Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 20
  • Item
    Thumbnail Image
    Precision oncology using a clinician-directed, tailored approach to molecular profiling
    Lam, M ; Tran, B ; Beck, S ; Tie, J ; Herath, D ; Whittle, J ; Kwan, EM ; Fox, SB ; Fellowes, A ; Ananda, S ; Lipton, L ; Gibbs, P ; Rosenthal, MA ; Desai, J (WILEY, 2018-02)
    AIM: Precision oncology involves molecularly matching patients to targeted agents usually in early drug development (EDD) programs. Molecular profiling (MP) identifies actionable targets. Comprehensive commercial MP platforms are costly and in resource limited environments, a more practical approach to MP is necessary to support EDD and precision oncology. We adopted a clinician-directed, tailored approach to MP to enrol patients onto molecularly targeted trials. We report the feasibility of this approach. METHODS: All patients referred to the Royal Melbourne Hospital (RMH) EDD between September 2013 and September 2015 were identified in a prospective database. Key captured data included clinicopathological data, MP platform ordered (if any), molecular targets identified and subsequent enrolment onto clinical trials. EDD-clinician decisions to order MP and the platform utilized was guided by patient consultation, tumor type, trial availability and requirement for molecular information. RESULTS: We identified 377 patients referred to RMH EDD. A total of 216 (57%) had MP ordered. The remainder had known actionable targets (19%), or were inappropriate for clinical trials (24%). In those undergoing MP, 187 genetic aberrations were found in 113 patients with 98 considered actionable targets in 86 patients. Ninety-eight (25%) patients were enrolled onto a clinical trial, including 40 (11%) receiving molecularly matched treatments. Median progression-free survival was improved in patients enrolled onto molecularly matched trials compared to those on unmatched trials (3.6 months vs 1.9 months, HR 0.58 [0.38-0.89], P  =  0.013). CONCLUSION: A clinician-directed, tailored approach to the use of MP is feasible, resulting in 11% of patients enrolled onto molecularly matched trials.
  • Item
    Thumbnail Image
    Wither surgical oncology?
    Pham, TD ; Narasimhan, V ; Guerra, G ; Kong, J ; Desai, J ; Ramsay, R ; Heriot, A (WILEY, 2019-01)
  • Item
    Thumbnail Image
    Treatment of patients with primary retroperitoneal sarcoma: predictors of outcome from an Australian specialist sarcoma centre
    Snow, HA ; Hitchen, TX ; Head, J ; Herschtal, A ; Bae, S ; Chander, S ; Chu, J ; Hendry, S ; Ngan, SY ; Desai, J ; Choong, PFM ; Henderson, M ; Gyorki, DE (WILEY, 2018-11)
    BACKGROUND: Several unanswered questions surround the management of retroperitoneal sarcoma (RPS). Guidelines recommend treatment by a multidisciplinary team at a specialized referral centre. The objective of this study was to describe the management of RPS at an Australian specialist sarcoma centre, comparing outcomes to international standards and analysing for predictors of local failure. METHODS: A retrospective review of a prospectively maintained database was performed on patients with RPS treated between 2008 and 2016. A 5-year outcome analyses focussed on patients undergoing curative-intent surgery for primary, non-metastatic RPS. RESULTS: Eighty-eight patients underwent surgery for primary RPS. Five-year overall survival was 66%, 5-year freedom from local recurrence was 65% and 5-year freedom from distant metastasis was 71%. Overall survival was associated with tumour grade (hazard ratio (HR) 6.1, P < 0.001) and histologic organ invasion (HR 5.7, P < 0.001). Variables associated with improved freedom from local recurrence were macroscopically complete resection (HR 0.14, P < 0.001) and neoadjuvant radiotherapy (HR 0.33, P = 0.014). Treatment at a specialist sarcoma centre was associated with a higher rate of preoperative biopsy and neoadjuvant radiotherapy (both with P < 0.001). There was a trend towards improved local control for patients undergoing surgery at a specialist centre (P = 0.055). CONCLUSION: This is the largest Australian series of RPS and outcomes are comparable to major international sarcoma centres. Patients treated at a specialist centre had higher rates of preoperative diagnosis and tailored therapy which was associated with improved outcomes. Patients with suspected RPS should be referred to a specialist centre for optimal preoperative evaluation and multidisciplinary management.
  • Item
    No Preview Available
    KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors
    Hong, DS ; Fakih, MG ; Strickler, JH ; Desai, J ; Durm, GA ; Shapiro, GI ; Falchook, GS ; Price, TJ ; Sacher, A ; Denlinger, CS ; Bang, Y-J ; Dy, GK ; Krauss, JC ; Kuboki, Y ; Kuo, JC ; Coveler, AL ; Park, K ; Kim, TW ; Barlesi, F ; Munster, PN ; Ramalingam, SS ; Burns, TF ; Meric-Bernstam, F ; Henary, H ; Ngang, J ; Ngarmchamnanrith, G ; Kim, J ; Houk, BE ; Canon, J ; Lipford, JR ; Friberg, G ; Lito, P ; Govindan, R ; Li, BT (MASSACHUSETTS MEDICAL SOC, 2020-09-24)
    BACKGROUND: No therapies for targeting KRAS mutations in cancer have been approved. The KRAS p.G12C mutation occurs in 13% of non-small-cell lung cancers (NSCLCs) and in 1 to 3% of colorectal cancers and other cancers. Sotorasib is a small molecule that selectively and irreversibly targets KRASG12C. METHODS: We conducted a phase 1 trial of sotorasib in patients with advanced solid tumors harboring the KRAS p.G12C mutation. Patients received sotorasib orally once daily. The primary end point was safety. Key secondary end points were pharmacokinetics and objective response, as assessed according to Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1. RESULTS: A total of 129 patients (59 with NSCLC, 42 with colorectal cancer, and 28 with other tumors) were included in dose escalation and expansion cohorts. Patients had received a median of 3 (range, 0 to 11) previous lines of anticancer therapies for metastatic disease. No dose-limiting toxic effects or treatment-related deaths were observed. A total of 73 patients (56.6%) had treatment-related adverse events; 15 patients (11.6%) had grade 3 or 4 events. In the subgroup with NSCLC, 32.2% (19 patients) had a confirmed objective response (complete or partial response) and 88.1% (52 patients) had disease control (objective response or stable disease); the median progression-free survival was 6.3 months (range, 0.0+ to 14.9 [with + indicating that the value includes patient data that were censored at data cutoff]). In the subgroup with colorectal cancer, 7.1% (3 patients) had a confirmed response, and 73.8% (31 patients) had disease control; the median progression-free survival was 4.0 months (range, 0.0+ to 11.1+). Responses were also observed in patients with pancreatic, endometrial, and appendiceal cancers and melanoma. CONCLUSIONS: Sotorasib showed encouraging anticancer activity in patients with heavily pretreated advanced solid tumors harboring the KRAS p.G12C mutation. Grade 3 or 4 treatment-related toxic effects occurred in 11.6% of the patients. (Funded by Amgen and others; CodeBreaK100 ClinicalTrials.gov number, NCT03600883.).
  • Item
    Thumbnail Image
    POTENTIAL MECHANISMS OF RESISTANCE IDENTIFIED THROUGH ANALYSIS OF MULTIPLE BIOMARKERS IN IMMUNE HOT NON-RESPONDERS WITH NON-SMALL CELL LUNG CANCER (NSCLC) TREATED WITH TISLELIZUMAB
    Desai, J ; Zhou, Q ; Deva, S ; Zhao, J ; Wang, J ; Tan, W ; Ma, X ; Zhang, Y ; Shen, Z ; Wu, X ; Leaw, S ; Zhang, J ; Wu, Y-L (BMJ PUBLISHING GROUP, 2020-11)
    Background Tislelizumab, an anti-PD-1 monoclonal antibody, has demonstrated clinical benefit for patients with NSCLC. The underlying response and resistance mechanisms to tislelizumab treatment remain unknown. Methods Baseline tumor samples from 59 nonsquamous (NSQ) and 41 squamous (SQ) NSCLC patients treated with tislelizumab monotherapy (NCT02407990 and NCT04068519) were tested for gene mutations using large panel next generation sequencing and RNA expression using gene expression profiling (GEP; Precision Immuno-Oncology Panel, HTG Molecular Diagnostics). GEP analyses of NSQ and SQ NSCLC were performed separately due to different gene expression patterns. Results The ORR, mPFS, and mOS in this pooled NSCLC cohort were 15.2% (95% CI: 9.0, 23.6), 4.1 months (95% CI: 2.20, 6.11), and 15.1 months (95% CI: 11.20, NE), respectively, with a median study follow-up of 15.3 months (95% CI: 14.06, 15.90). Non-responders (NRs) exhibited distinct tumor and immune gene signature profiles and could be clustered into two subgroups: NR1 and NR2. Compared with responders, NR1 had elevated cell cycle signatures in both NSQ (P=0.2) and SQ (P=0.03) cohorts, and a trend of decreased inflamed gene signature profiles. However, NR2 showed comparable or even higher tumor inflammation (18-gene), and CD8+ T-cell signature scores in both NSQ and SQ cohorts and could be classified as immune hot. To explore the resistance mechanisms of immune hot NRs, differentially expressed gene analyses between immune hot NR2 and responders were performed. M2 macrophage and Treg signature scores were higher in NR2 in both NSQ (M2, P=0.05; Treg, P=0.03) and SQ (M2, P=0.05 [subgroup of NR2]; Treg, P=0.03) cohorts; significantly higher expression of immune regulatory genes included PIK3CD, CCR2, CD244, IRAK3, and MAP4K1 (P<0.05) in NSQ and PIK3CD, CCR2, CD40, CD163, and MMP12 (P<0.05) in SQ. Significantly higher epithelial–mesenchymal transition (EMT) and angiogenesis gene expression, including SNAI1, FAP, VEGFC, and TEK (P<0.05) genes, were also observed in SQ NR2. Moreover, gene mutation analysis identified seven immune hot NR patients harboring either driver mutations (RET fusion, ROS1 fusion, BRAF, and PIK3CA amp) or well-established resistance mutations (loss of function mutation in JAK2, STK11, and MDM2 amplification). Conclusions Despite the presence of immune hot features, a subgroup of tislelizumab NRs with NSCLC were identified. High levels of immune suppressive factors, such as M2 macrophage and Treg signatures, angiogenesis, and EMT genes, as well as the existence of driver/resistance mutations, may indicate mechanisms of resistance of immune hot NRs, highlighting potential novel treatment targets. Acknowledgements Editorial assistance was provided by Agnieszka Laskowski, PhD, and Elizabeth Hermans, PhD (OPEN Health Medical Communications, Chicago, IL), and funded by the study sponsor. Trial Registration NCT02407990 and NCT04068519
  • Item
    Thumbnail Image
    A PHASE II STUDY OF CONTINUOUS LOW DOSE PANOBINOSTAT IN PAEDIATRIC PATIENTS WITH MALIGNANT RHABDOID TUMORS/ATYPICAL TERATOID RHABDOID TUMORS
    Wood, P ; Desai, J ; Waldeck, K ; Cain, J ; Gottardo, N ; Strong, R ; Kinross, K ; Carr, M ; Jones, J ; Wong, L ; Ziegler, D ; Hansford, J ; Michael, M ; Ashley, D (OXFORD UNIV PRESS INC, 2020-12)
    Abstract

    BACKGROUND

    Panobinostat treatment has been shown to terminally differentiate malignant rhabdoid tumor (MRT)/atypical teratoid rhabdoid tumors (ATRT) in pre-clinical models. This is an open label, phase II study of panobinostat in patients with newly diagnosed or relapsed MRT/ATRT. AIMS: To assess the anti-tumor activity of low dose, continuous panobinostat, its associated toxicities, the biological activity of low dose panobinostat by measuring histone acetylation status in peripheral mononuclear cells (PMNC), and markers of differentiation in fresh tumor tissue specimens.

    METHODS

    Following cycles of induction and consolidation chemotherapy and/or radiation treatment, patients were enrolled and commenced on panobinostat as a continuous daily oral dose starting at 10mg/m2 following a three-week wash out period between therapies. Real-time acetylation status, measuring acetylated H4 on PMNC, was performed to determine the pharmacodynamics of panobinostat. Patients were monitored for drug toxicities with the possibility of dose reductions in decrements of 2mg/m2.

    RESULTS

    Six patients with newly diagnosed ATRT/MRT and one patient with relapsed MRT have been enrolled to date. The average age at enrollment was 2.5 years. Currently, six patients (85.7%) remain on study with a mean treatment duration of 170 days (range 44–327 days). One patient was removed from study at day 44 due to disease progression. The main dose-limiting toxicity observed to date has been myelosuppression. Panobinostat, at a dose of 10mg/m2, caused significant acetylation of H4 in PMNC.

    CONCLUSIONS

    Treatment with panobinostat appears to be well tolerated in infants with MRT/ATRT, with successful real-time pharmacodynamic assessment of H4 acetylation.
  • Item
    Thumbnail Image
    New biomarkers for checkpoint inhibitor therapy
    Burdett, N ; Desai, J (ELSEVIER, 2020-03)
    Immune checkpoint inhibitor blockade has vastly changed treatment paradigms and improved outcomes of many solid organ malignancies. The achievements of the last decade have transformed the outcomes of several tumour types, most notably metastatic melanoma. There are, however, still large numbers of patients who receive checkpoint inhibitor therapy and do not respond. In addition to potential lack of efficacy, checkpoint inhibitors also come with a unique and sometimes devastating side-effect profile. There exists a strong need for biomarkers to accurately predict response, improve treatment selection and avoid exposing patients to toxicity where there is minimal likelihood of response. There is a wide range of methodologies investigating predictive biomarkers in this space; in this review, we address the major putative biomarkers of interest. These include conventional serum tests such as lymphocyte indices and lactate dehydrogenase, and more novel research markers such as interleukin-6 and T receptor clonality. We discuss tumorous factors that may be of interest in certain tumour types, and finally gene expression profiling. Significant research continues into many of these potential predictive biomarkers in response to the emergent need to better select patients who will benefit from treatment.
  • Item
    Thumbnail Image
    Therapeutic strategies to remodel immunologically cold tumors
    Wang, M ; Wang, S ; Desai, J ; Trapani, JA ; Neeson, PJ (WILEY, 2020)
    Immune checkpoint inhibitors (ICIs) induce a durable response in a wide range of tumor types, but only a minority of patients outside these 'responsive' tumor types respond, with some totally resistant. The primary predictor of intrinsic immune resistance to ICIs is the complete or near-complete absence of lymphocytes from the tumor, so-called immunologically cold tumors. Here, we propose two broad approaches to convert 'cold' tumors into 'hot' tumors. The first is to induce immunogenic tumor cell death, through the use of oncolytic viruses or bacteria, conventional cancer therapies (e.g. chemotherapy or radiation therapy) or small molecule drugs. The second approach is to target the tumor microenvironment, and covers diverse options such as depleting immune suppressive cells; inhibiting transforming growth factor-beta; remodelling the tumor vasculature or hypoxic environment; strengthening the infiltration and activation of antigen-presenting cells and/or effector T cells in the tumor microenvironment with immune modulators; and enhancing immunogenicity through personalised cancer vaccines. Strategies that successfully modify cold tumors to overcome their resistance to ICIs represent mechanistically driven approaches that will ultimately result in rational combination therapies to extend the clinical benefits of immunotherapy to a broader cancer cohort.
  • Item
    Thumbnail Image
    Survival in early phase immuno-oncology trials: Development and validation of a prognostic index
    Day, D ; Guo, C ; Kanjanapan, Y ; Tran, B ; Spreafico, A ; Joshua, AM ; Wang, L ; Abdul Razak, AR ; Leighl, NB ; Hansen, AR ; Butler, MO ; Siu, LL ; Desai, J ; Bedard, PL (Oxford University Press, 2019-12-01)
    Background: Immuno-oncology (IO) is rapidly evolving in early drug development. We aimed to develop and prospectively validate a prognostic index for patients treated in IO phase I trials to assist with patient selection. Methods: The development cohort included 192 advanced solid tumor patients treated in 13 IO phase I trials, targeting immune checkpoint and/or co-stimulatory molecules. A prognostic scoring system was developed from multivariate survival analysis of 10 clinical factors, and subsequently validated in two independent validation cohorts (n = 152 and n = 80). Results: In the development cohort, median age was 57.5 years (range = 20.4-84.8 years). Median progression-free survival and overall survival (OS) were 13.4 and 73.6 weeks, respectively, 90-day mortality was 16%, and overall response rate was 20%. In multivariate analysis, Eastern Cooperative Oncology Group performance status greater than or equal to 1 (hazard ratio [HR] = 3.2, 95% confidence interval [CI] = 1.8 to 5.7; P < .001), number of metastatic sites greater than 2 (HR = 2.0, 95% CI = 1.3 to 3.1; P = .003), and albumin less than the lower limit of normal (HR = 1.8, 95% CI = 1.2 to 2.7; P = .007) were independent prognostic factors; comprising the Princess Margaret Immuno-oncology Prognostic Index (PM-IPI). Patients with a score of 2-3 compared with patients with a score of 0-1 had shorter OS (HR = 3.4, 95% CI = 1.9 to 6.1; P < .001), progression-free survival (HR = 2.3, 95% CI = 1.7 to 3.2; P < .001), higher 90-day mortality (odds ratio = 8.1, 95% CI = 3.0 to 35.4; P < .001), and lower overall response rate (odds ratio = 0.4, 95% CI = 0.2 to 0.8; P = .019). The PM-IPI retained prognostic ability in both validation cohorts and performed better than previously published phase I prognostic scores for predicting OS in all three cohorts. Conclusions: The PM-IPI is a validated prognostic score for patients treated in phase IIO trials and may aid in improving patient selection.
  • Item
    Thumbnail Image
    CART-WHEEL.org: An Ethically Approved Online Database for Patient-Entered Data to Facilitate Rare Cancer Research
    Kee, D ; Parker, C ; Bae, S ; Tucker, KM ; Harrison, M ; Tohidi-Esfahani, I ; Black, M ; Delahunty, R ; Ananda, S ; Friedlander, M ; Cunliffe, HE ; Gibbs, P ; Desai, J ; Trotman, J ; Scott, CL (LIPPINCOTT WILLIAMS & WILKINS, 2020-02-21)
    PURPOSE: Rare cancers are challenging for researchers, as clinicians and scientists have difficulty recruiting sufficient patient cases to power studies appropriately. Likewise, patients often are frustrated by a lack of specific information or evidence base for their cancer and, although eager to participate in research, have limited opportunities. We established CART-WHEEL.org, an online patient-entered database, to directly engage patients in the research process, collect rare cancer data, and facilitate their entry into additional research. PATIENTS AND METHODS: Patients access CART-WHEEL.org directly online. Clinical information is collected from users via a streamlined questionnaire developed collaboratively with consumer groups to ensure accessibility and relevance. Data collected include the following: patient demographics, comorbidities, and risk factors and tumor diagnostic, biomarker, and treatment history. Patients can download a medical summary for personal use; consent for research use of data; and indicate willingness to be contacted about other research or clinical trials. We describe data collected to date and its validation, and we provide examples of how CART-WHEEL.org can facilitate rare cancer research. RESULTS: From January 2010 to March 2018, 558 patients provided consent and entered their rare cancer data. One hundred distinct rare tumor types and patients from 22 countries were included. Validation of data entered by 21 patients with sarcoma against a hospital database demonstrated accuracy sufficient to facilitate future research in key fields, such as tumor site (95%) and histopathologic diagnosis (90%). Examples of CART-WHEEL-based disease-specific projects, subsequent recruitment to other rare cancer projects, and rare cancer patient cases of interest are described. CONCLUSIONS: Online platforms like CART-WHEEL.org can engage consumers directly, facilitating collection of patient-entered rare cancer data for hypothesis generation, and connect patients with researchers to enable specific rare cancer research and clinical trials.