Poster Abstracts

IF 1.4 4区 医学 Q4 ONCOLOGY
{"title":"Poster Abstracts","authors":"","doi":"10.1111/ajco.14010","DOIUrl":null,"url":null,"abstract":"<p>Dianheng Bu<sup>1</sup>, Javier Torres<sup>1,2</sup>, Arvind Sahu<sup>1 </sup></p><p><i><sup>1</sup>Goulburn Valley Health, Shepperton, Australia</i></p><p><i><sup>2</sup>Department of Rural Health, Shepparton Medical School, Melbourne University, Shepperton, Australia </i></p><p><b>Background</b>: Long-term adherence to endocrine therapy is challenging, even in the clinical trial setting. Non-adherence to treatment has an increased risk of cancer recurrence and poorer survival.<sup>1</sup> The evidence surrounding the adherence rate to endocrine therapy is sparse overall. The study aims to measure the adherence rate to adjuvant endocrine therapy for early breast cancer and factors contributing to non-adherence in the regional setting.</p><p><b>Methods</b>: A cross-sectional survey was conducted at Goulburn Valley Health between January and June 2023. Female patients receiving adjuvant oral endocrine therapy were included. Adherence was measured using the validated 6-item Simplified Medication Adherence Questionnaire (SMAQ).<sup>2</sup></p><p><b>Results</b>: Fifty-one patients (mean age: 60.1 years) participated. The adherence rate, as determined by the SMAQ, was 64.7% (33 out of 51). Age and the number of side effects significantly impacted adherence. Adherent patients had a mean age of 64.3 years, while non-adherent patients had a mean age of 52.4 years (<i>p</i> &lt; 0.001) (Figure 1). The mean number of side effects was 1.8 for adherent patients and 3.8 for non-adherent patients (<i>p</i> &lt; 0.01). No significant differences were found in adherence based on cancer stage, ECOG status, or type of endocrine therapy.</p><p><b>Conclusion</b>: The prevalence of adherence to oral adjuvant endocrine therapy for early breast cancer in the regional population was below 65%. Younger age and experiencing more side effects were associated with lower adherence rates. These findings emphasize the need for early interventions to improve adherence and optimize treatment outcomes in this population.</p><p><b>References</b>:</p><p>1. Chlebowski RT, Kim J, Haque R. Adherence to endocrine therapy in breast cancer adjuvant and prevention settings. <i>Cancer Prev Res</i>. 2014;7(4):378-387.</p><p>2. Ortega Suarez FJ, Sanchez Plumed J, Lorenzo AD. Validation on the simplified medication adherence questionnaire (SMAQ) in renal transplant patients on tacrolimus. <i>Nefrologia</i>. 2011;31(6):690-696.</p><p>Caitlin Cocks<sup>1</sup>, Sowmya Cheruvu<sup>1</sup>, Hilary L. Martin<sup>1</sup></p><p><i><sup>1</sup>Fiona Stanley Hospital, Murdoch, Australia </i></p><p><b>Background</b>: COVID-19 mortality rates of up to 24% have been reported in oncology patient cohorts prior to the availability of antiviral medications and vaccinations.<sup>1</sup> The COVID-19 pandemic was managed in Western Australia (WA) with strict vaccination mandates and border restrictions. This study aims to assess the impact of COVID-19 infection on oncology patients following the WA border opening in March 2022.</p><p><b>Methods</b>: Medical oncology patients at a tertiary hospital in WA who tested positive for COVID-19 between March and May 2022 were prospectively identified. Case notes were reviewed for data collection, including treatment delays, cancer outcomes, and mortality at 30 days, 60 days, and 12 months post-infection.</p><p><b>Results</b>: Thirty-seven patients tested positive for COVID-19 with 64.9% symptomatic. 78.4% of patients had received at least two COVID-19 immunizations prior to infection. Breast cancer was the most common malignancy (40.5%), followed by non-small cell lung cancer (21.6%). Twenty-nine patients were on active therapy, nine on chemotherapy, and three on immunotherapy. Twenty patients were treated with anti-viral therapy. Eight patients were hospitalized. Treatment delays affected 76.9% of survivors with a median delay of 2 weeks. Ten patients had disease progression in the subsequent 12 months. Five patients reported a second COVID-19 infection and recovered without complication. A total of 13 patients died during follow-up: eight within 30 days, three between 30 and 60 days, and two between 60 days and 12 months. No direct deaths related to COVID-19 were reported.</p><p><b>Conclusion</b>: Reassuringly, there were no deaths directly related to COVID-19. These findings support the use of antivirals and vaccination in this cancer patient population and contrast with the early high rates of COVID-19 mortality.</p><p><b>Reference</b>:</p><p>Russell B, Moss CL, Shah V, et al. Risk of COVID-19 death in cancer patients: an analysis from Guy's Cancer Centre and King's College Hospital in London. <i>Br J Cancer</i>. 2021;125(7):939-947. https://doi.org/10.1038/s41416-021-01500-z.</p><p>Andrew Fantoni<sup>1</sup>, Brandon Lau<sup>2</sup>, Jess McNeill<sup>3</sup>, Christine Henneker<sup>3</sup>, Shaouli Shahid<sup>4</sup>, Wei-Sen Lam<sup>1 </sup></p><p><i><sup>1</sup>Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia</i></p><p><i><sup>2</sup>Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Australia</i></p><p><i><sup>3</sup>WA Country Health Service, Central Office, East Perth, Australia</i></p><p><i><sup>4</sup>Centre for Aboriginal Studies, Curtin University of Technology, Bentley, Australia </i></p><p><b>Background</b>: Telehealth could offer greater availability to specialist oncology care whilst maintaining patient proximity to home. The Western Australia Country Health Service (WACHS) TeleChemotherapy model was implemented in the Pilbara (Karratha) in September 2019, and in the Kimberley (Broome) and Wheatbelt (Narrogin) in March 2020. We examined the safety of this model with regards to treatment utilization and toxicity outcomes.</p><p><b>Methods</b>: Retrospective data were collected for all patients referred to a WACHS TeleChemotherapy service between September 2019 and February 2022; including patient demographics, cancer diagnosis, treatment intent, cycles and lines of treatment received, treatment delays, and toxicity (adverse events [AEs] as per CTCAEv5.0, deaths, and hospital admissions). Descriptive statistics were used to report safety outcomes.</p><p><b>Results</b>: Of 159 patients referred for treatment, 154 (97%) were accepted. Twenty-one withdrew due to progressive deterioration or preference for metropolitan treatment. The final cohort (<i>n</i> = 133) had a median age of 61 years (range 24–90) with 54% male, 23% Aboriginal and 94% ECOG≤1. A total of 1034 cycles of treatment were delivered, representing 171 lines of therapy. 74% were palliative in intent. Mean dose intensity was maintained at 90% (±14%). Forty-seven treatment lines (27%) were subject to dose reduction, 62% prior to TeleChemotherapy. Fifty-eight lines (34%) were subject to significant dose delay, due to toxicity (53%) or patient preference (38%). Sixty-three lines (37%) were associated with grade ≥3 AEs. There were no significant associations between incidence of AEs and regimen. ≥1 hospitalization occurred in 32% (<i>n</i> = 54) of treatment lines. Eleven deaths were recorded, one related to toxicity. Forty-eight immunotherapy-containing lines were delivered, four lines (8%) with grade≥3 immune-related AEs and no deaths.</p><p><b>Conclusion</b>: This study affirms the safety of the WACHS TeleChemotherapy model including low rates of immune-mediated toxicity. Dose intensity was not compromised, and treatment delays were due to expected toxicities or patient preferences rather than logistical issues.</p><p>Jocelyn Finney<sup>1</sup>, Isabella Wilson<b><sup>1</sup></b>, Ben Felmingham<sup>2,3,4</sup>, Sanjeev Kumar<sup>1</sup>, Alina Mahmood<sup>1</sup>, Lisa Horvath<sup>1,5</sup>, David Celermajer<sup>6</sup>, Rachel Conyers<sup>7 </sup></p><p><i><sup>1</sup>Chris O'Brien Lifehouse, Sydney, Australia</i></p><p><i><sup>2</sup>Murdoch Children's Research Institute, Melbourne, Australia</i></p><p><i><sup>3</sup>Children's Cancer Centre, The Royal Children's Hospital, Melbourne, Australia</i></p><p><i><sup>4</sup>Cardiac Regeneration Laboratory, Murdoch Children's Research Institute, Melbourne, Australia</i></p><p><i><sup>5</sup>Sydney Medical School</i><i>, University of Sydney, Sydney, Australia</i></p><p><i><sup>6</sup>Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia</i></p><p><i><sup>7</sup>Department of Paediatrics, University of Melbourne, Melbourne, Australia </i></p><p><b>Background</b>: Cancer therapy related cardiac dysfunction (CTRCD) is a significant side effect of several chemotherapy therapies, targeted agents, and immunotherapies integral to treatment. Anthracyclines are an example of a commonly used chemotherapy drug that can cause irreversible early and late onset heart failure which are currently under-documented. In Australia, there are currently no other multi-center, longitudinal studies examining CTRCD in adults. COBLH aims to use the Australian Cardio-Oncology Registry (ACOR) to explore the epidemiology, clinical presentation, and management of CTRCD amongst adults receiving cardiotoxic therapies. Additionally, a biobanking sub-study will attempt to identify gene polymorphisms predisposing individuals to developing anthracycline cardiotoxicity (ACT).</p><p><b>Methods</b>: ACOR is a prospective observational study involving 14 sites across Australia. COBLH patient criteria includes non-palliative, high-risk adult patients treated with cardiotoxic therapies within the last 5 years. Information is obtained from medical records and de-identified upon entry into ACOR. ACOR collects data on patient demographics, medical history, cancer diagnosis and treatment, cardiac assessments, and the management of CTRCD. These instruments are collected at baseline prior to treatment, and at 6-month intervals thereafter. Patients who have developed ACT will be asked to participate in the biobanking study, which will involve a one-off 12 mL peripheral blood draw to be processed at an external site for whole genome exome sequencing and the development of pluripotent stem cell derived cardiomyocytes.</p><p><b>Results</b>: At the time of submission, 19 of 220 recruited patients at COBLH have developed CTRCD. This includes 10 patients with ACT, three with left ventricular dysfunction, three with suspected anthracycline related cardiomyopathies, and three with symptomatic heart failure. ACOR anticipates full registered site capacity to capture 600 patients annually.</p><p><b>Conclusion</b>: COBLH will use ACOR to study long-term CTRCD in adult patients with the biobanking study seeking to prove a causal association between genetic predisposition and ACT.</p><p><b>Acknowledgments</b>: 2,3.</p><p>Adriana Gangemi, Bella Nguyen, Wei-sen Lam<sup> </sup></p><p><i>Fiona Stanley Hospital, Perth, Australia </i></p><p><b>Background</b>: Anaplastic thyroid cancers (ATCS) are rare aggressive tumors, with extremely poor prognosis despite multimodality treatments. Recently data have shown that a combination of lenvatinib, a tyrosine kinase inhibitor, and pembrolizumab, a checkpoint inhibitor that functions by inhibition of PD-1, can improve response rate and survival in patients with ATC.<sup>1,2</sup> Our case showcases a real-world example of this combination treatment.</p><p><b>Methods and results</b>: This case describes a 67-year-old female initially diagnosed in September 2021, presenting with enlarging neck lump. Thyroidectomy and bilateral neck dissection revealed a 42 mm anaplastic thyroid carcinoma, vascular invasion present, extrathyroidal extension in strap muscle, 0/25 lymph node involved, clear margins. This patient completed adjuvant chemoradiation with low dose doxorubicin in December 2021. Disease recurred locally in August 2022 and was deemed not suitable for resection. She was started on pembrolizumab and lenvatinib (20 mg) on September 9, 2022 and her symptoms improved over 4 weeks. The PET scan after 8 weeks of treatment showed almost a complete response and a PET 6 months post-treatment continued to show no convincing evidence of recurrence (Figure 1).</p><p><b>Conclusion</b>: In the context of an extremely poor prognostic disease, this case showed that the combination of lenvatinib and pembrolizumab for ATCs is a game-changer.</p><p><b>References</b>:</p><p>1. Dierks C, Seufert J, Ruf J, et al. 1915P The lenvatinib/pembrolizumab combination induces long lasting and complete responses in patients with metastatic anaplastic or poorly differentiated thyroid carcinoma: results from a retrospective study and first results from the prospective phase II ATLEP trial. <i>Ann Oncol</i>. 2020;31:S1085.</p><p>2. Luongo C, Porcelli T, Sessa F, et al. Combination of lenvatinib and pembrolizumab as salvage treatment for paucicellular variant of anaplastic thyroid cancer: a case report. <i>Curr Oncol</i>. 2021;28(6):5401-5407.</p><p>Grace Gard, Joanna Oakley, Vanessa Wong, Kelsey Serena, Michael Harold, Jo Cockwill, Katya Gray, Helen Anderson, Graeme Down, Judi Price, Peter Gibbs<sup> </sup></p><p><i>WEHI, Parkville, Australia </i></p><p><b>Background/aims</b>: In cancer research, there is growing interest in consumers and researchers working together in partnership, yet the practicalities of partnering and how to optimize the co-design process have not been well established. A research group of health care professionals and consumers aimed to create a Personalized Care Plan for patients with newly diagnosed locally advanced rectal cancer. The group has reflected on and documented their experience of co-design.</p><p><b>Methods</b>: Our research group includes consumers from the consumer program at WEHI, multidisciplinary colorectal cancer clinicians, and project officers. Together they created the Personalized Care Plan template, to be provided to patients and their general practitioner. The team's reflections on the co-design process have been captured using Gibbs’ Reflective Cycle. Patients are currently enrolling into a prospective cohort to evaluate the impact of the Personalized Care Plan.</p><p><b>Results</b>: In 2022, over six meetings, we created a two-page Personalized Care Plan to embed into the WEHI-CRC database. Personalized information for individual patients includes disease stage, planned treatment, and scheduled follow-up. Reflection on the co-design process highlighted the importance of establishing expectations, having expertise within the consumer group, and open communication and respect. Challenges included time commitment, power dynamics, diversity of representation, and loss of consumers due to health reasons and time availability. Responses reflected a positive attitude-change of the researchers on the value of consumer input. Both consumers and researchers communicated the high objective and affective value of contributing to the project.</p><p><b>Conclusion</b>: Reflecting on the co-design process for a patient education sheet, we found that essential components of a co-design group are having clear expectations, and strategies to address challenges such as time commitment and entrenched power dynamics. Meaningful input from consumers can objectively improve the outcome of the project while offering positive personal value for consumers and researchers involved.</p><p>Tushar Goyal, Jennifer Zhang, Bianca Devitt<sup> </sup></p><p><i>Eastern Health, Melbourne, Australia </i></p><p><b>Background</b>: There is growing interest and variability in the choice and timing of antibiotics in transitioning from intravenous to oral therapy. We aimed to understand the treatment and prescribing practices of neutropenic sepsis in our institution.</p><p><b>Methods</b>: A retrospective study was performed at a tertiary hospital across two sites in Melbourne. Adult patients admitted with solid organ malignancies who met the criteria for neutropenic sepsis between January 2020 and December 2021 were eligible. Analysis was performed using descriptive statistics (Table 1).</p><p><b>Results</b>: Sixty-nine patients were identified. Median time from presentation to intravenous antibiotics (ivABx) was 2 h (IQR = 3 h), duration of ivABx was 3 days and total duration of antibiotics was 7 days (IQR = 4 days), with no significant association between the duration and ANC nadir. Sixty-one patients (88%) were culture negative. Fifty-seven patients (83%) received oral (PO) antibiotic tail upon completion of ivAbx, with amoxicillin-clavulanate being the most commonly used (69%), and cephalosporins (13%) being more commonly prescribed after an infective focus was clinically evident. Fifty-three patients (77%) had been clinically stable and afebrile for more than 48 h before being switched to PO antibiotics. Patients with high-severity MASCC scores received a longer duration of total antibiotics and had longer length of stay as inpatients. Seven of these patients (21.2%) required ICU admission, with no patients from the low severity group being admitted to ICU. One patient experienced gastrointestinal adverse effects during deescalated antibiotic therapy. Hospital readmission rates due to recurrence of neutropenic sepsis were similar in patients who received oral antibiotics versus others.</p><p><b>Conclusion</b>: There were notable variations in the selection and duration of antibiotics for the management of culture-negative neutropenic sepsis, in comparison to guidelines. Future investigations could explore the potential benefits of early transition to oral antibiotics to assess their impact on clinical outcomes.</p><p>Nikki How<sup>1</sup>, Dhanvee Kandadai<sup>2</sup>, Paul Moroz<sup>3</sup>, David Morris<sup>4</sup>, David Ransom<sup>1 </sup></p><p><i><sup>1</sup>Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia</i></p><p><i><sup>2</sup>Royal Perth Hospital, Perth, Australia</i></p><p><i><sup>3</sup>Western Australian Peritonectomy Service, Joondalup, Australia</i></p><p><i><sup>4</sup>St. George Peritonectomy Unit, Kogarah, Australia </i></p><p><b>Background</b>: CRS-HIPEC is a potential treatment option for patients with primary peritoneal malignancies or secondary peritoneal carcinomatosis from dissemination of intraabdominal malignancies. Accumulating evidence for the survival benefits of CRS-HIPEC has contributed to its increasing use, with the aim of prolonged survival and curative intent. This study aims to report the long-term survival outcomes for Western Australian patients who had access to CRS-HIPEC through an Australian interstate program.</p><p><b>Methods</b>: This was a retrospective observational study of Western Australian patients who were transferred to St George Hospital in New South Wales for CRS-HIPEC between 2008 and 2013. Data was obtained from multiple Western Australian registers, supplemented by review of individual patient records. Overall survival was assessed using Kaplan–Meier methods and comparison with log-rank tests.</p><p><b>Results</b>: Forty-two patients received CRS-HIPEC in the period of assessment and had follow-up data available. Median time from diagnosis to CRS-HIPEC was 8.5 months. The most common diagnoses included 13 patients with disseminated peritoneal adenomucinosis (DPAM), 11 with appendiceal adenocarcinoma (AA), nine with colorectal cancer (CRC), three with peritoneal mesothelioma (PM), and three with peritoneal mucinous carcinomatosis (PMCA). Median overall survival across all patients was 50 months, with a 5-year overall survival of 50%. The 5-year overall survival rates for DPAM, AA, and CRC were 75%, 32%, and 40%, respectively. Survival beyond 6 years occurred in 16 patients including nine patients with DPAM, three with AA, two with ovarian malignancies, one with CRC, and one with PM. Patients with PMCA had a comparatively lower survival probability (<i>p</i> &lt; 0.0001).</p><p><b>Conclusion</b>: These outcomes are consistent with results from other centers. The increasing demand led to the establishment of a state CRS-HIPEC Centre in Western Australia. It is worth noting that even with peritoneal metastases, survival beyond 6 years can be achieved in patients with AA, CRC, and PM.</p><p>Joshua Hurwitz<sup>1,2,3</sup>, Greta Beale<sup>1</sup>, James Pham<sup>1,2</sup>, Lauren Brown<sup>1,3</sup>, Anthony Rodrigues<sup>1,2,3</sup>, Anthony M Joshua<sup>1,2,3</sup>, Susan Clark<sup>1,2,3 </sup></p><p><i><sup>1</sup>The</i> Kinghorn <i>Cancer Centre, Darlinghurst, Australia</i></p><p><i><sup>2</sup>St</i> Vincent's <i>Clinical School, University of New South Wales, Darlinghurst, Australia</i></p><p><i><sup>3</sup>Gavan Institute of Medical Research, Darlinghurst, Australia </i></p><p><b>Background</b>: Vitamin C is a key cofactor in modulating epigenetic landscapes, genomic stability, and potential cancer growth (1). While rare in the general population, vitamin C deficiency is frequently observed in cancer patients. Denmark and New Zealand research identified inadequate vitamin C levels in cancer patients at baseline and further reduction with treatment (2, 3, 4). This study aimed to determine baseline vitamin C levels in solid cancer patients and the impact of systemic therapy in our own patient cohort.</p><p><b>Methods</b>: This prospective observational study aimed to recruit 31 subjects, 25 metastatic cancer patients and six healthy controls. Patients were recruited at The Kinghorn Cancer Centre. Information collected included demographics, patient characteristics, and malignancy type. Plasma vitamin C concentrations were collected before systemic therapy and after two treatment cycles. The normal vitamin C reference range was 40–80 μmol/L, and low was &lt;40 μmol/L.</p><p><b>Results</b>: Twenty-eight patients were recruited; five did not provide pathology samples for vitamin C levels – of the 23 remaining, 14 patients had a second vitamin C level (Table 1). At baseline, healthy controls had lower vitamin C levels (55 vs. 57) than their cancer counterparts. Vitamin C levels were higher after systemic therapy than before treatment (57 vs. 49). Three patients had weight loss before diagnosis, and one required nutritional supplementation. Treatment included multi-agent chemotherapy (38%), immunotherapy (24%), and single-agent chemotherapy (19%).</p><p><b>Conclusion</b>: In our cohort, vitamin C levels pre- and post-systemic therapy were within physiological parameters, in contrast to prior cancer studies across larger patient cohorts reporting marked vitamin C deficiency (2, 3, 4). Baseline vitamin C was lower, but not significantly, in the healthy controls. Further research with larger sample sizes is required to investigate vitamin C levels in solid malignancies, with systemic therapy and its role in cancer management.</p><p>Mandeep Singh Kalsi, Inosha Jayasekara, Frances Barnett<sup> </sup></p><p><i>Northern</i> Health<i>, Epping, Melbourne, Australia </i></p><p><b>Background</b>: Chemotherapy that was usually delivered only in hospital environments is now administered primarily in outpatient (OP) environment. There are financial incentives for OP chemotherapy delivery.</p><p><b>Methods</b>: We conducted a retrospective analysis to evaluate the outcomes of patients who were given inpatient (IP) chemotherapy agents for solid organ malignancies over a period of 3 years at a metropolitan hospital.</p><p><b>Results</b>: Small cell cancer of lung primary (20) was the common malignancy in 47 patients who received inpatient chemotherapy. There were 13 patients with gastro-intestinal primary, five patients with testicular cancer, four with breast cancer, three with ovarian cancer, one with cancer of unknown primary, and one with Kaposi sarcoma. Most patients’ inferred Eastern Cooperative Oncology Group (ECOG) performance status was 0% and 15% patients had ECOG status of 3. Thirty-day mortality was 6% and 60-day mortality was 23%. Sixty-day mortality was associated with higher ECOG status. Thirty-nine patients received subsequent chemotherapy sessions.</p><p><b>Conclusion</b>: IP chemotherapy administration was associated with high 60-day mortality. And high mortality was associated with higher ECOG score. More detailed analysis of patient characteristics is required to co-relate with mortality that may help in decision making for oncologists to decide for inpatient chemotherapy.</p><p>Muhammad A. Khattak<sup>1</sup>, Georgina V. Long<sup>2</sup>, Alexander M. M. Eggermont<sup>3</sup>, Jeffrey E. Gershenwald<sup>4</sup>, Dirk Schadendorf<sup>5</sup>, Paolo A. Ascierto<sup>6</sup>, Reinhard Dummer<sup>7</sup>, Axel Hauschild<sup>8</sup>, Matteo S. Carlino<sup>9</sup>, Antoni Ribas<sup>10</sup>, Caroline Robert<sup>11</sup>, Richard A. Scolyer<sup>12</sup>, Vernon K. Sondak<sup>13</sup>, Jonathan E. Cohen<sup>14</sup>, Jinchun Zhang<sup>15</sup>, Dmitri Grebennik<sup>15</sup>, Clemens Krepler<sup>15</sup>, Jason J. Luke<sup>16 </sup></p><p><i><sup>1</sup>Fiona</i> Stanley <i>Hospital and Edith Cowan University, Perth, Australia</i></p><p><sup>2</sup>Melanoma <i>Institute Australia, The University of Sydney, Mater and Royal North Shore Hospitals, Sydney, Australia</i></p><p><i><sup>3</sup>Faculty of Medicine University Medical Center and Princess Máxima Center, Utrecht, The Netherlands; Comprehensive</i> <i>Cancer Center Munich, Technical University Munich and Ludwig Maximilian University, Munich, Germany</i></p><p><i><sup>4</sup>The University of Texas MD Anderson Cancer Center, Houston, Texas, USA</i></p><p><i><sup>5</sup>University</i> Hospital <i>Essen, University Duisburg-Essen, West German Cancer Centre (WTZ), and German Cancer Consortium, Partner Site Essen, Essen, Germany</i></p><p><i><sup>6</sup>Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy</i></p><p><sup>7</sup>University <i>of Zurich and University Hospital Zurich, Zurich, Switzerland</i></p><p><i><sup>8</sup>University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany</i></p><p><i><sup>9</sup>Sydney</i> Medical <i>School, Faculty of Medicine and Health Sciences, University of Sydney, Camperdown, Melanoma Institute Australia, Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, Australia</i></p><p><sup>10</sup>Jonsson <i>Comprehensive Cancer Center, UCLA and Parker Institute for Cancer Immunotherapy, Los Angeles, California, USA</i></p><p><i><sup>11</sup>Université Paris Saclay, Le Kremlin Bicêtre and Gustave Roussy, Villejuif, France</i></p><p><sup>12</sup>Melanoma <i>Institute Australia, The University of Sydney, Faculty of Medicine and Health, The University of Sydney, Charles Perkins Centre, The University of Sydney, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, Australia</i></p><p><i><sup>13</sup>H. Lee</i> Moffitt <i>Cancer Center, Tampa, Florida, USA</i></p><p><i><sup>14</sup>Sharett</i> Institute <i>of Oncology and The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel</i></p><p><sup>15</sup>Merck <i>&amp; Co., Inc., Rahway, New Jersey, USA</i></p><p><i><sup>16</sup>UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, Pennsylvania, USA </i></p><p><b>Background</b>: Adjuvant pembrolizumab monotherapy improved recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) in patients with high-risk resected melanoma. Vibostolimab, an anti-TIGIT antibody, demonstrated antitumor activity and manageable safety in combination with pembrolizumab in solid tumors in the phase 1 KEYVIBE-001 study. The efficacy and safety of adjuvant coformulated vibostolimab with pembrolizumab in patients with resected high-risk melanoma in the double-blind, randomized, phase 3 KEYVIBE-010 study (NCT05665595).</p><p><b>Methods</b>: Patients ≥12 years old, with surgically resected stage IIB or IIC (pathologic or clinical), III, or IV cutaneous melanoma, and an ECOG PS of 0 or 1 (&gt;18 years), Karnofsky performance status ≥70 (≥16–&lt;18 years), or Lansky play-performance scale ≥70 (&lt;16 years) and have no evidence of metastatic disease will be enrolled. Patients with ocular, mucosal, or conjunctival melanoma will be excluded. Resection must occur within 12 weeks before randomization. Patients will need to provide an archival tumor tissue sample or newly obtained biopsy of a tumor lesion not previously irradiated. Patients will be stratified by risk (stage IIB/IIC/clinical IIB and stage IIC/IIIA/IIIB vs. stage IIIC/IIID/IV) and region (Asia vs. rest of the world). Approximately 1560 patients will be randomly assigned 1:1 to intravenous coformulated vibostolimab 200 mg with pembrolizumab 200 mg or pembrolizumab 200 mg (2 mg/kg up to 200 mg for adolescents) every 3 weeks for up to 17 cycles. The primary end point is RFS by investigator review. Secondary end points are DMFS by investigator review, overall survival, safety, and quality-of-life. Hazard ratios and 95% CIs will be estimated using a stratified Cox regression model with the Efron methods of handling ties, with treatment as a covariate. Between-treatment differences will be evaluated using a stratified log-rank test. Enrollment is ongoing.</p><p>Chee Lee<sup>1</sup>, Sandy Simon<b><sup>1</sup></b>, Danka Zebic<sup>1</sup>, Yeh Chen Lee<sup>1</sup>, Katrina Diamante<sup>1</sup>, Lenna Lai<sup>1</sup>, Rachel O'Connell<sup>1</sup>, Anna deFazio<sup>2</sup>, Sonia Yip<sup>1</sup>, Lyndal Anderson<sup>3</sup>, Bo Gao<sup>4</sup>, Jeffrey C. Goh<sup>5</sup>, Kate Webber<sup>6</sup>, Sally Baron-Hay<sup>7</sup>, Christopher Steer<sup>8</sup>, Linda Mileshkin<sup>9</sup>, Tarek Meniawy<sup>10</sup>, Janine Lombard<sup>11</sup>, Martin Stockler<sup>1</sup>, Michael Friedlander<sup>12 </sup></p><p><i><sup>1</sup>NHMRC</i> Clinical <i>Trials Centre, The University of Sydney, Camperdown, Australia</i></p><p><i><sup>2</sup>The University of Sydney, The Westmead Institute for Medical Research and Westmead Hospital, Westmead, Australia</i></p><p><i><sup>3</sup>Royal Prince Alfred Hospital, Camperdown, Australia</i></p><p><i><sup>4</sup>Westmead Hospital, Westmead, Australia</i></p><p><i><sup>5</sup>Royal Brisbane and Women's Hospital, Brisbane, Australia</i></p><p><i><sup>6</sup>Monash Medical Centre, Clayton, Australia</i></p><p><i><sup>7</sup>Royal North Shore Hospital, St Leonards, Australia</i></p><p><i><sup>8</sup>Border Medical Oncology Research Unit, Albury, Australia</i></p><p><i><sup>9</sup>Peter MacCallum Cancer Centre, Melbourne, Australia</i></p><p><i><sup>10</sup>Sir Charles Gairdner Hospital, Nedlands, Australia</i></p><p><i><sup>11</sup>Newcastle Private Hospital, New Lambton Heights, Australia</i></p><p><i><sup>12</sup>Prince of Wales Hospital and Royal Hospital for Women, Randwick, Australia </i></p><p>Aromatase inhibitors (AIs) are used to treat hormone receptor positive (HR+) advanced gynecological cancers. However, response rates are significantly lower than HR+ breast cancer treated with AIs. Alterations in the CDK4/6 pathway and mutations of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) are postulated to cause AI resistance. In HR+ metastatic breast cancer, combination of AI plus CDK4/6 inhibitor (ribociclib) or PIK3CA inhibitor (alpelisib) improves clinical outcomes. PARAGON-II trial (ACTRN12621000639820) aims to evaluate the activity of letrozole plus alpelisib or ribociclib, in PIK3CA mutant (mut) and PIK3CA wild-type (wt) HR+ advanced gynecological cancers, respectively.</p><p>This is a phase II, open label, basket trial, with six cohorts of common and rare HR+ gynecological cancers. Target population is patients with recurrent/metastatic cancer, with good performance status and for whom chemotherapy is not indicated. PIK3CA mutational status is determined by tissue or ctDNA analysis. The study consists of PIK3CA mut <i>endometrial</i> cancer (cohort A) and any other PIK3CA mut tumors (F). PIK3CA wt/unknown cohorts consist of endometrial cancer (B), platinum resistant high-grade epithelial ovarian cancers (C), endometrial stromal sarcoma, leiomyosarcoma, granulosa cell tumors (D), and low grade serous ovarian cancers (E). In the PIK3CA mut cohorts (A and F), participants will receive letrozole 2.5 mg plus alpelisib 300 mg daily until disease progression or intolerance. In the PIK3CA wt/unknown cohorts (B–E), participants will receive letrozole 2.5 mg daily plus ribociclib 600 mg daily (3-weeks-on/1-week-off). A total of 182 participants will be recruited. The primary endpoint is objective response rate. Secondary endpoints include disease control rate, duration of response, 6-month progression free survival (PFS), and median PFS, patient reported outcomes and adverse events. Tumor and blood samples are being collected for translational studies.</p><p>PARAGON-II trial started recruitment in May 2022. As of June 30, 2023, 64 participants have been recruited from 13 sites. Recruitment is expected to be completed by December 2024.</p><p>AIs are used to treat hormone receptor positive (HR+) advanced gynecological cancers. However, response rates are significantly lower than HR+ breast cancer treated with AIs. Alterations in the CDK4/6 pathway and mutations of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) are postulated to cause AI resistance. In HR+ metastatic breast cancer, combination of AI plus CDK4/6 inhibitor (ribociclib) or PIK3CA inhibitor (alpelisib) improves clinical outcomes.</p><p>Su Saint Lee<sup>1,2,3</sup>, Wei Chua<sup>1,4,5</sup>, Nancy Huang<sup>1,2,3</sup>, Weng Ng<sup>1,4,5</sup>, Aflah Roohullah<sup>1,2</sup>, Annette Tognela<sup>2</sup>, Ray Asghari<sup>3</sup>, Mahtab Farzin<sup>6</sup>, Tara Roberts<sup>4,5,7</sup>, Therese Becker<sup>4,5,7</sup>, Bin Wang<sup>6</sup>, Soon Lee<sup>5,6,7</sup>, Stephanie Lim<sup>3,4,5</sup></p><p><i><sup>1</sup>Department of Medical Oncology, Liverpool Hospital, Liverpool, Australia</i></p><p><i><sup>2</sup>Department of Medical Oncology, Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, Australia</i></p><p><i><sup>3</sup>School of Medicine, Western Sydney University, Campbelltown, Australia</i></p><p><i><sup>4</sup>Department of</i> <i>Medical Oncology, Bankstown-Lidcombe Hospital, Bankstown, Australia</i></p><p><i><sup>5</sup>Department of Anatomical Pathology, Liverpool Hospital, Liverpool, Australia</i></p><p><i><sup>6</sup>Ingham Institute of Applied Medical Research, Liverpool, Australia</i></p><p><i><sup>7</sup>South Western Clinical School, University of New South Wales, Liverpool, Australia</i></p><p><b>Background</b>: Tumor mutational profile affects prognosis in metastatic colorectal cancer (mCRC) but it remains unclear why some tumors behave exceptionally well or poorly. We aim to identify genes associated with exceptional cases using targeted next generation sequencing (NGS).</p><p><b>Methods</b>: Patients with RAS/BRAF-wildtype and MMR-proficient mCRC from the South Western Sydney Anatomical Pathology tissue bank from 2010 to 2018 with overall survival of &lt;6 months or &gt;3 years or atypical treatment responses were identified. Clinicopathological variables and survival outcomes were obtained. Random sample of eight patients underwent testing with a 30-gene (Qiagen) or 50-gene or 161-gene (ThermoFisher) NGS panel to establish workflow, feasibility and obtain preliminary data. Mutations identified were classified according to ESCAT tiers of significance. All cases will have 161-gene panel NGS.</p><p><b>Results</b>: Fifty-two good performers (GP) and 14 poor performers (PP) were identified. Median age was 65 years in GP and 51 in PP. Tumors were left-sided in 87% of GP and 77% of PP. Median overall survival from diagnosis of metastatic disease was 56.4 months (20.6–127.1 months) in GP and 4.8 months (1.4–15.4 months) in PP. 58% of GP and 14% of PP had ≥2 lines of chemotherapy. To date we have NGS data for six GP and two PP patients (four patients with 30-gene, two with 50-gene, and two with 161-gene panels). Two GP patients had TP53 mutations, another a PIK3CA mutation (all Tier II), while a fourth had an EGFR mutation of unknown significance. No mutations were detected in 2GP and two PP patients. 161-Gene panel testing is underway for patients who so far had limited testing.</p><p><b>Conclusion</b>: Targeted NGS panels may identify genes in exceptional performers in RAS/BRAF-wildtype mCRC, whose overall survival varies by more than 10-fold. Preliminary data shows differences in TP53 and PIK3CA in GP. Final 161-gene NGS results of all cases are underway.</p><p>Yeh Chen Lee<sup>1</sup>, Elizabeth Barnes<sup>1</sup>, Katherine Francis<sup>1</sup>, Danka Zebic<sup>1,2</sup>, Sandy Simon<sup>1</sup>, Gemma Blunt<sup>1</sup>, Pearly Khaw<sup>3</sup>, Viet Do<sup>4</sup>, Elizabeth Christie<sup>3</sup>, Rachel Delahunty<sup>3</sup>, Rebecca Mercieca-Bebber<sup>1</sup>, <i>Sonia</i> Yip<sup>1</sup>, Jeffrey Goh<sup>5</sup>, Janine Lombard<sup>6</sup>, Kate Webber<sup>7</sup>, Sudarshan Selva-Nayagam<sup>8</sup>, Ganessan Kichenadasse<sup>9</sup>, Tarek Meniawy<sup>10</sup>, Michelle Harrison<sup>11</sup>, Michelle Wilson<sup>12</sup>, Martin Stockler<sup>1</sup>, Linda Mileshkin<sup>3 </sup></p><p><i><sup>1</sup>NHMRC Clinical Trials Centre, University of Sydney, Camperdown, Australia</i></p><p><i><sup>2</sup>Prince of Wales Hospital and Royal</i> <i>Hospital for Women, Randwick, Australia</i></p><p><i><sup>3</sup>PeterMacCallum Cancer Centre, Melbourne, Australia</i></p><p><i><sup>4</sup>Liverpool Cancer Therapy Centre, Liverpool, Australia</i></p><p><i><sup>5</sup>Royal Brisbane and Women's Hospital, Brisbane, Australia</i></p><p><i><sup>6</sup>Calvary Mater Newcastle, Newcastle, Australia</i></p><p><i><sup>7</sup>Monash Medical Centre, Clayton, Australia</i></p><p><i><sup>8</sup>Royal Adelaide Hospital, Adelaide, Australia</i></p><p><i><sup>9</sup>Flinders Medical Centre, Bedford Park, Australia</i></p><p><i><sup>10</sup>Sir Charles Gairdner Hospital, Nedlands, Australia</i></p><p><i><sup>11</sup>Chris O'Brien Lifehouse, Camperdown, Australia</i></p><p><i><sup>12</sup>Auckland City Hospital, Auckland, New Zealand </i></p><p><b>Background</b>: Despite primary surgery, adjuvant chemoradiation, and chemotherapy, people with high-risk endometrial cancer (EC) remain at significant risk of relapse, following which the median survival time is approximately 12 months. Immune checkpoint inhibitors have shown activity in advanced EC. It is recognized that radiation and chemotherapy can have immune-stimulating properties and can synergize with immunotherapy, providing rationale for combining these treatments in high-risk curable EC. The aim of ADELE is to determine if adding tislelizumab to standard adjuvant chemotherapy after post-operative pelvic chemoradiation for high-risk EC improves outcomes.</p><p><b>Methods</b>: This is an open label, non-comparative, multicenter, randomized (2:1) phase 2 trial (Figure 1). The target population is people with high-risk FIGO stage IA–IVA EC who have had primary surgery and are planned for adjuvant pelvic chemoradiation followed by chemotherapy. Participants will be registered prior to commencing adjuvant pelvic chemoradiation and vaginal vault brachytherapy (if cervical stromal invasion) and subsequently randomized to one of two treatment groups in a ratio of 2:1 (experimental to control). The control group will receive chemotherapy (carboplatin plus paclitaxel) for four cycles. The experimental group will receive tislelizumab with chemotherapy for four cycles, followed by tislelizumab alone for another eight cycles. The primary outcome is failure free survival at 1 year. Secondary outcomes include feasibility, adverse events, effects on patient-reported outcomes, failure free survival, and overall survival. Tumor and blood samples will be collected for translational studies, with correlative objectives including impact of adjuvant therapy on ctDNA levels and potential predictive or prognostic biomarkers for response to immunotherapy.</p><p><b>Progress</b>: ADELE started recruitment in March 2022 and will enroll 135 participants. As of June 30, 2023, 17 participants have been randomized from 11 sites. Clinical trial identifier: ACTRN12621000273886.</p><p><b>Acknowledgments</b>: ADELE is an investigator-initiated study, funded by an MRFF Research Grant and Beigene Ltd.</p><p>Meghana Maddula<sup>1,2,3</sup>, Peter Fox<sup>1,4 </sup></p><p><i><sup>1</sup>Central West Cancer Centre, Orange Health Service, Orange, Australia</i></p><p><i><sup>2</sup>Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, Australia</i></p><p><i><sup>3</sup>Blacktown Haematology and Cancer Centre, Blacktown Hospital, Sydney, Australia</i></p><p><i><sup>4</sup>Western Sydney University, Bathurst Clinical School, Bathurst, Australia </i></p><p><b>Background</b>: Positron emission tomography (PET) is integral to the diagnosis, monitoring, and treatment of malignancy. Whilst PET utilization has increased, inequity in access remains across Australia. The OECD average in 2019 was 4 PET per 1000 persons. In this study, we aim to identify local trends in PET usage and evaluate geospatial disparity.</p><p><b>Methods</b>: In this retrospective population study, we evaluated PET scan utilization for Medicare funded oncological indications between 2001 and 2022. Data was extracted from public Medicare statistics and stratified by population density, state/territory, and tumor type. Additional Remoteness Area data were obtained from 2018 to 2021 and AIHW data informed cancer incidence.</p><p><b>Results</b>: Over 22 years, 1,124,431 PET scans were attended, with 2,746,117 incident cancers. This corresponds to 2.2 PET per 1000 persons and 40.9 per 100 cancers but had increased to 5.4 per 1000 persons and 89.2 per 100 cancers by the end of study period (Table 1). PET uptake per capita increased nationwide with a compound annual growth rate of 18.4%. The greatest per capita usage of PET scans was in New South Wales followed by Western Australia, with the greatest annualized growth in the Northern Territory (28.6%) and Tasmania (24.2%). PET scanner availability by state/territory did not correlate with PET utilization (<i>R</i> = .15, <i>p</i> = 0.72). PET scans per 100 incident cancers was highest in lymphoma (201.9) and head and neck (147.0), and lowest in neurological (14.1) and colorectal cancers (39.4). By Remoteness Area, PET per 1000 persons was lower in remote (3.1) and very remote (1.9) compared to major cities (4.4).</p><p><b>Conclusion</b>: Australian PET scan usage has increased significantly since the introduction of Medicare subsidized PET in 2001 and is now above the OECD average. Geospatial disparity remains in PET usage between states/territories and based on rurality. Further research into variations in PET utilization is required to bridge the gap.</p><p>Rhiannon Mellor<sup>1,2,3*</sup>, Jessica Smith<sup>4,5*</sup>, Anna Mislang<sup>6,7* </sup></p><p><sup>*</sup>All first authors</p><p><i><sup>1</sup>Chris O'Brien Lifehouse, Camperdown, Australia</i></p><p><i><sup>2</sup>University of New South Wales, Sydney, Australia<sup>3</sup>The Garvan Institute of</i> <i>Medical Research, Darlinghurst, Australia</i></p><p><i><sup>4</sup>Macquarie University Hospital, Macquarie University, Australia</i></p><p><i><sup>5</sup>Western Sydney University, Parramatta, Australia</i></p><p><i><sup>6</sup>Department of Medical Oncology, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, Australia</i></p><p><i><sup>7</sup>College of Medicine and Public Health, Flinders University, Bedford Park, Australia </i></p><p><b>Background</b>: Medical oncologists operate in an increasingly complex environment, regardless of their practice location. Over the last 3 years, the COVID-19 pandemic has increased this complexity. International studies report that up to 71% of oncologists exhibit evidence of burnout,<sup>1</sup> and this has worsened since COVID-19.<sup>2</sup> Burnout negatively impacts the psychological well-being of the individual healthcare professional and the level of care they provide, significantly costing individuals and the broader healthcare organization. This is a critical issue that must be urgently addressed.</p><p><b>Aims and objectives</b>: This study aims to determine the current prevalence of burnout amongst trainees and Fellows of the Royal Australasian College of Physicians (FRACP) in Medical Oncology in Australia. Secondary objectives are to (1) examine the drivers of burnout among oncologists in Australia, (2) assess the impact of burnout, and (3) collect data on potential interventions to address burnout and clinician wellbeing, as identified by oncologists.</p><p><b>Methods and results</b>: In this cross-sectional study, FRACP and trainee medical oncologists in Australia will be invited to complete an anonymous online questionnaire. The questionnaire will assess burnout with the Maslach Burnout Inventory (MBI) and measure the consequences of burnout using the Patient Health Questionnaire-4 (PHQ-4) to screen for anxiety and depression, and the Stanford self-valuation tool. Factors that contribute to both wellbeing and burnout will also be examined, along with potential work-based interventions to improve wellbeing.</p><p><b>Conclusion</b>: The prevalence and drivers of burnout amongst medical oncologists in Australia are poorly understood. This study will provide essential data for a solutions-based, <i>multifactorial</i> approach to improve clinician wellbeing.</p><p><b>Acknowledgments</b>: Thank you to Bethan Richards and Jia Liu for their input on questionnaire development.</p><p><b>References</b>:</p><p>1. Banerjee S, Califano R, Corral J, et al. Professional burnout in European young oncologists. <i>Ann Oncol</i>. 2017; 28(7):1590-1596.</p><p>2. Banerjee S, Lim KHJ, Murali K, et al. The impact of COVID-19 on oncology professionals. <i>ESMO Open</i>. 2021;6(2):100058.</p><p>Hanh Nguyen, Bella Nguyen<sup> </sup></p><p><i>Fiona Stanley Hospital, Murdoch, Australia </i></p><p><b>Background</b>: For patients with borderline resectable (BR) and locally advanced (LA) pancreatic cancers, neoadjuvant chemotherapy (NAT) is routinely offered to downstage their disease for potential pancreatectomy. In many cases, however, pancreatectomy remains contraindicated after NAT and alternative therapies are required. This study aims to retrospectively compare the characteristics of patients with BR and LA pancreatic cancer who received versus those who did not receive pancreatectomy after NAT within the South Metropolitan Health Service (SMHS) in Perth Australia from 2015 to 2022.</p><p><b>Methods</b>: Patient electronic medical records (eMR) were used to identify patients who received NAT for their BR or LA pancreatic cancer. Data on patient demographics, clinical characteristics, and adjuvant therapies received post-NAT were attained for further analysis.</p><p><b>Results</b>: There were 19 patients who received NAT to downstage their BR or LA disease for potential resection. Out of these patients, 36.8% (<i>n</i> = 7) successfully underwent pancreatectomy whilst 57.9% (<i>n</i> = 11) did not qualify for surgery and 5.3% (<i>n</i> = 1) were lost to follow-up. Patients undergoing pancreatectomy were more likely to have a recorded ECOG status of zero (85.7% vs. 54.5%). Other characteristics were comparable between patients who underwent pancreatectomy versus those who did not undergo pancreatectomy, respectively; mean age (61 vs. 59 years), sex (male = 72.7% vs. 71.4%), tumor location (pancreatic head = 90.9% vs. 100%), and CA-19-9 level at diagnosis (2448.5 vs. 2999.4). Adjuvant chemotherapy (42.9% vs. 91%) and radiation therapy (28.6% vs. 63.6%) were less frequently utilized for patients who did versus those who did not receive pancreatectomy, respectively.</p><p><b>Conclusion</b>: Aside from ECOG status, there were no significant demographic or clinical differences amongst BR or LA pancreatic patients who did versus those who did not undergo pancreatectomy after NAT. Patients who underwent pancreatectomy were less likely to require subsequent adjuvant chemotherapy and radiation therapy.</p><p>Hanh Nguyen, Bella Nguyen<sup> </sup></p><p><i>Fiona Stanley Hospital, Murdoch, Australia </i></p><p><b>Background</b>: Whilst surgery is the only curative treatment modality for pancreatic cancer, local blood vessel involvement often precludes its resectability and classifies the disease as borderline resectable (BR) or locally advanced (LA). Neoadjuvant chemotherapy (NAT) is often offered to downstage BR and LA pancreatic cancers for potential resection. This study aims to retrospectively review the clinical management of patients with BR and LA pancreatic cancer within the South Metropolitan Health Service (SMHS) in Perth Australia from 2015 to 2022. Primary outcomes include pancreatectomy rates post-NAT, patient progression-free survival, and overall survival (OS).</p><p><b>Methods</b>: Patient electronic medical records (eMR) were used to identify all pancreatic cancer patients who received FOLFIRINOX or GnP (nanoparticle albumin-bound paclitaxel) as NAT for their BR or LA pancreatic cancer within the SMHS from 2015 to 2022.</p><p><b>Results</b>: Of 19 patients who received NAT, 15 received FOLFIRINOX and four received GnP. Of those who received FOLFIRINIX, 40.0% (<i>n</i> = 6) successfully underwent pancreatectomy whilst 53.3% (<i>n</i> = 8) did not qualify for surgery and 6.7% (<i>n</i> = 1) were lost to follow-up. Of those who received GnP, only one patient underwent pancreatectomy. Reasons precluding pancreatectomy after NAT included inadequate NAT response/progressive disease (63.6%, <i>n</i> = 7) and poor performance status (36.4%, <i>n</i> = 11). Pancreatectomy specimens were staged from IA–IIB, most were grade 2 (85.7%; <i>n</i> = 6) and resected with negative margins (R0) (57.1%, <i>n</i> = 4). The median survival (30 vs. 13 months) and median progression-free survival (20 vs. 7 months) for patients who underwent pancreatectomy were higher than those who did not undergo pancreatectomy, respectively.</p><p><b>Conclusion</b>: Pancreatectomy was performed in two in five patients after FOLFIRINOX NAT and one in four patients after GnP NAT. These rates reflect real-world patients who are less likely to be surgically fit compared to published trial data such as CONKO-007.7.</p><p>Ek Leone Oh, Elizabeth Steinepreis, Hilary Martin </p><p><i>Fiona Stanley Hospital, Murdoch, Australia </i></p><p><b>Background</b>: Management for lobular breast cancer, which comprises approximately 10% of breast cancer, is generally extrapolated from trials comprised predominantly of ductal breast cancer patients. This study aims to investigate treatment outcomes of patients treated for metastatic lobular breast cancer.</p><p><b>Methods</b>: We performed a retrospective review of patients diagnosed with metastatic lobular breast cancer at a single center between January 2017 and April 31, 2023. Clinical benefit from treatment was defined as stable disease, partial response, or complete response.</p><p><b>Results</b>: Twenty-one patients with metastatic lobular breast cancer were identified. Sixteen (76.2%) had hormone-receptor positive HER2-negative breast cancer (HR+ HER2−), two (9.5%) had triple-negative breast cancer, and three (14.3%) had HER2-positive hormone-receptor positive (HER2+HR+) disease. Treatment and outcomes for HR+HER2− subgroup are shown in Table 1. Among the patients with HR+HER2− subgroup, 14 (87.5%) received a combination of CDK4/6 inhibitors and non-steroidal aromatase inhibitor (NSAI) of whom 13 started this as first-line therapy for metastatic disease and six (42.9%) remain on this treatment at data-cutoff. Seven (43.8%) patients with HR+HER− disease received chemotherapy at some point in their treatment. Both patients with triple-negative lobular breast cancer experienced rapidly progressive disease on all agents trialled (olaparib, paclitaxel, carboplatin/gemcitabine, and eribulin). The three patients with HER2+HR+ disease received a combination of taxane or vinorelbine, pertuzumab, and trastuzumab as first-line therapy. All remain on this treatment with a median duration of 34 months (range 19–68 months) at data-cutoff.</p><p><b>Conclusion</b>: HR+ HER2− lobular breast cancer treated with CDK4/6 inhibitors and NSAI had similar PFS compared to the registration trial data supporting the use of this combination for lobular cancer. There were also some responses seen to chemotherapy, with all patients who received capecitabine obtaining clinical benefit. Patients with HER2-positive disease had excellent outcomes, while outcomes were poor for triple-negative lobular breast cancer. Both patients with triple-negative lobular breast cancer experienced rapidly progressive disease on all agents trialed (olaparib, paclitaxel, carboplatin/gemcitabine, and eribulin). The three patients with HER2+HR+ disease received a combination of taxane or vinorelbine, pertuzumab, and trastuzumab as first-line therapy. All remain on this treatment with a median duration of 34 months (range 19–68 months) at data-cutoff.</p><p>Hayley Roberts<sup>1,2</sup>, Benjamin Solomon<sup>2</sup>, Susan Harden<sup>2</sup>, Senthil Lingaratnam<sup>2</sup>, Marliese Alexander<sup>2 </sup></p><p><i><sup>1</sup>Alfred</i> Health<i>, Melbourne, Australia, Australia</i></p><p><i><sup>2</sup>Peter</i> MacCallum <i>Cancer Centre, Melbourne, Australia </i></p><p><b>Background</b>: Lung cancer remains the leading cause of cancer death in Australia, despite recent progress in immunotherapy, tyrosine kinase inhibitors (TKIs), and radiotherapy. Thirty-day mortality after systemic anti-cancer therapy (SACT) has been suggested as a quality indicator for lung cancer and introduced as a benchmark in the UK. However, while multiple international researchers have reported 30-day mortality rates, the exact method for calculating these rates differs widely between studies.</p><p><b>Methods</b>: Patients ≥18 years of age with a histologically confirmed diagnosis of lung cancer who received palliative systemic anti-cancer treatment at Peter MacCallum Cancer Centre between January 2015 and December 2022 were identified using a lung cancer database (AURORA). Information was collected on patient, disease, and treatment characteristics. An annual 30-day mortality index following SACT was calculated (patients who died within 30 days of SACT, as a proportion of all patients given palliative-intent treatment for lung cancer in that year).</p><p><b>Results</b>: We found annual rates of 30-day mortality after palliative-intent SACT for lung cancer of between 9% and 15% (Figure 1). Patients who received SACT within 30 days of death were more likely to have received TKIs or immunotherapy, and to have had an ECOG performance status of two or more, and less likely to have had small cell histology. </p><p><b>Conclusions</b>: The rates of 30-day mortality in our center are comparable to internationally published rates. While 30-day mortality after SACT is a useful benchmark, several factors must be taken into account when comparing between institutions, including SACT intent (curative or palliative), number of lines of treatment, histological subtype, and type of treatment (especially for TKIs). Ensuring consistent methods of calculation is important, especially reporting time period and denominator population (total number of patients on SACT over a period of time, or total number of patients who have died).</p><p>Rahul Solanki<sup>1,2</sup>, Dylan Bartholomeusz<sup>1,2</sup>, Michael Cilento<sup>3,4</sup>, Eve Hopping<sup>3</sup>, Harsh Kanhere<sup>1,2,3</sup>, Timothy Price<sup>1,2,3 </sup></p><p><sup>1</sup><i>Royal Adelaide Hospital, Adelaide, Australia</i></p><p><sup>2</sup><i>The University of Adelaide, Adelaide, Australia</i></p><p><sup>3</sup><i>The Queen Elizabeth Hospital, Woodville South, Australia</i></p><p><sup>4</sup><i>South Australian Immunogenomics Cancer Institute, The University of Adelaide, Adelaide, Australia</i></p><p><b>Background</b>: A 78-year-old lady was referred for an FDG PET scan for staging of esophageal cancer. She was receiving endoscopic surveillance for Barrett's esophagus. Her last endoscopy showed two nodular lesions in the distal esophagus and biopsies revealed HER2 positive poorly differentiated esophageal adenocarcinoma.</p><p><b>Methods</b>: The CT staging scan showed distal esophageal thickening and enlarged regional lymph nodes. Her 18FDG staging PET scan showed increased FDG uptake in the distal esophagus at the primary site but also revealed extensive non-regional bilateral FDG avid lymphadenopathy and skeletal lesions suggesting metastatic disease.</p><p><b>Results</b>: After review in the upper gastrointestinal (UGI) multidisciplinary team (MDT) meeting, noting a past history of sarcoidosis, she underwent biopsy of the right axillary and right inguinal lymph nodes, which showed well-formed non-necrotizing granulomas suggestive of granulomatous lymphadenitis. There were no malignant cells detected. Following repeat review in the UGI MDT, the consensus decision was that this was a case of localized esophageal cancer with FDG avid nodal and bony lesions likely due to sarcoidosis. She was commenced on trimodality treatment with neoadjuvant chemoradiotherapy (chemoRT), which included weekly administration of dexamethasone along with chemotherapy, with the plan for subsequent curative esophageal resection. The repeat 18FDG PET scan post-chemoRT showed significant reduction of FDG uptake in the primary site and also reduction of the widespread 18FDG avid lymph nodes and bony lesions. The sarcoidosis is thought to have responded with dexamethasone with a plan for close follow-up Figure 1).</p><p><b>Conclusion</b>: This case emphasizes the complexity of decision making in staging patients with malignancy and concomitant inflammatory granulomatous disease and reveals the benefit of clinical history, MDT discussion and biopsy when clinical discrepancies arise.</p><p>Jennifer Soon<sup>1</sup><b><sup>,2</sup></b>, Fanny Franchini<sup>1</sup>, Maarten IJzerman<sup>1,3</sup>, Grant McArthur<sup>1,2,4 </sup></p><p><sup>1</sup><i>University of Melbourne, Parkville, Australia</i></p><p><i><sup>2</sup>Peter</i> MacCallum <i>Cancer Centre, Melbourne, Australia</i></p><p><i><sup>3</sup>Erasmus School of Health Policy and Management, Rotterdam, The Netherlands</i></p><p><i><sup>4</sup>Victorian Comprehensive Cancer Centre Alliance, Melbourne, Australia </i></p><p><b>Purpose</b>: With healthcare costs rising, focus is shifting toward maximizing health outcomes per dollar. One approach to optimizing value is through de-escalation, which is the rationalization of routine treatment without compromising patient outcomes. Since 2013, successful clinical trials of high-cost immune checkpoint, BRAF and MEK inhibition have led to these drugs becoming ubiquitous in melanoma management. Pipeline therapies such as relatlimab and tumor infiltrating lymphocyte (TIL) therapy are expected to have a similar or even greater cost. The purpose of this scoping review is to establish current and emerging opportunities to de-escalate the use of systemic therapies in the treatment of cutaneous melanoma. It will also seek to comment on the proportion of studies that include patient-reported outcomes and quality of life measures. We hypothesize that neoadjuvant and response-directed strategies will be identified as well as the emerging potential of prognostic and predictive tools.</p><p><b>Methods</b>: A systematic search strategy has been developed for MEDLINE and PubMed from January 1, 2013 to June 30, 2023. Additional texts will be sourced from grey literature and reference scanning. Two authors will screen abstracts and full-texts facilitated by the Covidence software. Disagreements will be resolved by consensus or a third reviewer. The first author will perform data extraction whilst a second author will review a random selection of papers to ensure consistent interpretation. De-escalation strategies will be categorized by concept, potential impact on resource utilization and patient outcomes, and strength of evidence. Data will be synthesized qualitatively and quantitatively. Results will be reported following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR). Preliminary results will be available in August 2023. The final results of this scoping review will directly inform a melanoma consumer and clinician survey exploring their perspectives on de-escalation strategies.</p><p>Joanne Tang, Sudarshan Selva-Nayagam<sup> </sup></p><p><i>Central Adelaide Local Health Network, Adelaide, Australia </i></p><p><b>Background</b>: Sequencing of adjuvant treatment and the use of weekly cisplatin with concurrent radiotherapy in endometrial cancer has not been clearly established. This audit aims to compare toxicity and treatment completion rates in patients who undergo chemotherapy then radiotherapy ± chemotherapy (ChtRt) to patients who undergo radiotherapy ± chemotherapy then chemotherapy (RttCh), and to establish the toxicity and completion rates of concurrent weekly cisplatin (1WC) versus 3-weekly cisplatin (3WC) during radiotherapy (CRTx).</p><p><b>Methods</b>: Patient demographics, treatment regimens, and toxicity profiles were collected for endometrial cancer patients who were treated with adjuvant radiotherapy ± chemotherapy, with chemotherapy preceding or following. Data from patient medical records were extracted across two tertiary hospitals between January 2015 and October 2021 and analyzed.</p><p><b>Results</b>: The study included 19 patients in the ChtRt and 15 patients in the RttCh group. Overall toxicity outcomes were similar between ChtRt and RttCh groups, except patients in the ChtRt group had higher rates of diarrhea during radiotherapy (74% vs. 20%). Hematological toxicity during CRTx was higher in ChtRt group (<i>n</i> = 6) (64% vs. 11% all grade toxicity). ChtRt patients had a higher rate of chemotherapy cessation during radiotherapy (67% vs. 18%), whilst RttCh patients had a higher rate of chemotherapy interruption/cessation (60% vs. 42%) (Table 1). Across both groups, 16 patients had 1WC, and five had 3WC. Overall grade &gt;3 toxicity was higher in the 1WC group (56% vs. 20%), with 31% of patients ceasing 1WC during CRTx, compared to none in the 3WC.</p><p><b>Conclusion</b>: In this small retrospective study, overall toxicity was similar across sequencing approaches, however treatment completion of each modality was higher in whichever modality was started first. 1WC CRTx had a higher toxicity profile compared to 3WC, and the ongoing use of this modified protocol in the absence of clear benefit is currently under review.</p><p>Christina Teng, Anew Killen, Charlotte Lemech<sup> </sup></p><p><i>Scientia Clinical</i> Research<i>, Randwick, Australia </i></p><p><b>Aim</b>: To identify and summarize the demographic factors of patients referred and enrolled to Scientia Clinical Research (SCR) Phase 1 oncology trials.</p><p><b>Background</b>: Phase 1 trials are first-in-human studies evaluating the safety, efficacy, and pharmacokinetics of newly developed medications. SCR is a dedicated Phase 1 trial unit located in NSW, Australia.</p><p><b>Methods</b>: Descriptive statistics were used to summarize de-identified data from the clinical records of patients referred to the SCR Phase 1 unit.</p><p><b>Results</b>: In the 2022 calendar year, 168 patients were referred to SCR for an opinion about early phase clinical trials, from 51 referrers. Of those referred, 139 patients had a first consultation. Figure 1 summarizes the outcomes of the patients referred. The mean age at time of referral was 59 years, (standard deviation 12, median 61, range 12–83). The most common primary diagnoses were upper gastrointestinal (29%), colorectal (23%), and gynecological (23%) cancers. Of the patients where referral location was available, the majority resided in metropolitan Sydney (47%). Regional NSW residents comprised 29% and interstate referrals 9%. Five initial consultations were conducted with the assistance of an interpreter (three Mandarin, one Cambodian, one Auslan). Of these, two patients proceeded to trial participation. In 2022, 52 patients consented and commenced trial screening over 21 trials; 29 were female (56%). Median age was 71. Residents of metropolitan Sydney comprised 50% of screened patients. Four patients were ineligible at screening; 48 commenced trial treatment. Median time from referral to starting trial was 14 days. The most common diagnosis of recruited patients was gynecological cancer (31%), followed by upper gastrointestinal (21%) and colorectal (20%) cancers.</p><p><b>Conclusion</b>: In 2022, referrals and trial participants to phase 1 oncology trials at SCR represented a diverse geographical population. Patients from culturally and linguistically diverse backgrounds were under-represented.</p><p>Lucy Porter<sup>1</sup>, Phillip Parente<sup>2,3</sup>, Grace Gard<sup>4,5</sup>, Benjamin Brady<sup>6</sup>, Wasek Faisal<sup>7,8</sup>, Dishan Herath<sup>4</sup>, Margaret Lee<sup>2,3</sup>, Peter Gibbs<sup>4,5</sup>, Ben Markman<sup>5,6</sup>, Rachel Wong<sup>2,3 </sup></p><p><i><sup>1</sup>School of Medicine</i>, Faculty <i>of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia</i></p><p><i><sup>2</sup>Department of Medical Oncology, Eastern Health, Melbourne, Australia</i>,</p><p><i><sup>3</sup>Eastern Health Clinical School, Monash University, Melbourne, Australia</i></p><p><i><sup>4</sup>Department of Medical Oncology, Western Health, Melbourne, Australia</i>,</p><p><i><sup>5</sup>Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia</i></p><p><i><sup>6</sup>Department of Medical Oncology, Cabrini Health, Melbourne, Australia</i></p><p><i><sup>7</sup>Grampians Integrated Cancer Service, Grampians Health, Ballarat, Australia</i></p><p><i><sup>8</sup>School of Health, La Trobe University, Melbourne, Australia </i></p><p><b>Background</b>: The treatment of early-stage non-small cell lung cancer (NSCLC) is evolving, with data increasingly supporting the role of immunotherapy and targeted agents in adjuvant and neo-adjuvant settings. Atezolizumab, a PD-L1 inhibitor, has been PBS-subsidized since November 2022 for resected Stage II–IIIa NSCLC patients with PD-L1 ≥ 50 who are EGFR/ALK wild-type and treated with platinum-based chemotherapy. The proportion of patients who receive molecular testing and are eligible for PBS-subsidized atezolizumab treatment in Australian hospitals is unknown.</p><p><b>Methods</b>: Patients enrolled in the multi-site INHALE lung cancer registry between February 2020 and January 2023 at four Australian sites (<i>n</i> = 448) were retrospectively analyzed. NSCLC patients who underwent surgical resection were included (<i>n</i> = 115) and analyzed for stage, molecular testing, treatment details, and outcomes.</p><p><b>Results</b>: A total of 90/115 resected NSCLC patients were treated with curative intent. 63/90 received surgery alone, 25/90 received adjuvant systemic therapy and/or radiotherapy, and 2/90 received neo-adjuvant systemic therapy and/or radiotherapy. 7/90 experienced disease recurrence/progression.</p><p>A total of 70/90 curative-intent resected patients had PD-L1 testing. 62/70 within 30 days and 67/70 within 90 days of surgery. 56/90 underwent genomic testing. Testing results are shown in Table 1.</p><p>Of 70 PD-L1 tested patients, one was eligible for adjuvant atezolizumab as per current PBS criteria. 67/70 were ineligible (PD-L1 &lt; 50%, ineligible-stage, no platinum-based chemotherapy), and 2/70 were PBS-eligibility unknown (genomic testing not performed). PD-L1 and genomic testing rates were higher in metropolitan versus regional settings (70%–90% vs. 40% and 15%–88% vs. 50%, respectively), and private versus public settings (90% vs. 70% and 88% vs. 15%–65%, respectively).</p><p><b>Conclusion</b>: In this real-world analysis of resected NSCLC patients, very few routine care patients were eligible for PBS-subsidized atezolizumab. We anticipate that PD-L1 and genomic testing rates will increase in response to the availability of adjuvant atezolizumab on the PBS and as evidence supporting neo-adjuvant/adjuvant use of immunotherapy and/or targeted agents continues to emerge.</p><p>Joanne Tang<sup>1</sup>, Brian Stein<sup>2,3 </sup></p><p><i><sup>1</sup>Central Adelaide Local</i> Health <i>Network, Adelaide, Australia</i></p><p><i><sup>2</sup>Icon Cancer Centre</i>, Adelaide<i>, Australia</i>,</p><p><i><sup>3</sup>University of Adelaide</i>, Adelaide<i>, Australia </i></p><p><b>Background</b>: EviQ is a consensus driven evidence-based repository of cancer treatment protocols widely used in Australia and serves as a de facto standard. A standardized assessment of the clinical benefit of the protocols published on EviQ has not been undertaken. Based on the references in EviQ we aimed to determine the feasibility of scoring all solid tumor protocols to assess the benefit of protocols and explore correlations between ESMO-MCBS scoring and treatment indication, tumor stream and trial endpoint.</p><p><b>Methods</b>: We graded all solid tumor protocols published on EviQ until April 31, 2022 with ESMO-MBCS version 1.1, excluding functional duplicates (e.g., weight-based vs. flat dose immunotherapy). We only used the “Evidence” section for reference as this is the evidentiary justification for protocol publication. In addition to technically unevaluable protocols, we regarded protocols using phase II data in common scenarios, lacking a standard comparator, or based on conglomerated data as unevaluable.</p><p><b>Results</b>: Of 478 included protocols, 301 (63%) were evaluable. Intent was curative in 97 and palliative in 204. Overall, 140 (47%) of evaluated protocols met the MCBS threshold for significant benefit; with 75% of curative but only 33% of palliative protocols being of “substantive benefit”. Of the 57 protocols that had toxicity or quality-of-life benefit, 88% were palliative and 47% were chemotherapy regimens. Of low benefit protocols, 49% were chemotherapy-based, and response-based outcomes were over-represented in palliative protocols. There was no difference in scores by tumor stream. Of the 177 unevaluable protocols, 31% lacked a standard comparator and 42% used phase II data in non-orphan settings.</p><p><b>Conclusion</b>: Within EviQ, 47% of evaluable protocols are ESMO-MCBS “substantial benefit”; most are curative. Few protocols gained points for less toxicity. We suggest adding ESMO-MCBS score to EviQ to aid in regimen consideration.</p><p>Sophie Tran<sup>1,2</sup>, Cassana Moore<sup>2</sup>, Danielle Roscoe<sup>2</sup>, Hieu Chau<sup>2</sup>, Sachin Joshi<sup>2,3</sup>, Quan Tran<sup>2</sup>, Rohan Nair<sup>2,3</sup>, Mahesh Iddawela<sup>2,3,4 </sup></p><p><i><sup>1</sup>Eastern</i> Health<i>, Box Hill, Australia</i></p><p><i><sup>2</sup>Latrobe</i> Regional <i>Health, Traralgon, Australia</i></p><p><i><sup>3</sup>Alfred Health, Melbourne, Australia</i></p><p><i><sup>4</sup>Department of Rural Health, Monash University, Traralgon, Australia </i></p><p><b>Background</b>: Brain metastases (BM) is a common occurrence in non-small cell lung cancer (NSCLC) and is associated with high morbidity and mortality. The advent of new systemic treatments and radiation techniques has led to improved outcomes in this prognostically poor group. As such, early detection at diagnosis and surveillance is important. This project aims to review the characteristics, current practice, management and outcomes of lung cancer BM at a single regional center to help inform development of local protocols.</p><p><b>Methods</b>: Patients with lung cancer BM were identified from the William Buckland Radiotherapy Centre medical records and Latrobe Regional Health lung cancer database. A total of 101 patients were identified between 2017 and 2022. Demographic and clinical information data were collected.</p><p><b>Results</b>: The median age at lung cancer diagnosis was 68 years, 59% were male and 71% ECOG 0 or 1. At initial diagnosis, 66% (67/101) had metastatic disease and of these patients, 60% (40/67) had BM. Initial brain imaging was performed in 68% (67/101). Targetable oncogenic aberrations were found in 13% (13/101). The median time to BM was 10 months and median survival from BM diagnosis was 4 months. 37% (37/101) of patients had surgical resection, 20% (20/101) had stereotactic radiation (SRS) alone, 32% (32/101) had whole brain radiation therapy (WBRT) and 12% (12/101) had no local therapy. The surgery or SRS group had better survival outcomes compared to those who had WBRT or no local treatment following BM diagnosis (log rank <i>χ</i><sup>2</sup>(2) = 7.590, <i>p</i> = 0.02) (Figure 1).</p><p><b>Conclusion</b>: The incidence of BM at initial diagnosis is higher than published data with prognosis following BM diagnosis poor. This may be reflective of the regional demographic of patients who present more advanced. This underlines the importance of establishing local guidelines in intracranial disease detection and screening for earlier diagnosis and timely management to improve outcomes.</p><p>Niamh Walsh<sup>1,2</sup>, Rosalie Stephens<sup>3</sup>, Alvin Tan<sup>4</sup>, Vanessa Durandt<sup>5</sup>, Jennifer McLachlan<sup>6</sup>, Jody Jordan<sup>7</sup>, Kate Gregory<sup>8</sup>, Sean Sutton<sup>2</sup>, Catherine Barrow<sup>2</sup>, Annie Wong<sup>1,2 </sup></p><p><i><sup>1</sup>University of</i> Otago<i>, Wellington, New Zealand</i></p><p><i><sup>2</sup>Wellington Blood &amp; Cancer Centre, Wellington, New Zealand</i></p><p><i><sup>3</sup>Regional Blood and Cancer Service, Auckland, New Zealand</i></p><p><i><sup>4</sup>Medical Oncology, Waikato, New Zealand</i></p><p><i><sup>5</sup>Oncology and Haematology, Dunedin, New Zealand</i></p><p><i><sup>6</sup>Canterbury</i> Regional <i>Cancer and Haematology Service, Christchurch, New Zealand</i></p><p><i><sup>7</sup>Midcentral District Cancer, Hawkes Bay, New Zealand</i></p><p><i><sup>8</sup>Nelson Cancer</i> Centre<i>, Nelson, New Zealand </i></p><p><b>Background</b>: Most patients with metastatic melanoma will develop brain metastases (BM), with poor prognosis. Immune checkpoint inhibitors (ICI) are active for melanoma BM. However, landmark clinical trials are not representative of the “real-world” population, due to exclusion of patients with symptomatic BM, poor performance status or steroid use.<sup>1,2</sup> This study reports the real-world outcomes of ICI for melanoma BM in New Zealand (NZ).</p><p><b>Methods</b>: A retrospective audit of patients with melanoma BM treated with ICI between September 1, 2016, and September 1, 2020 was conducted across seven NZ cancer centers. Baseline demographics, tumor characteristics, treatment, intracranial tumor response rates, intracranial progression-free survival (iPFS), and overall survival (OS) were recorded.</p><p><b>Results</b>: A total of 144 patients received at least one dose of ICI. Most patients (93%) received anti-PD1 monotherapy. Almost a quarter of patients had baseline performance status (ECOG 2–3), 56% were symptomatic and 33% had corticosteroids. Intracranial response was seen in 36%, and median-iPFS was 9 months. The 6- and 12-month-iPFS rates were 55% and 45%, respectively. Seventy-seven (53%) patients received local therapies, for example, surgery or radiation. The median OS was 15 months, and a third of patients were alive at 2 years (Figure 1). The toxicity of ICI was 28% and 15% for grade 1–2 and 3–4 events, respectively. Of those patients still alive at 2 years, 23% were asymptomatic.</p><p><b>Conclusions</b>: Most patients had symptomatic disease and received anti-PD1 monotherapy. The intracranial tumor response rate for anti-PD1 monotherapy was higher than expected in this symptomatic population. However, this must be interpreted with caution given the retrospective study design. Nonetheless, this study provides real-world data for melanoma BM in NZ.</p><p><b>Acknowledgments</b>: Cancer Society Central Districts and Graduate Women Otago Scholarship.</p><p><b>References</b>:</p><p>1. Tawbi HA, Forsyth PA, Algazi A, et al. Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain. <i>N Engl J Med</i>. 2018;379(8):722–730. https://doi.org/10.1056/NEJMoa1805453</p><p>2. Long GV, Atkinson V, Lo S, Sandhu, et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. <i>Lancet Oncol</i>. 2018;19(5):672–681. https://doi.org/10.1016/S1470-2045(18)30139-6</p><p>Isabella Wilson<sup>1</sup>, Madeleine Strach<sup>1,2</sup>, Vivek Bhai<sup>1</sup>, Michelle Harrison<sup>1</sup>, Whiter Tang<sup>1</sup>, Peter Grimison<sup>1 </sup></p><p><i><sup>1</sup>Chris O'Brien</i> Lifehouse<i>, Sydney, Australia</i></p><p><i><sup>2</sup>The University of Sydney Faculty of Medicine and Health, Sydney, Australia </i></p><p><b>Background</b>: Gemcitabine and docetaxel (GD) is a standard chemotherapy regimen for soft tissue sarcoma (STS). The GeDDiS phase 3 trial compared GD with doxorubicin in the 1st-line setting, using a 90-min gemcitabine infusion time, reporting a response rate of 20% and median progression-free survival (PFS) of 5.4 months.<sup>1</sup> We examined the real-world efficacy and toxicity of GD in the 1st and later line using a 30-min gemcitabine infusion time.</p><p><b>Methods</b>: A retrospective analysis was conducted of patients with unresectable or metastatic STS receiving ≥1 cycle of GD between July 2018 and October 2022. Data collected included demographics, tumor characteristics, dose intensity, toxicity, response, PFS, and overall survival (OS).</p><p><b>Results</b>: Thirty-eight patients were included for analysis. Median follow-up was 19 months (range 3–30). Histologic subtypes (<i>n</i>) were leiomyosarcoma (20), synovial sarcoma (4), undifferentiated pleomorphic sarcoma (4), other (10). Line of treatment (<i>n</i>) was 1st line (10), 2nd line (13), and ≥3rd line (15). Median number of cycles was 6 (range 1–12). Sixteen patients (42%) required dose modification during the 1st three cycles. Response rate was 42%, including 5% with complete response. At the time of data collection, 33 patients had disease progression and 24 patients had died. PFS (median, 6-month rate) was 4.5 months and 44%. OS (median, 12-month rate) was 15 months and 65% (Figure 1). Grade 3/4 toxicity included neutropenia (8%), febrile neutropenia (3%), and anemia (21%). There was one death possibly attributable to GD.</p><p><b>Conclusion</b>: This data demonstrates efficacy of GD comparable to other published data and favorable toxicity in a real-world population despite shorter gemcitabine infusion time and heavily pre-treated population.</p><p><b>Reference</b>:</p><p>1. Seddon B, Strauss SJ, Whelan J, et al. Gemcitabine and docetaxel versus doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): a randomized controlled phase 3 trial. <i>Lancet Oncol</i>. 2017;18(10):1397-1410.</p><p>Vanessa Wong<sup>1,2,3</sup>, Michael Harold<sup>1</sup>, Bill Karanatsios<sup>2</sup>, Eleonora Kay<sup>4</sup>, Duncan Colyer<sup>4</sup>, Peter Gibbs<sup>1,2</sup>, Shehara Mendis<sup>1,2 </sup></p><p><i><sup>1</sup>Walter &amp; Eliza</i> Hall <i>Institute of Medical Research, Parkville, Australia</i></p><p><i><sup>2</sup>Western Health, St</i> Albans<i>, Australia</i></p><p><i><sup>3</sup>Ballarat Health</i> Services<i>, Ballarat, Australia</i></p><p><i><sup>4</sup>Victorian Comprehensive Cancer Centre (VCCC) Alliance, Parkville, Australia</i></p><p><b>Background</b>: Cancer clinical trials are becoming progressively more complicated and expensive to run, making pragmatic trial designs that limit complexity and/or cost an attractive alternative. One such pragmatic trial design is to conduct a clinical trial using an existing prospective clinical registry to collect patient, treatment, and trial endpoint data. As such, investigator-initiated registry-based clinical trials (Reg-CTs) in cancer populations are emerging in Victoria, supported by the Victorian Comprehensive Cancer Centre (VCCC) Alliance. The perspectives of clinicians and study staff actively involved in this novel trial methodology is of interest.</p><p><b>Methods</b>: A survey was constructed by a panel of clinicians and trials staff. The key focus was barriers and enablers of Reg-CTs. Invitations to participate in the survey were sent to clinical investigators, study coordinators, research nurses, research officers and clinical trials assistants at Victorian sites involved in Reg-CTs.</p><p><b>Results</b>: A total of 42/100 (42%) potential respondents replied, including 18 (43%) investigators and 16 (38%) study coordinators. Thirty-eight (90%) were actively recruiting to Reg-CTs and 20 (48%) had been involved in Reg-CTs for over 2 years. The most important benefits perceived were involvement in trials asking relevant unanswered clinical questions and reduced study-related activity for site staff and patients. Funding was seen as the most important barrier to initiating Reg-CTs, and patient eligibility the largest barrier to enrollment. The single greatest challenge perceived in running Reg-CTs was having clear demarcation of staff responsibility for trial data entry versus registry data entry.</p><p><b>Conclusion</b>: Clinical trial teams overall appreciate multiple benefits of involvement in Reg-CTs, including reduced burden of trial-related procedures. Identifying sufficient patient candidates remains a challenge in Reg-CTs. Though generally considered cost-effective in design and conduct, adequate funding for sites to run these trials was perceived as the single most important barrier to increasing uptake of Reg-CT trials.</p><p>Nicholas Yeo<sup>1</sup>, Sayeda Naher<sup>2 </sup></p><p><i><sup>1</sup>Sutherland Hospital Day Oncology Unit, Caringbah, Australia</i></p><p><i><sup>2</sup>Illawarra Cancer</i> Care <i>Centre, Wollongong, Australia </i></p><p><b>Background</b>: Metastatic breast cancer represents a heterogenous group of patients. Whilst treatment and survival have improved, it remains the 5th most common cause of cancer death in Australia. There is a paucity of data describing the outcomes for patients with metastatic breast cancer treated in regional centers. We aim to describe the experiences within the Illawarra Shoalhaven Local Health District (ISLHD) and provide real-world data on the treatment patterns for these patients, including any factors influencing treatment response.</p><p><b>Methods</b>: We retrospectively reviewed patients with metastatic breast cancer with available electronic medical records from 2011 to 2020, in the ISLHD. Standard demographic, clinicopathological characteristics, treatment data, and outcomes were assessed.</p><p><b>Results</b>: We identified 169 patients with a mean age of 65 (interquartile range [IQR] 53–79). Most women had hormone-receptor positive breast cancers number (<i>n</i> = 104, 62%), followed by HER2-positive (<i>n</i> = 45, 26%), and triple negative breast cancers (TNBC) (<i>n</i> = 20, 12%). Amongst them, 125 (73.5%) of patients had de novo metastatic disease. The average number of lines of treatment received was 2 (IQR 1–3). The most prescribed chemotherapy regimens were taxanes (38.2%), followed by capecitabine (13.2%). Number of metastases (odds ratio [OR] 1.33, 95% CI 1.03–1.71, <i>p</i> = 0.03) and triple negative disease (OR 8.17, 95% CI 4.6–14.5, <i>p</i> &lt; 0.001) were associated with poorer survival. There were no significant differences between de novo and recurrent disease (OR .80, 95% CI .53–1.22, <i>p</i> = 0.30).</p><p><b>Conclusion</b>: This is the first retrospective review describing the treatment patterns and outcomes for patients with metastatic breast cancer within the Illawarra Shoalhaven Local Health District. Whilst treatment and survival have improved, it is evident that triple negative breast cancers remain an aggressive subgroup. There is a need for better treatments for this subgroup of patients.</p>","PeriodicalId":8633,"journal":{"name":"Asia-Pacific journal of clinical oncology","volume":null,"pages":null},"PeriodicalIF":1.4000,"publicationDate":"2023-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/ajco.14010","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Asia-Pacific journal of clinical oncology","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1111/ajco.14010","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q4","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Dianheng Bu1, Javier Torres1,2, Arvind Sahu

1Goulburn Valley Health, Shepperton, Australia

2Department of Rural Health, Shepparton Medical School, Melbourne University, Shepperton, Australia 

Background: Long-term adherence to endocrine therapy is challenging, even in the clinical trial setting. Non-adherence to treatment has an increased risk of cancer recurrence and poorer survival.1 The evidence surrounding the adherence rate to endocrine therapy is sparse overall. The study aims to measure the adherence rate to adjuvant endocrine therapy for early breast cancer and factors contributing to non-adherence in the regional setting.

Methods: A cross-sectional survey was conducted at Goulburn Valley Health between January and June 2023. Female patients receiving adjuvant oral endocrine therapy were included. Adherence was measured using the validated 6-item Simplified Medication Adherence Questionnaire (SMAQ).2

Results: Fifty-one patients (mean age: 60.1 years) participated. The adherence rate, as determined by the SMAQ, was 64.7% (33 out of 51). Age and the number of side effects significantly impacted adherence. Adherent patients had a mean age of 64.3 years, while non-adherent patients had a mean age of 52.4 years (p < 0.001) (Figure 1). The mean number of side effects was 1.8 for adherent patients and 3.8 for non-adherent patients (p < 0.01). No significant differences were found in adherence based on cancer stage, ECOG status, or type of endocrine therapy.

Conclusion: The prevalence of adherence to oral adjuvant endocrine therapy for early breast cancer in the regional population was below 65%. Younger age and experiencing more side effects were associated with lower adherence rates. These findings emphasize the need for early interventions to improve adherence and optimize treatment outcomes in this population.

References:

1. Chlebowski RT, Kim J, Haque R. Adherence to endocrine therapy in breast cancer adjuvant and prevention settings. Cancer Prev Res. 2014;7(4):378-387.

2. Ortega Suarez FJ, Sanchez Plumed J, Lorenzo AD. Validation on the simplified medication adherence questionnaire (SMAQ) in renal transplant patients on tacrolimus. Nefrologia. 2011;31(6):690-696.

Caitlin Cocks1, Sowmya Cheruvu1, Hilary L. Martin1

1Fiona Stanley Hospital, Murdoch, Australia 

Background: COVID-19 mortality rates of up to 24% have been reported in oncology patient cohorts prior to the availability of antiviral medications and vaccinations.1 The COVID-19 pandemic was managed in Western Australia (WA) with strict vaccination mandates and border restrictions. This study aims to assess the impact of COVID-19 infection on oncology patients following the WA border opening in March 2022.

Methods: Medical oncology patients at a tertiary hospital in WA who tested positive for COVID-19 between March and May 2022 were prospectively identified. Case notes were reviewed for data collection, including treatment delays, cancer outcomes, and mortality at 30 days, 60 days, and 12 months post-infection.

Results: Thirty-seven patients tested positive for COVID-19 with 64.9% symptomatic. 78.4% of patients had received at least two COVID-19 immunizations prior to infection. Breast cancer was the most common malignancy (40.5%), followed by non-small cell lung cancer (21.6%). Twenty-nine patients were on active therapy, nine on chemotherapy, and three on immunotherapy. Twenty patients were treated with anti-viral therapy. Eight patients were hospitalized. Treatment delays affected 76.9% of survivors with a median delay of 2 weeks. Ten patients had disease progression in the subsequent 12 months. Five patients reported a second COVID-19 infection and recovered without complication. A total of 13 patients died during follow-up: eight within 30 days, three between 30 and 60 days, and two between 60 days and 12 months. No direct deaths related to COVID-19 were reported.

Conclusion: Reassuringly, there were no deaths directly related to COVID-19. These findings support the use of antivirals and vaccination in this cancer patient population and contrast with the early high rates of COVID-19 mortality.

Reference:

Russell B, Moss CL, Shah V, et al. Risk of COVID-19 death in cancer patients: an analysis from Guy's Cancer Centre and King's College Hospital in London. Br J Cancer. 2021;125(7):939-947. https://doi.org/10.1038/s41416-021-01500-z.

Andrew Fantoni1, Brandon Lau2, Jess McNeill3, Christine Henneker3, Shaouli Shahid4, Wei-Sen Lam

1Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia

2Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Australia

3WA Country Health Service, Central Office, East Perth, Australia

4Centre for Aboriginal Studies, Curtin University of Technology, Bentley, Australia 

Background: Telehealth could offer greater availability to specialist oncology care whilst maintaining patient proximity to home. The Western Australia Country Health Service (WACHS) TeleChemotherapy model was implemented in the Pilbara (Karratha) in September 2019, and in the Kimberley (Broome) and Wheatbelt (Narrogin) in March 2020. We examined the safety of this model with regards to treatment utilization and toxicity outcomes.

Methods: Retrospective data were collected for all patients referred to a WACHS TeleChemotherapy service between September 2019 and February 2022; including patient demographics, cancer diagnosis, treatment intent, cycles and lines of treatment received, treatment delays, and toxicity (adverse events [AEs] as per CTCAEv5.0, deaths, and hospital admissions). Descriptive statistics were used to report safety outcomes.

Results: Of 159 patients referred for treatment, 154 (97%) were accepted. Twenty-one withdrew due to progressive deterioration or preference for metropolitan treatment. The final cohort (n = 133) had a median age of 61 years (range 24–90) with 54% male, 23% Aboriginal and 94% ECOG≤1. A total of 1034 cycles of treatment were delivered, representing 171 lines of therapy. 74% were palliative in intent. Mean dose intensity was maintained at 90% (±14%). Forty-seven treatment lines (27%) were subject to dose reduction, 62% prior to TeleChemotherapy. Fifty-eight lines (34%) were subject to significant dose delay, due to toxicity (53%) or patient preference (38%). Sixty-three lines (37%) were associated with grade ≥3 AEs. There were no significant associations between incidence of AEs and regimen. ≥1 hospitalization occurred in 32% (n = 54) of treatment lines. Eleven deaths were recorded, one related to toxicity. Forty-eight immunotherapy-containing lines were delivered, four lines (8%) with grade≥3 immune-related AEs and no deaths.

Conclusion: This study affirms the safety of the WACHS TeleChemotherapy model including low rates of immune-mediated toxicity. Dose intensity was not compromised, and treatment delays were due to expected toxicities or patient preferences rather than logistical issues.

Jocelyn Finney1, Isabella Wilson1, Ben Felmingham2,3,4, Sanjeev Kumar1, Alina Mahmood1, Lisa Horvath1,5, David Celermajer6, Rachel Conyers

1Chris O'Brien Lifehouse, Sydney, Australia

2Murdoch Children's Research Institute, Melbourne, Australia

3Children's Cancer Centre, The Royal Children's Hospital, Melbourne, Australia

4Cardiac Regeneration Laboratory, Murdoch Children's Research Institute, Melbourne, Australia

5Sydney Medical School, University of Sydney, Sydney, Australia

6Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia

7Department of Paediatrics, University of Melbourne, Melbourne, Australia 

Background: Cancer therapy related cardiac dysfunction (CTRCD) is a significant side effect of several chemotherapy therapies, targeted agents, and immunotherapies integral to treatment. Anthracyclines are an example of a commonly used chemotherapy drug that can cause irreversible early and late onset heart failure which are currently under-documented. In Australia, there are currently no other multi-center, longitudinal studies examining CTRCD in adults. COBLH aims to use the Australian Cardio-Oncology Registry (ACOR) to explore the epidemiology, clinical presentation, and management of CTRCD amongst adults receiving cardiotoxic therapies. Additionally, a biobanking sub-study will attempt to identify gene polymorphisms predisposing individuals to developing anthracycline cardiotoxicity (ACT).

Methods: ACOR is a prospective observational study involving 14 sites across Australia. COBLH patient criteria includes non-palliative, high-risk adult patients treated with cardiotoxic therapies within the last 5 years. Information is obtained from medical records and de-identified upon entry into ACOR. ACOR collects data on patient demographics, medical history, cancer diagnosis and treatment, cardiac assessments, and the management of CTRCD. These instruments are collected at baseline prior to treatment, and at 6-month intervals thereafter. Patients who have developed ACT will be asked to participate in the biobanking study, which will involve a one-off 12 mL peripheral blood draw to be processed at an external site for whole genome exome sequencing and the development of pluripotent stem cell derived cardiomyocytes.

Results: At the time of submission, 19 of 220 recruited patients at COBLH have developed CTRCD. This includes 10 patients with ACT, three with left ventricular dysfunction, three with suspected anthracycline related cardiomyopathies, and three with symptomatic heart failure. ACOR anticipates full registered site capacity to capture 600 patients annually.

Conclusion: COBLH will use ACOR to study long-term CTRCD in adult patients with the biobanking study seeking to prove a causal association between genetic predisposition and ACT.

Acknowledgments: 2,3.

Adriana Gangemi, Bella Nguyen, Wei-sen Lam 

Fiona Stanley Hospital, Perth, Australia 

Background: Anaplastic thyroid cancers (ATCS) are rare aggressive tumors, with extremely poor prognosis despite multimodality treatments. Recently data have shown that a combination of lenvatinib, a tyrosine kinase inhibitor, and pembrolizumab, a checkpoint inhibitor that functions by inhibition of PD-1, can improve response rate and survival in patients with ATC.1,2 Our case showcases a real-world example of this combination treatment.

Methods and results: This case describes a 67-year-old female initially diagnosed in September 2021, presenting with enlarging neck lump. Thyroidectomy and bilateral neck dissection revealed a 42 mm anaplastic thyroid carcinoma, vascular invasion present, extrathyroidal extension in strap muscle, 0/25 lymph node involved, clear margins. This patient completed adjuvant chemoradiation with low dose doxorubicin in December 2021. Disease recurred locally in August 2022 and was deemed not suitable for resection. She was started on pembrolizumab and lenvatinib (20 mg) on September 9, 2022 and her symptoms improved over 4 weeks. The PET scan after 8 weeks of treatment showed almost a complete response and a PET 6 months post-treatment continued to show no convincing evidence of recurrence (Figure 1).

Conclusion: In the context of an extremely poor prognostic disease, this case showed that the combination of lenvatinib and pembrolizumab for ATCs is a game-changer.

References:

1. Dierks C, Seufert J, Ruf J, et al. 1915P The lenvatinib/pembrolizumab combination induces long lasting and complete responses in patients with metastatic anaplastic or poorly differentiated thyroid carcinoma: results from a retrospective study and first results from the prospective phase II ATLEP trial. Ann Oncol. 2020;31:S1085.

2. Luongo C, Porcelli T, Sessa F, et al. Combination of lenvatinib and pembrolizumab as salvage treatment for paucicellular variant of anaplastic thyroid cancer: a case report. Curr Oncol. 2021;28(6):5401-5407.

Grace Gard, Joanna Oakley, Vanessa Wong, Kelsey Serena, Michael Harold, Jo Cockwill, Katya Gray, Helen Anderson, Graeme Down, Judi Price, Peter Gibbs 

WEHI, Parkville, Australia 

Background/aims: In cancer research, there is growing interest in consumers and researchers working together in partnership, yet the practicalities of partnering and how to optimize the co-design process have not been well established. A research group of health care professionals and consumers aimed to create a Personalized Care Plan for patients with newly diagnosed locally advanced rectal cancer. The group has reflected on and documented their experience of co-design.

Methods: Our research group includes consumers from the consumer program at WEHI, multidisciplinary colorectal cancer clinicians, and project officers. Together they created the Personalized Care Plan template, to be provided to patients and their general practitioner. The team's reflections on the co-design process have been captured using Gibbs’ Reflective Cycle. Patients are currently enrolling into a prospective cohort to evaluate the impact of the Personalized Care Plan.

Results: In 2022, over six meetings, we created a two-page Personalized Care Plan to embed into the WEHI-CRC database. Personalized information for individual patients includes disease stage, planned treatment, and scheduled follow-up. Reflection on the co-design process highlighted the importance of establishing expectations, having expertise within the consumer group, and open communication and respect. Challenges included time commitment, power dynamics, diversity of representation, and loss of consumers due to health reasons and time availability. Responses reflected a positive attitude-change of the researchers on the value of consumer input. Both consumers and researchers communicated the high objective and affective value of contributing to the project.

Conclusion: Reflecting on the co-design process for a patient education sheet, we found that essential components of a co-design group are having clear expectations, and strategies to address challenges such as time commitment and entrenched power dynamics. Meaningful input from consumers can objectively improve the outcome of the project while offering positive personal value for consumers and researchers involved.

Tushar Goyal, Jennifer Zhang, Bianca Devitt 

Eastern Health, Melbourne, Australia 

Background: There is growing interest and variability in the choice and timing of antibiotics in transitioning from intravenous to oral therapy. We aimed to understand the treatment and prescribing practices of neutropenic sepsis in our institution.

Methods: A retrospective study was performed at a tertiary hospital across two sites in Melbourne. Adult patients admitted with solid organ malignancies who met the criteria for neutropenic sepsis between January 2020 and December 2021 were eligible. Analysis was performed using descriptive statistics (Table 1).

Results: Sixty-nine patients were identified. Median time from presentation to intravenous antibiotics (ivABx) was 2 h (IQR = 3 h), duration of ivABx was 3 days and total duration of antibiotics was 7 days (IQR = 4 days), with no significant association between the duration and ANC nadir. Sixty-one patients (88%) were culture negative. Fifty-seven patients (83%) received oral (PO) antibiotic tail upon completion of ivAbx, with amoxicillin-clavulanate being the most commonly used (69%), and cephalosporins (13%) being more commonly prescribed after an infective focus was clinically evident. Fifty-three patients (77%) had been clinically stable and afebrile for more than 48 h before being switched to PO antibiotics. Patients with high-severity MASCC scores received a longer duration of total antibiotics and had longer length of stay as inpatients. Seven of these patients (21.2%) required ICU admission, with no patients from the low severity group being admitted to ICU. One patient experienced gastrointestinal adverse effects during deescalated antibiotic therapy. Hospital readmission rates due to recurrence of neutropenic sepsis were similar in patients who received oral antibiotics versus others.

Conclusion: There were notable variations in the selection and duration of antibiotics for the management of culture-negative neutropenic sepsis, in comparison to guidelines. Future investigations could explore the potential benefits of early transition to oral antibiotics to assess their impact on clinical outcomes.

Nikki How1, Dhanvee Kandadai2, Paul Moroz3, David Morris4, David Ransom

1Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia

2Royal Perth Hospital, Perth, Australia

3Western Australian Peritonectomy Service, Joondalup, Australia

4St. George Peritonectomy Unit, Kogarah, Australia 

Background: CRS-HIPEC is a potential treatment option for patients with primary peritoneal malignancies or secondary peritoneal carcinomatosis from dissemination of intraabdominal malignancies. Accumulating evidence for the survival benefits of CRS-HIPEC has contributed to its increasing use, with the aim of prolonged survival and curative intent. This study aims to report the long-term survival outcomes for Western Australian patients who had access to CRS-HIPEC through an Australian interstate program.

Methods: This was a retrospective observational study of Western Australian patients who were transferred to St George Hospital in New South Wales for CRS-HIPEC between 2008 and 2013. Data was obtained from multiple Western Australian registers, supplemented by review of individual patient records. Overall survival was assessed using Kaplan–Meier methods and comparison with log-rank tests.

Results: Forty-two patients received CRS-HIPEC in the period of assessment and had follow-up data available. Median time from diagnosis to CRS-HIPEC was 8.5 months. The most common diagnoses included 13 patients with disseminated peritoneal adenomucinosis (DPAM), 11 with appendiceal adenocarcinoma (AA), nine with colorectal cancer (CRC), three with peritoneal mesothelioma (PM), and three with peritoneal mucinous carcinomatosis (PMCA). Median overall survival across all patients was 50 months, with a 5-year overall survival of 50%. The 5-year overall survival rates for DPAM, AA, and CRC were 75%, 32%, and 40%, respectively. Survival beyond 6 years occurred in 16 patients including nine patients with DPAM, three with AA, two with ovarian malignancies, one with CRC, and one with PM. Patients with PMCA had a comparatively lower survival probability (p < 0.0001).

Conclusion: These outcomes are consistent with results from other centers. The increasing demand led to the establishment of a state CRS-HIPEC Centre in Western Australia. It is worth noting that even with peritoneal metastases, survival beyond 6 years can be achieved in patients with AA, CRC, and PM.

Joshua Hurwitz1,2,3, Greta Beale1, James Pham1,2, Lauren Brown1,3, Anthony Rodrigues1,2,3, Anthony M Joshua1,2,3, Susan Clark1,2,3 

1The Kinghorn Cancer Centre, Darlinghurst, Australia

2St Vincent's Clinical School, University of New South Wales, Darlinghurst, Australia

3Gavan Institute of Medical Research, Darlinghurst, Australia 

Background: Vitamin C is a key cofactor in modulating epigenetic landscapes, genomic stability, and potential cancer growth (1). While rare in the general population, vitamin C deficiency is frequently observed in cancer patients. Denmark and New Zealand research identified inadequate vitamin C levels in cancer patients at baseline and further reduction with treatment (2, 3, 4). This study aimed to determine baseline vitamin C levels in solid cancer patients and the impact of systemic therapy in our own patient cohort.

Methods: This prospective observational study aimed to recruit 31 subjects, 25 metastatic cancer patients and six healthy controls. Patients were recruited at The Kinghorn Cancer Centre. Information collected included demographics, patient characteristics, and malignancy type. Plasma vitamin C concentrations were collected before systemic therapy and after two treatment cycles. The normal vitamin C reference range was 40–80 μmol/L, and low was <40 μmol/L.

Results: Twenty-eight patients were recruited; five did not provide pathology samples for vitamin C levels – of the 23 remaining, 14 patients had a second vitamin C level (Table 1). At baseline, healthy controls had lower vitamin C levels (55 vs. 57) than their cancer counterparts. Vitamin C levels were higher after systemic therapy than before treatment (57 vs. 49). Three patients had weight loss before diagnosis, and one required nutritional supplementation. Treatment included multi-agent chemotherapy (38%), immunotherapy (24%), and single-agent chemotherapy (19%).

Conclusion: In our cohort, vitamin C levels pre- and post-systemic therapy were within physiological parameters, in contrast to prior cancer studies across larger patient cohorts reporting marked vitamin C deficiency (2, 3, 4). Baseline vitamin C was lower, but not significantly, in the healthy controls. Further research with larger sample sizes is required to investigate vitamin C levels in solid malignancies, with systemic therapy and its role in cancer management.

Mandeep Singh Kalsi, Inosha Jayasekara, Frances Barnett 

Northern Health, Epping, Melbourne, Australia 

Background: Chemotherapy that was usually delivered only in hospital environments is now administered primarily in outpatient (OP) environment. There are financial incentives for OP chemotherapy delivery.

Methods: We conducted a retrospective analysis to evaluate the outcomes of patients who were given inpatient (IP) chemotherapy agents for solid organ malignancies over a period of 3 years at a metropolitan hospital.

Results: Small cell cancer of lung primary (20) was the common malignancy in 47 patients who received inpatient chemotherapy. There were 13 patients with gastro-intestinal primary, five patients with testicular cancer, four with breast cancer, three with ovarian cancer, one with cancer of unknown primary, and one with Kaposi sarcoma. Most patients’ inferred Eastern Cooperative Oncology Group (ECOG) performance status was 0% and 15% patients had ECOG status of 3. Thirty-day mortality was 6% and 60-day mortality was 23%. Sixty-day mortality was associated with higher ECOG status. Thirty-nine patients received subsequent chemotherapy sessions.

Conclusion: IP chemotherapy administration was associated with high 60-day mortality. And high mortality was associated with higher ECOG score. More detailed analysis of patient characteristics is required to co-relate with mortality that may help in decision making for oncologists to decide for inpatient chemotherapy.

Muhammad A. Khattak1, Georgina V. Long2, Alexander M. M. Eggermont3, Jeffrey E. Gershenwald4, Dirk Schadendorf5, Paolo A. Ascierto6, Reinhard Dummer7, Axel Hauschild8, Matteo S. Carlino9, Antoni Ribas10, Caroline Robert11, Richard A. Scolyer12, Vernon K. Sondak13, Jonathan E. Cohen14, Jinchun Zhang15, Dmitri Grebennik15, Clemens Krepler15, Jason J. Luke16 

1Fiona Stanley Hospital and Edith Cowan University, Perth, Australia

2Melanoma Institute Australia, The University of Sydney, Mater and Royal North Shore Hospitals, Sydney, Australia

3Faculty of Medicine University Medical Center and Princess Máxima Center, Utrecht, The Netherlands; Comprehensive Cancer Center Munich, Technical University Munich and Ludwig Maximilian University, Munich, Germany

4The University of Texas MD Anderson Cancer Center, Houston, Texas, USA

5University Hospital Essen, University Duisburg-Essen, West German Cancer Centre (WTZ), and German Cancer Consortium, Partner Site Essen, Essen, Germany

6Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy

7University of Zurich and University Hospital Zurich, Zurich, Switzerland

8University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany

9Sydney Medical School, Faculty of Medicine and Health Sciences, University of Sydney, Camperdown, Melanoma Institute Australia, Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, Australia

10Jonsson Comprehensive Cancer Center, UCLA and Parker Institute for Cancer Immunotherapy, Los Angeles, California, USA

11Université Paris Saclay, Le Kremlin Bicêtre and Gustave Roussy, Villejuif, France

12Melanoma Institute Australia, The University of Sydney, Faculty of Medicine and Health, The University of Sydney, Charles Perkins Centre, The University of Sydney, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, Australia

13H. Lee Moffitt Cancer Center, Tampa, Florida, USA

14Sharett Institute of Oncology and The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel

15Merck & Co., Inc., Rahway, New Jersey, USA

16UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, Pennsylvania, USA 

Background: Adjuvant pembrolizumab monotherapy improved recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) in patients with high-risk resected melanoma. Vibostolimab, an anti-TIGIT antibody, demonstrated antitumor activity and manageable safety in combination with pembrolizumab in solid tumors in the phase 1 KEYVIBE-001 study. The efficacy and safety of adjuvant coformulated vibostolimab with pembrolizumab in patients with resected high-risk melanoma in the double-blind, randomized, phase 3 KEYVIBE-010 study (NCT05665595).

Methods: Patients ≥12 years old, with surgically resected stage IIB or IIC (pathologic or clinical), III, or IV cutaneous melanoma, and an ECOG PS of 0 or 1 (>18 years), Karnofsky performance status ≥70 (≥16–<18 years), or Lansky play-performance scale ≥70 (<16 years) and have no evidence of metastatic disease will be enrolled. Patients with ocular, mucosal, or conjunctival melanoma will be excluded. Resection must occur within 12 weeks before randomization. Patients will need to provide an archival tumor tissue sample or newly obtained biopsy of a tumor lesion not previously irradiated. Patients will be stratified by risk (stage IIB/IIC/clinical IIB and stage IIC/IIIA/IIIB vs. stage IIIC/IIID/IV) and region (Asia vs. rest of the world). Approximately 1560 patients will be randomly assigned 1:1 to intravenous coformulated vibostolimab 200 mg with pembrolizumab 200 mg or pembrolizumab 200 mg (2 mg/kg up to 200 mg for adolescents) every 3 weeks for up to 17 cycles. The primary end point is RFS by investigator review. Secondary end points are DMFS by investigator review, overall survival, safety, and quality-of-life. Hazard ratios and 95% CIs will be estimated using a stratified Cox regression model with the Efron methods of handling ties, with treatment as a covariate. Between-treatment differences will be evaluated using a stratified log-rank test. Enrollment is ongoing.

Chee Lee1, Sandy Simon1, Danka Zebic1, Yeh Chen Lee1, Katrina Diamante1, Lenna Lai1, Rachel O'Connell1, Anna deFazio2, Sonia Yip1, Lyndal Anderson3, Bo Gao4, Jeffrey C. Goh5, Kate Webber6, Sally Baron-Hay7, Christopher Steer8, Linda Mileshkin9, Tarek Meniawy10, Janine Lombard11, Martin Stockler1, Michael Friedlander12 

1NHMRC Clinical Trials Centre, The University of Sydney, Camperdown, Australia

2The University of Sydney, The Westmead Institute for Medical Research and Westmead Hospital, Westmead, Australia

3Royal Prince Alfred Hospital, Camperdown, Australia

4Westmead Hospital, Westmead, Australia

5Royal Brisbane and Women's Hospital, Brisbane, Australia

6Monash Medical Centre, Clayton, Australia

7Royal North Shore Hospital, St Leonards, Australia

8Border Medical Oncology Research Unit, Albury, Australia

9Peter MacCallum Cancer Centre, Melbourne, Australia

10Sir Charles Gairdner Hospital, Nedlands, Australia

11Newcastle Private Hospital, New Lambton Heights, Australia

12Prince of Wales Hospital and Royal Hospital for Women, Randwick, Australia 

Aromatase inhibitors (AIs) are used to treat hormone receptor positive (HR+) advanced gynecological cancers. However, response rates are significantly lower than HR+ breast cancer treated with AIs. Alterations in the CDK4/6 pathway and mutations of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) are postulated to cause AI resistance. In HR+ metastatic breast cancer, combination of AI plus CDK4/6 inhibitor (ribociclib) or PIK3CA inhibitor (alpelisib) improves clinical outcomes. PARAGON-II trial (ACTRN12621000639820) aims to evaluate the activity of letrozole plus alpelisib or ribociclib, in PIK3CA mutant (mut) and PIK3CA wild-type (wt) HR+ advanced gynecological cancers, respectively.

This is a phase II, open label, basket trial, with six cohorts of common and rare HR+ gynecological cancers. Target population is patients with recurrent/metastatic cancer, with good performance status and for whom chemotherapy is not indicated. PIK3CA mutational status is determined by tissue or ctDNA analysis. The study consists of PIK3CA mut endometrial cancer (cohort A) and any other PIK3CA mut tumors (F). PIK3CA wt/unknown cohorts consist of endometrial cancer (B), platinum resistant high-grade epithelial ovarian cancers (C), endometrial stromal sarcoma, leiomyosarcoma, granulosa cell tumors (D), and low grade serous ovarian cancers (E). In the PIK3CA mut cohorts (A and F), participants will receive letrozole 2.5 mg plus alpelisib 300 mg daily until disease progression or intolerance. In the PIK3CA wt/unknown cohorts (B–E), participants will receive letrozole 2.5 mg daily plus ribociclib 600 mg daily (3-weeks-on/1-week-off). A total of 182 participants will be recruited. The primary endpoint is objective response rate. Secondary endpoints include disease control rate, duration of response, 6-month progression free survival (PFS), and median PFS, patient reported outcomes and adverse events. Tumor and blood samples are being collected for translational studies.

PARAGON-II trial started recruitment in May 2022. As of June 30, 2023, 64 participants have been recruited from 13 sites. Recruitment is expected to be completed by December 2024.

AIs are used to treat hormone receptor positive (HR+) advanced gynecological cancers. However, response rates are significantly lower than HR+ breast cancer treated with AIs. Alterations in the CDK4/6 pathway and mutations of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) are postulated to cause AI resistance. In HR+ metastatic breast cancer, combination of AI plus CDK4/6 inhibitor (ribociclib) or PIK3CA inhibitor (alpelisib) improves clinical outcomes.

Su Saint Lee1,2,3, Wei Chua1,4,5, Nancy Huang1,2,3, Weng Ng1,4,5, Aflah Roohullah1,2, Annette Tognela2, Ray Asghari3, Mahtab Farzin6, Tara Roberts4,5,7, Therese Becker4,5,7, Bin Wang6, Soon Lee5,6,7, Stephanie Lim3,4,5

1Department of Medical Oncology, Liverpool Hospital, Liverpool, Australia

2Department of Medical Oncology, Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, Australia

3School of Medicine, Western Sydney University, Campbelltown, Australia

4Department of Medical Oncology, Bankstown-Lidcombe Hospital, Bankstown, Australia

5Department of Anatomical Pathology, Liverpool Hospital, Liverpool, Australia

6Ingham Institute of Applied Medical Research, Liverpool, Australia

7South Western Clinical School, University of New South Wales, Liverpool, Australia

Background: Tumor mutational profile affects prognosis in metastatic colorectal cancer (mCRC) but it remains unclear why some tumors behave exceptionally well or poorly. We aim to identify genes associated with exceptional cases using targeted next generation sequencing (NGS).

Methods: Patients with RAS/BRAF-wildtype and MMR-proficient mCRC from the South Western Sydney Anatomical Pathology tissue bank from 2010 to 2018 with overall survival of <6 months or >3 years or atypical treatment responses were identified. Clinicopathological variables and survival outcomes were obtained. Random sample of eight patients underwent testing with a 30-gene (Qiagen) or 50-gene or 161-gene (ThermoFisher) NGS panel to establish workflow, feasibility and obtain preliminary data. Mutations identified were classified according to ESCAT tiers of significance. All cases will have 161-gene panel NGS.

Results: Fifty-two good performers (GP) and 14 poor performers (PP) were identified. Median age was 65 years in GP and 51 in PP. Tumors were left-sided in 87% of GP and 77% of PP. Median overall survival from diagnosis of metastatic disease was 56.4 months (20.6–127.1 months) in GP and 4.8 months (1.4–15.4 months) in PP. 58% of GP and 14% of PP had ≥2 lines of chemotherapy. To date we have NGS data for six GP and two PP patients (four patients with 30-gene, two with 50-gene, and two with 161-gene panels). Two GP patients had TP53 mutations, another a PIK3CA mutation (all Tier II), while a fourth had an EGFR mutation of unknown significance. No mutations were detected in 2GP and two PP patients. 161-Gene panel testing is underway for patients who so far had limited testing.

Conclusion: Targeted NGS panels may identify genes in exceptional performers in RAS/BRAF-wildtype mCRC, whose overall survival varies by more than 10-fold. Preliminary data shows differences in TP53 and PIK3CA in GP. Final 161-gene NGS results of all cases are underway.

Yeh Chen Lee1, Elizabeth Barnes1, Katherine Francis1, Danka Zebic1,2, Sandy Simon1, Gemma Blunt1, Pearly Khaw3, Viet Do4, Elizabeth Christie3, Rachel Delahunty3, Rebecca Mercieca-Bebber1, Sonia Yip1, Jeffrey Goh5, Janine Lombard6, Kate Webber7, Sudarshan Selva-Nayagam8, Ganessan Kichenadasse9, Tarek Meniawy10, Michelle Harrison11, Michelle Wilson12, Martin Stockler1, Linda Mileshkin

1NHMRC Clinical Trials Centre, University of Sydney, Camperdown, Australia

2Prince of Wales Hospital and Royal Hospital for Women, Randwick, Australia

3PeterMacCallum Cancer Centre, Melbourne, Australia

4Liverpool Cancer Therapy Centre, Liverpool, Australia

5Royal Brisbane and Women's Hospital, Brisbane, Australia

6Calvary Mater Newcastle, Newcastle, Australia

7Monash Medical Centre, Clayton, Australia

8Royal Adelaide Hospital, Adelaide, Australia

9Flinders Medical Centre, Bedford Park, Australia

10Sir Charles Gairdner Hospital, Nedlands, Australia

11Chris O'Brien Lifehouse, Camperdown, Australia

12Auckland City Hospital, Auckland, New Zealand 

Background: Despite primary surgery, adjuvant chemoradiation, and chemotherapy, people with high-risk endometrial cancer (EC) remain at significant risk of relapse, following which the median survival time is approximately 12 months. Immune checkpoint inhibitors have shown activity in advanced EC. It is recognized that radiation and chemotherapy can have immune-stimulating properties and can synergize with immunotherapy, providing rationale for combining these treatments in high-risk curable EC. The aim of ADELE is to determine if adding tislelizumab to standard adjuvant chemotherapy after post-operative pelvic chemoradiation for high-risk EC improves outcomes.

Methods: This is an open label, non-comparative, multicenter, randomized (2:1) phase 2 trial (Figure 1). The target population is people with high-risk FIGO stage IA–IVA EC who have had primary surgery and are planned for adjuvant pelvic chemoradiation followed by chemotherapy. Participants will be registered prior to commencing adjuvant pelvic chemoradiation and vaginal vault brachytherapy (if cervical stromal invasion) and subsequently randomized to one of two treatment groups in a ratio of 2:1 (experimental to control). The control group will receive chemotherapy (carboplatin plus paclitaxel) for four cycles. The experimental group will receive tislelizumab with chemotherapy for four cycles, followed by tislelizumab alone for another eight cycles. The primary outcome is failure free survival at 1 year. Secondary outcomes include feasibility, adverse events, effects on patient-reported outcomes, failure free survival, and overall survival. Tumor and blood samples will be collected for translational studies, with correlative objectives including impact of adjuvant therapy on ctDNA levels and potential predictive or prognostic biomarkers for response to immunotherapy.

Progress: ADELE started recruitment in March 2022 and will enroll 135 participants. As of June 30, 2023, 17 participants have been randomized from 11 sites. Clinical trial identifier: ACTRN12621000273886.

Acknowledgments: ADELE is an investigator-initiated study, funded by an MRFF Research Grant and Beigene Ltd.

Meghana Maddula1,2,3, Peter Fox1,4 

1Central West Cancer Centre, Orange Health Service, Orange, Australia

2Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, Australia

3Blacktown Haematology and Cancer Centre, Blacktown Hospital, Sydney, Australia

4Western Sydney University, Bathurst Clinical School, Bathurst, Australia 

Background: Positron emission tomography (PET) is integral to the diagnosis, monitoring, and treatment of malignancy. Whilst PET utilization has increased, inequity in access remains across Australia. The OECD average in 2019 was 4 PET per 1000 persons. In this study, we aim to identify local trends in PET usage and evaluate geospatial disparity.

Methods: In this retrospective population study, we evaluated PET scan utilization for Medicare funded oncological indications between 2001 and 2022. Data was extracted from public Medicare statistics and stratified by population density, state/territory, and tumor type. Additional Remoteness Area data were obtained from 2018 to 2021 and AIHW data informed cancer incidence.

Results: Over 22 years, 1,124,431 PET scans were attended, with 2,746,117 incident cancers. This corresponds to 2.2 PET per 1000 persons and 40.9 per 100 cancers but had increased to 5.4 per 1000 persons and 89.2 per 100 cancers by the end of study period (Table 1). PET uptake per capita increased nationwide with a compound annual growth rate of 18.4%. The greatest per capita usage of PET scans was in New South Wales followed by Western Australia, with the greatest annualized growth in the Northern Territory (28.6%) and Tasmania (24.2%). PET scanner availability by state/territory did not correlate with PET utilization (R = .15, p = 0.72). PET scans per 100 incident cancers was highest in lymphoma (201.9) and head and neck (147.0), and lowest in neurological (14.1) and colorectal cancers (39.4). By Remoteness Area, PET per 1000 persons was lower in remote (3.1) and very remote (1.9) compared to major cities (4.4).

