients are under-represented in Phase III clinical trials, and as a consequence the efficacy of adjuvant therapy in older patients with pancreatic cancer is not clear. We aimed to assess the use and efficacy of adjuvant chemotherapy in older patients with pancreatic cancer.Methods:We assessed a community cohort of 439 patients with a diagnosis of pancreatic ductal adenocarcinoma who underwent operative resection in centres associated with the Australian Pancreatic Cancer Genome Initiative.Results:The median age of the cohort was 67 years. Overall only 47% of all patients received adjuvant therapy. Patients who received adjuvant chemotherapy were predominantly younger, had later stage disease, more lymph node involvement and more evidence of perineural invasion than the group that did not receive adjuvant treatment. Overall, adjuvant chemotherapy was associated with prolonged survival (median 22.1 vs 15.8 months; P<0.0001). Older patients (aged ⩾70) were less likely to receive adjuvant chemotherapy (51.5% vs 29.8%; P<0.0001). Older patients had a particularly poor outcome when adjuvant therapy was not delivered (median survival=13.1 months; HR 1.89, 95% CI: 1.27–2.78, P=0.002).Conclusion:Patients aged ⩾70 are less likely to receive adjuvant therapy although it is associated with improved outcome. Increased use of adjuvant therapy in older individuals is encouraged as they constitute a large proportion of patients with pancreatic cancer.


Cancer | 2014

Clinical and pathologic features of familial pancreatic cancer

Jeremy L. Humphris; Amber L. Johns; Skye H. Simpson; Mark J. Cowley; Marina Pajic; David K. Chang; Adnan Nagrial; Venessa T. Chin; Lorraine A. Chantrill; Mark Pinese; R. Scott Mead; Anthony J. Gill; Jaswinder S. Samra; James G. Kench; Elizabeth A. Musgrove; Katherine M. Tucker; Allan D. Spigelman; Nic Waddell; Sean M. Grimmond; Andrew V. Biankin

Inherited predisposition to pancreatic cancer contributes significantly to its incidence and presents an opportunity for the development of early detection strategies. The genetic basis of predisposition remains unexplained in a high proportion of patients with familial PC (FPC).


Small GTPases | 2015

The dynamics of Rho GTPase signaling and implications for targeting cancer and the tumor microenvironment

Marina Pajic; David Herrmann; Claire Vennin; James R.W. Conway; Venessa T. Chin; Anna-Karin E. Johnsson; Heidi C. E. Welch; Paul Timpson

Numerous large scale genomics studies have demonstrated that cancer is a molecularly heterogeneous disease, characterized by acquired changes in the structure and DNA sequence of tumor genomes. More recently, the role of the equally complex tumor microenvironment in driving the aggressiveness of this disease is increasingly being realized. Tumor cells are surrounded by activated stroma, creating a dynamic environment that promotes cancer development, metastasis and chemoresistance. The Rho family of small GTPases plays an essential role in the regulation of cell shape, cytokinesis, cell adhesion, and cell motility. Importantly, these processes need to be considered in the context of a complex 3-dimensional (3D) environment, with reciprocal feedback and cross-talk taking place between the tumor cells and host environment. Here we discuss the role of molecular networks involving Rho GTPases in cancer, and the therapeutic implications of inhibiting Rho signaling in both cancer cells and the emerging concept of targeting the surrounding stroma.

Collaboration


Dive into the Venessa T. Chin's collaboration.

Top Co-Authors

Avatar

Adnan Nagrial

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Lorraine A. Chantrill

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amber L. Johns

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Marina Pajic

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Angela Chou

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Anthony J. Gill

Kolling Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark Pinese

Garvan Institute of Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge