Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 21
  • Item
    Thumbnail Image
    Management of adverse events from the treatment of encorafenib plus cetuximab for patients with BRAF V600E-mutant metastatic colorectal cancer: insights from the BEACON CRC study
    Tabernero, J ; Velez, L ; Trevino, TL ; Grothey, A ; Yaeger, R ; Van Cutsem, E ; Wasan, H ; Desai, J ; Ciardiello, F ; Yoshino, T ; Gollerkeri, A ; Maharry, K ; Christy-Bittel, J ; Kopetz, S (ELSEVIER, 2021-12)
    Colorectal cancer is the second leading cause of cancer deaths worldwide, with a 5-year relative survival of 14% in patients with metastatic colorectal cancer (mCRC). Patients with BRAF V600E mutations, which occur in ∼10%-15% of patients with mCRC, have a poorer prognosis compared with those with wild-type BRAF tumours. The combination of the BRAF inhibitor encorafenib with the epidermal growth factor receptor inhibitor cetuximab currently represents the only chemotherapy-free targeted therapy approved in the USA and Europe for previously treated patients with BRAF V600E-mutated mCRC. As a class, BRAF inhibitors are associated with dermatologic, gastrointestinal, and renal events, as well as pyrexia and secondary skin malignancies. Adverse event (AE) profiles of specific BRAF inhibitors vary, however, and are affected by the specific agents given in combination. In patients with mCRC, commonly reported AEs of cetuximab monotherapy include infusion reactions and dermatologic toxicities. Data from the phase III BEACON CRC study indicate that the combination of encorafenib with cetuximab has a distinct safety profile. Here we review the most frequently reported AEs that occurred with this combination in BEACON CRC and best practices for managing and mitigating AEs that require more than standard supportive care.
  • Item
    Thumbnail Image
    Activity of the Gamma Secretase Inhibitor AL101 in Desmoid Tumors: A Case Report of 2 Adult Cases
    Chan, D ; Kaplan, J ; Gordon, G ; Desai, J (MDPI, 2021-10)
    Desmoid tumors (aggressive fibromatosis) are soft tissue mesenchymal tumors that can be locally invasive and life-threatening. Depending on the location, these tumors are often unresectable or tend to recur after surgery. To date, there are no approved systemic therapies for desmoid tumors. These tumors typically harbor mutations in the β-catenin oncogene CTNNB1 or the tumor suppressor gene adenomatous polyposis coli, resulting in constitutive activation of the WNT pathway. The Notch pathway is part of the underlying cause for desmoid tumor development, possibly due to crosstalk with the WNT pathway, providing a rationale for Notch inhibition as a therapeutic strategy. The gamma secretase activation of the Notch receptor can be targeted with investigational gamma secretase inhibitors. In this case report, we follow the course of 2 patients with desmoid tumors treated with the highly potent, parenterally administered investigational gamma secretase inhibitor AL101, resulting in long-lasting responses. Case 1 reports on a patient with a mesenteric desmoid tumor who participated in a phase 1 trial and then transitioned into a compassionate use program; Case 2 reports on a patient with recurrent pelvic tumors receiving AL101 through a compassionate use program. After tumor progression on other systemic therapies, Cases 1 and 2 had confirmed partial responses (41% and 60% maximal tumor size decrease from baseline) recorded after 1.0 and 1.6 years of treatment with AL101, with a duration of response of 8.6+ and 2.6+ years, respectively. Also, in a phase 1 study of AL102, a potent orally administered gamma secretase inhibitor that shares structural features with AL101, a patient with a desmoid tumor was noted to have tumor shrinkage. Formal clinical testing of AL102 for the treatment of patients with desmoid tumors that are not amenable to surgery or are refractory to/recurrent from other prior therapies is currently underway.
  • Item
    Thumbnail Image
    Criteria-based curation of a therapy-focused compendium to support treatment recommendations in precision oncology
    Lin, FP ; Thavaneswaran, S ; Grady, JP ; Ballinger, M ; Kansara, M ; Oakes, SR ; Desai, J ; Lee, CK ; Simes, J ; Thomas, DM (NATURE PORTFOLIO, 2021-06-23)
    While several resources exist that interpret therapeutic significance of genomic alterations in cancer, many regional real-world issues limit access to drugs. There is a need for a pragmatic, evidence-based, context-adapted tool to guide clinical management based on molecular biomarkers. To this end, we have structured a compendium of approved and experimental therapies with associated biomarkers following a survey of drug regulatory databases, existing knowledge bases, and published literature. Each biomarker-disease-therapy triplet was categorised using a tiering system reflective of key therapeutic considerations: approved and reimbursed therapies with respect to a jurisdiction (Tier 1), evidence of efficacy or approval in another jurisdiction (Tier 2), evidence of antitumour activity (Tier 3), and plausible biological rationale (Tier 4). Two resistance categories were defined: lack of efficacy (Tier R1) or antitumor activity (Tier R2). Based on this framework, we curated a digital resource focused on drugs relevant in the Australian healthcare system (TOPOGRAPH: Therapy Oriented Precision Oncology Guidelines for Recommending Anticancer Pharmaceuticals). As of November 2020, TOPOGRAPH comprised 2810 biomarker-disease-therapy triplets in 989 expert-appraised entries, including 373 therapies, 199 biomarkers, and 106 cancer types. In the 345 therapies catalogued, 84 (24%) and 65 (19%) were designated Tiers 1 and 2, respectively, while 271 (79%) therapies were supported by preclinical studies, early clinical trials, retrospective studies, or case series (Tiers 3 and 4). A companion algorithm was also developed to support rational, context-appropriate treatment selection informed by molecular biomarkers. This framework can be readily adapted to build similar resources in other jurisdictions to support therapeutic decision-making.
  • Item
    Thumbnail Image
    Pexidartinib improves physical functioning and stiffness in patients with tenosynovial giant cell tumor: results from the ENLIVEN randomized clinical trial
    van de Sande, M ; Tap, WD ; Gelhorn, HL ; Ye, X ; Speck, RM ; Palmerini, E ; Stacchiotti, S ; Desai, J ; Wagner, AJ ; Alcindor, T ; Ganjoo, K ; Martin-Broto, J ; Wang, Q ; Shuster, D ; Gelderblom, H ; Healey, JH (Medical Journal Sweden AB, 2021-07-04)
    Background and purpose - The ENLIVEN trial showed that, after 25 weeks, pexidartinib statistically significantly reduced tumor size more than placebo in patients with symptomatic, advanced tenosynovial giant cell tumor (TGCT) for whom surgery was not recommended. Here, we detail the effect of pexidartinib on patient-reported physical function and stiffness in ENLIVEN.Patients and methods - This was a planned analysis of patient-reported outcome data from ENLIVEN, a double-blinded, randomized phase 3 trial of adults with symptomatic, advanced TGCT treated with pexidartinib or placebo. Physical function was assessed using the Patient-Reported Outcomes Measurement Information System (PROMIS)-physical function (PF), and worst stiffness was assessed using a numerical rating scale (NRS). A mixed model for repeated measures was used to compare changes in PROMIS-PF and worst stiffness NRS scores from baseline to week 25 between treatment groups. Response rates for the PROMIS-PF and worst stiffness NRS at week 25 were calculated based on threshold estimates from reliable change index and anchor-based methods.Results - Between baseline and week 25, greater improvements in physical function and stiffness were experienced by patients receiving pexidartinib than patients receiving placebo (change in PROMIS-PF = 4.1 [95% confidence interval (CI) 1.8-6.3] vs. -0.9 [CI -3.0 to 1.2]; change in worst stiffness NRS = -2.5 [CI -3.0 to -1.9] vs. -0.3 [CI -0.9 to 0.3]). Patients receiving pexidartinib had higher response rates than patients receiving placebo for meaningful improvements in physical function and stiffness. Improvements were sustained after 50 weeks of pexidartinib treatment.Interpretation - Pexidartinib treatment provided sustained, meaningful improvements in physical function and stiffness for patients with symptomatic, advanced TGCT.
  • Item
    No Preview Available
    The Role of Systemic Therapies in the Management of Soft Tissue Sarcoma
    Burdett, N ; Bae, S ; Hamilton, A ; Desai, J (Springer Singapore, 2021)
  • Item
    Thumbnail Image
    Encorafenib Plus Cetuximab as a New Standard of Care for Previously Treated BRAF V600E-Mutant Metastatic Colorectal Cancer: Updated Survival Results and Subgroup Analyses from the BEACON Study
    Tabernero, J ; Grothey, A ; Van Cutsem, E ; Yaeger, R ; Wasan, H ; Yoshino, T ; Desai, J ; Ciardiello, F ; Loupakis, F ; Hong, YS ; Steeghs, N ; Guren, TK ; Arkenau, H-T ; Garcia-Alfonso, P ; Elez, E ; Gollerkeri, A ; Maharry, K ; Christy-Bittel, J ; Kopetz, S (LIPPINCOTT WILLIAMS & WILKINS, 2021-02-01)
    PURPOSE: BEACON CRC evaluated encorafenib plus cetuximab with or without binimetinib versus investigators' choice of irinotecan or FOLFIRI plus cetuximab in patients with BRAFV600E-mutant metastatic colorectal cancer (mCRC), after progression on 1-2 prior regimens. In the previously reported primary analysis, encorafenib, binimetinib plus cetuximab (ENCO/BINI/CETUX; triplet) and encorafenib plus cetuximab (ENCO/CETUX; doublet) regimens improved overall survival (OS) and objective response rate (ORR; by blinded central review) versus standard of care. The purpose of this analysis was to report updated efficacy and safety data. METHODS: In this open-label, phase III trial, 665 patients with BRAF V600E-mutant mCRC were randomly assigned 1:1:1 to receive triplet, doublet, or control. Primary end points were OS and independently reviewed ORR comparing triplet to control. OS for doublet versus control was a key secondary end point. Updated analyses include 6 months of additional follow-up and ORR for all randomized patients. RESULTS: Patients received triplet (n = 224), doublet (n = 220), or control (n = 221). Median OS was 9.3 months (95% CI, 8.2 to 10.8) for triplet and 5.9 months (95% CI, 5.1 to 7.1) for control (hazard ratio [HR], 0.60 [95% CI, 0.47 to 0.75]). Median OS for doublet was 9.3 months (95% CI, 8.0 to 11.3) (HR v control, 0.61 [95% CI, 0.48 to 0.77]). Confirmed ORR was 26.8% (95% CI, 21.1% to 33.1%) for triplet, 19.5% (95% CI, 14.5% to 25.4%) for doublet, and 1.8% (95% CI, 0.5% to 4.6%) for control. Adverse events were consistent with the prior primary analysis, with grade ≥ 3 adverse events in 65.8%, 57.4%, and 64.2% for triplet, doublet, and control, respectively. CONCLUSION: In the BEACON CRC study, encorafenib plus cetuximab improved OS, ORR, and progression-free survival in previously treated patients in the metastatic setting compared with standard chemotherapy. Based on the primary and updated analyses, encorafenib plus cetuximab is a new standard care regimen for previously treated patients with BRAF V600E mCRC.
  • 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
    Long-term outcomes of pexidartinib in tenosynovial giant cell tumors
    Gelderblom, H ; Wagner, AJ ; Tap, WD ; Palmerini, E ; Wainberg, ZA ; Desai, J ; Healey, JH ; van de Sande, MAJ ; Bernthal, NM ; Staals, EL ; Peterfy, CG ; Frezza, AM ; Hsu, HH ; Wang, Q ; Shuster, DE ; Stacchiotti, S (WILEY, 2021-03-15)
    BACKGROUND: The objective of this study was to report on the long-term effects of pexidartinib on tenosynovial giant cell tumor (TGCT). METHODS: This was a pooled analysis encompassing 3 pexidartinib-treated TGCT cohorts: 1) a phase 1 extension study (NCT01004861; 1000 mg/d; n = 39), 2) ENLIVEN patients randomized to pexidartinib (1000 mg/d for 2 weeks and then 800 mg/d; n = 61), and 3) ENLIVEN crossover patients (NCT02371369; 800 mg/d; n = 30). Eligible patients were 18 years old or older and had a histologically confirmed TGCT that was unresectable and symptomatic. Efficacy endpoints included the best overall response (complete or partial response) and the duration of response (DOR) by the Response Evaluation Criteria in Solid Tumors (RECIST) and the tumor volume score (TVS). The safety assessment included the frequency of treatment-emergent adverse events (TEAEs) and hepatic laboratory abnormalities (aminotransferase elevations and mixed/cholestatic hepatotoxicity). The data cutoff was May 31, 2019. RESULTS: One hundred thirty patients with TGCT received pexidartinib (median treatment duration, 19 months; range, 1 to 76+ months); 54 (42%) remained on treatment at the end of the analysis (26 months after initial data cut of March 2017). The RECIST overall response rate (ORR) was 60%; the TVS ORR was 65%. The median times to response were 3.4 (RECIST) and 2.8 months (TVS), with 48 of the responding patients (62%) achieving a RECIST partial response by 6 months and with 72 (92%) doing so by 18 months. The median DOR was reached for TVS (46.8 months). Reported TEAEs were mostly low-grade, with hair color changes being most frequent (75%). Most liver abnormalities (92%) were aminotransferase elevations; 4 patients (3%) experienced mixed/cholestatic hepatotoxicity (all within the first 2 months of treatment), which was reversible in all cases (recovery spanned 1-7 months). CONCLUSIONS: This study demonstrates the prolonged efficacy and tolerability of long-term pexidartinib treatment for TGCT.