Conclusion: Australian PET scan usage has increased significantly since the introduction of Medicare subsidized PET in 2001 and is now above the OECD average. Geospatial disparity remains in PET usage between states/territories and based on rurality. Further research into variations in PET utilization is required to bridge the gap.

Rhiannon Mellor1,2,3*, Jessica Smith4,5*, Anna Mislang6,7* 

*All first authors

1Chris O'Brien Lifehouse, Camperdown, Australia

2University of New South Wales, Sydney, Australia3The Garvan Institute of Medical Research, Darlinghurst, Australia

4Macquarie University Hospital, Macquarie University, Australia

5Western Sydney University, Parramatta, Australia

6Department of Medical Oncology, Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, Australia

7College of Medicine and Public Health, Flinders University, Bedford Park, Australia 

Background: Medical oncologists operate in an increasingly complex environment, regardless of their practice location. Over the last 3 years, the COVID-19 pandemic has increased this complexity. International studies report that up to 71% of oncologists exhibit evidence of burnout,1 and this has worsened since COVID-19.2 Burnout negatively impacts the psychological well-being of the individual healthcare professional and the level of care they provide, significantly costing individuals and the broader healthcare organization. This is a critical issue that must be urgently addressed.

Aims and objectives: This study aims to determine the current prevalence of burnout amongst trainees and Fellows of the Royal Australasian College of Physicians (FRACP) in Medical Oncology in Australia. Secondary objectives are to (1) examine the drivers of burnout among oncologists in Australia, (2) assess the impact of burnout, and (3) collect data on potential interventions to address burnout and clinician wellbeing, as identified by oncologists.

Methods and results: In this cross-sectional study, FRACP and trainee medical oncologists in Australia will be invited to complete an anonymous online questionnaire. The questionnaire will assess burnout with the Maslach Burnout Inventory (MBI) and measure the consequences of burnout using the Patient Health Questionnaire-4 (PHQ-4) to screen for anxiety and depression, and the Stanford self-valuation tool. Factors that contribute to both wellbeing and burnout will also be examined, along with potential work-based interventions to improve wellbeing.

Conclusion: The prevalence and drivers of burnout amongst medical oncologists in Australia are poorly understood. This study will provide essential data for a solutions-based, multifactorial approach to improve clinician wellbeing.

Acknowledgments: Thank you to Bethan Richards and Jia Liu for their input on questionnaire development.

References:

1. Banerjee S, Califano R, Corral J, et al. Professional burnout in European young oncologists. Ann Oncol. 2017; 28(7):1590-1596.

2. Banerjee S, Lim KHJ, Murali K, et al. The impact of COVID-19 on oncology professionals. ESMO Open. 2021;6(2):100058.

Hanh Nguyen, Bella Nguyen 

Fiona Stanley Hospital, Murdoch, Australia 

Background: For patients with borderline resectable (BR) and locally advanced (LA) pancreatic cancers, neoadjuvant chemotherapy (NAT) is routinely offered to downstage their disease for potential pancreatectomy. In many cases, however, pancreatectomy remains contraindicated after NAT and alternative therapies are required. This study aims to retrospectively compare the characteristics of patients with BR and LA pancreatic cancer who received versus those who did not receive pancreatectomy after NAT within the South Metropolitan Health Service (SMHS) in Perth Australia from 2015 to 2022.

Methods: Patient electronic medical records (eMR) were used to identify patients who received NAT for their BR or LA pancreatic cancer. Data on patient demographics, clinical characteristics, and adjuvant therapies received post-NAT were attained for further analysis.

Results: There were 19 patients who received NAT to downstage their BR or LA disease for potential resection. Out of these patients, 36.8% (n = 7) successfully underwent pancreatectomy whilst 57.9% (n = 11) did not qualify for surgery and 5.3% (n = 1) were lost to follow-up. Patients undergoing pancreatectomy were more likely to have a recorded ECOG status of zero (85.7% vs. 54.5%). Other characteristics were comparable between patients who underwent pancreatectomy versus those who did not undergo pancreatectomy, respectively; mean age (61 vs. 59 years), sex (male = 72.7% vs. 71.4%), tumor location (pancreatic head = 90.9% vs. 100%), and CA-19-9 level at diagnosis (2448.5 vs. 2999.4). Adjuvant chemotherapy (42.9% vs. 91%) and radiation therapy (28.6% vs. 63.6%) were less frequently utilized for patients who did versus those who did not receive pancreatectomy, respectively.

Conclusion: Aside from ECOG status, there were no significant demographic or clinical differences amongst BR or LA pancreatic patients who did versus those who did not undergo pancreatectomy after NAT. Patients who underwent pancreatectomy were less likely to require subsequent adjuvant chemotherapy and radiation therapy.

Hanh Nguyen, Bella Nguyen 

Fiona Stanley Hospital, Murdoch, Australia 

Background: Whilst surgery is the only curative treatment modality for pancreatic cancer, local blood vessel involvement often precludes its resectability and classifies the disease as borderline resectable (BR) or locally advanced (LA). Neoadjuvant chemotherapy (NAT) is often offered to downstage BR and LA pancreatic cancers for potential resection. This study aims to retrospectively review the clinical management of patients with BR and LA pancreatic cancer within the South Metropolitan Health Service (SMHS) in Perth Australia from 2015 to 2022. Primary outcomes include pancreatectomy rates post-NAT, patient progression-free survival, and overall survival (OS).

Methods: Patient electronic medical records (eMR) were used to identify all pancreatic cancer patients who received FOLFIRINOX or GnP (nanoparticle albumin-bound paclitaxel) as NAT for their BR or LA pancreatic cancer within the SMHS from 2015 to 2022.

Results: Of 19 patients who received NAT, 15 received FOLFIRINOX and four received GnP. Of those who received FOLFIRINIX, 40.0% (n = 6) successfully underwent pancreatectomy whilst 53.3% (n = 8) did not qualify for surgery and 6.7% (n = 1) were lost to follow-up. Of those who received GnP, only one patient underwent pancreatectomy. Reasons precluding pancreatectomy after NAT included inadequate NAT response/progressive disease (63.6%, n = 7) and poor performance status (36.4%, n = 11). Pancreatectomy specimens were staged from IA–IIB, most were grade 2 (85.7%; n = 6) and resected with negative margins (R0) (57.1%, n = 4). The median survival (30 vs. 13 months) and median progression-free survival (20 vs. 7 months) for patients who underwent pancreatectomy were higher than those who did not undergo pancreatectomy, respectively.

Conclusion: Pancreatectomy was performed in two in five patients after FOLFIRINOX NAT and one in four patients after GnP NAT. These rates reflect real-world patients who are less likely to be surgically fit compared to published trial data such as CONKO-007.7.

Ek Leone Oh, Elizabeth Steinepreis, Hilary Martin 

Fiona Stanley Hospital, Murdoch, Australia 

Background: Management for lobular breast cancer, which comprises approximately 10% of breast cancer, is generally extrapolated from trials comprised predominantly of ductal breast cancer patients. This study aims to investigate treatment outcomes of patients treated for metastatic lobular breast cancer.

Methods: We performed a retrospective review of patients diagnosed with metastatic lobular breast cancer at a single center between January 2017 and April 31, 2023. Clinical benefit from treatment was defined as stable disease, partial response, or complete response.

Results: Twenty-one patients with metastatic lobular breast cancer were identified. Sixteen (76.2%) had hormone-receptor positive HER2-negative breast cancer (HR+ HER2−), two (9.5%) had triple-negative breast cancer, and three (14.3%) had HER2-positive hormone-receptor positive (HER2+HR+) disease. Treatment and outcomes for HR+HER2− subgroup are shown in Table 1. Among the patients with HR+HER2− subgroup, 14 (87.5%) received a combination of CDK4/6 inhibitors and non-steroidal aromatase inhibitor (NSAI) of whom 13 started this as first-line therapy for metastatic disease and six (42.9%) remain on this treatment at data-cutoff. Seven (43.8%) patients with HR+HER− disease received chemotherapy at some point in their treatment. Both patients with triple-negative lobular breast cancer experienced rapidly progressive disease on all agents trialled (olaparib, paclitaxel, carboplatin/gemcitabine, and eribulin). The three patients with HER2+HR+ disease received a combination of taxane or vinorelbine, pertuzumab, and trastuzumab as first-line therapy. All remain on this treatment with a median duration of 34 months (range 19–68 months) at data-cutoff.

Conclusion: HR+ HER2− lobular breast cancer treated with CDK4/6 inhibitors and NSAI had similar PFS compared to the registration trial data supporting the use of this combination for lobular cancer. There were also some responses seen to chemotherapy, with all patients who received capecitabine obtaining clinical benefit. Patients with HER2-positive disease had excellent outcomes, while outcomes were poor for triple-negative lobular breast cancer. Both patients with triple-negative lobular breast cancer experienced rapidly progressive disease on all agents trialed (olaparib, paclitaxel, carboplatin/gemcitabine, and eribulin). The three patients with HER2+HR+ disease received a combination of taxane or vinorelbine, pertuzumab, and trastuzumab as first-line therapy. All remain on this treatment with a median duration of 34 months (range 19–68 months) at data-cutoff.

Hayley Roberts1,2, Benjamin Solomon2, Susan Harden2, Senthil Lingaratnam2, Marliese Alexander

1Alfred Health, Melbourne, Australia, Australia

2Peter MacCallum Cancer Centre, Melbourne, Australia 

Background: Lung cancer remains the leading cause of cancer death in Australia, despite recent progress in immunotherapy, tyrosine kinase inhibitors (TKIs), and radiotherapy. Thirty-day mortality after systemic anti-cancer therapy (SACT) has been suggested as a quality indicator for lung cancer and introduced as a benchmark in the UK. However, while multiple international researchers have reported 30-day mortality rates, the exact method for calculating these rates differs widely between studies.

Methods: Patients ≥18 years of age with a histologically confirmed diagnosis of lung cancer who received palliative systemic anti-cancer treatment at Peter MacCallum Cancer Centre between January 2015 and December 2022 were identified using a lung cancer database (AURORA). Information was collected on patient, disease, and treatment characteristics. An annual 30-day mortality index following SACT was calculated (patients who died within 30 days of SACT, as a proportion of all patients given palliative-intent treatment for lung cancer in that year).

Results: We found annual rates of 30-day mortality after palliative-intent SACT for lung cancer of between 9% and 15% (Figure 1). Patients who received SACT within 30 days of death were more likely to have received TKIs or immunotherapy, and to have had an ECOG performance status of two or more, and less likely to have had small cell histology. 

Conclusions: The rates of 30-day mortality in our center are comparable to internationally published rates. While 30-day mortality after SACT is a useful benchmark, several factors must be taken into account when comparing between institutions, including SACT intent (curative or palliative), number of lines of treatment, histological subtype, and type of treatment (especially for TKIs). Ensuring consistent methods of calculation is important, especially reporting time period and denominator population (total number of patients on SACT over a period of time, or total number of patients who have died).

Rahul Solanki1,2, Dylan Bartholomeusz1,2, Michael Cilento3,4, Eve Hopping3, Harsh Kanhere1,2,3, Timothy Price1,2,3 

1Royal Adelaide Hospital, Adelaide, Australia

2The University of Adelaide, Adelaide, Australia

3The Queen Elizabeth Hospital, Woodville South, Australia

4South Australian Immunogenomics Cancer Institute, The University of Adelaide, Adelaide, Australia

Background: A 78-year-old lady was referred for an FDG PET scan for staging of esophageal cancer. She was receiving endoscopic surveillance for Barrett's esophagus. Her last endoscopy showed two nodular lesions in the distal esophagus and biopsies revealed HER2 positive poorly differentiated esophageal adenocarcinoma.

Methods: The CT staging scan showed distal esophageal thickening and enlarged regional lymph nodes. Her 18FDG staging PET scan showed increased FDG uptake in the distal esophagus at the primary site but also revealed extensive non-regional bilateral FDG avid lymphadenopathy and skeletal lesions suggesting metastatic disease.

Results: After review in the upper gastrointestinal (UGI) multidisciplinary team (MDT) meeting, noting a past history of sarcoidosis, she underwent biopsy of the right axillary and right inguinal lymph nodes, which showed well-formed non-necrotizing granulomas suggestive of granulomatous lymphadenitis. There were no malignant cells detected. Following repeat review in the UGI MDT, the consensus decision was that this was a case of localized esophageal cancer with FDG avid nodal and bony lesions likely due to sarcoidosis. She was commenced on trimodality treatment with neoadjuvant chemoradiotherapy (chemoRT), which included weekly administration of dexamethasone along with chemotherapy, with the plan for subsequent curative esophageal resection. The repeat 18FDG PET scan post-chemoRT showed significant reduction of FDG uptake in the primary site and also reduction of the widespread 18FDG avid lymph nodes and bony lesions. The sarcoidosis is thought to have responded with dexamethasone with a plan for close follow-up Figure 1).

Conclusion: This case emphasizes the complexity of decision making in staging patients with malignancy and concomitant inflammatory granulomatous disease and reveals the benefit of clinical history, MDT discussion and biopsy when clinical discrepancies arise.

Jennifer Soon1,2, Fanny Franchini1, Maarten IJzerman1,3, Grant McArthur1,2,4 

1University of Melbourne, Parkville, Australia

2Peter MacCallum Cancer Centre, Melbourne, Australia

3Erasmus School of Health Policy and Management, Rotterdam, The Netherlands

4Victorian Comprehensive Cancer Centre Alliance, Melbourne, Australia 

Purpose: With healthcare costs rising, focus is shifting toward maximizing health outcomes per dollar. One approach to optimizing value is through de-escalation, which is the rationalization of routine treatment without compromising patient outcomes. Since 2013, successful clinical trials of high-cost immune checkpoint, BRAF and MEK inhibition have led to these drugs becoming ubiquitous in melanoma management. Pipeline therapies such as relatlimab and tumor infiltrating lymphocyte (TIL) therapy are expected to have a similar or even greater cost. The purpose of this scoping review is to establish current and emerging opportunities to de-escalate the use of systemic therapies in the treatment of cutaneous melanoma. It will also seek to comment on the proportion of studies that include patient-reported outcomes and quality of life measures. We hypothesize that neoadjuvant and response-directed strategies will be identified as well as the emerging potential of prognostic and predictive tools.

Methods: A systematic search strategy has been developed for MEDLINE and PubMed from January 1, 2013 to June 30, 2023. Additional texts will be sourced from grey literature and reference scanning. Two authors will screen abstracts and full-texts facilitated by the Covidence software. Disagreements will be resolved by consensus or a third reviewer. The first author will perform data extraction whilst a second author will review a random selection of papers to ensure consistent interpretation. De-escalation strategies will be categorized by concept, potential impact on resource utilization and patient outcomes, and strength of evidence. Data will be synthesized qualitatively and quantitatively. Results will be reported following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR). Preliminary results will be available in August 2023. The final results of this scoping review will directly inform a melanoma consumer and clinician survey exploring their perspectives on de-escalation strategies.

Joanne Tang, Sudarshan Selva-Nayagam 

Central Adelaide Local Health Network, Adelaide, Australia 

Background: Sequencing of adjuvant treatment and the use of weekly cisplatin with concurrent radiotherapy in endometrial cancer has not been clearly established. This audit aims to compare toxicity and treatment completion rates in patients who undergo chemotherapy then radiotherapy ± chemotherapy (ChtRt) to patients who undergo radiotherapy ± chemotherapy then chemotherapy (RttCh), and to establish the toxicity and completion rates of concurrent weekly cisplatin (1WC) versus 3-weekly cisplatin (3WC) during radiotherapy (CRTx).

Methods: Patient demographics, treatment regimens, and toxicity profiles were collected for endometrial cancer patients who were treated with adjuvant radiotherapy ± chemotherapy, with chemotherapy preceding or following. Data from patient medical records were extracted across two tertiary hospitals between January 2015 and October 2021 and analyzed.

Results: The study included 19 patients in the ChtRt and 15 patients in the RttCh group. Overall toxicity outcomes were similar between ChtRt and RttCh groups, except patients in the ChtRt group had higher rates of diarrhea during radiotherapy (74% vs. 20%). Hematological toxicity during CRTx was higher in ChtRt group (n = 6) (64% vs. 11% all grade toxicity). ChtRt patients had a higher rate of chemotherapy cessation during radiotherapy (67% vs. 18%), whilst RttCh patients had a higher rate of chemotherapy interruption/cessation (60% vs. 42%) (Table 1). Across both groups, 16 patients had 1WC, and five had 3WC. Overall grade >3 toxicity was higher in the 1WC group (56% vs. 20%), with 31% of patients ceasing 1WC during CRTx, compared to none in the 3WC.

Conclusion: In this small retrospective study, overall toxicity was similar across sequencing approaches, however treatment completion of each modality was higher in whichever modality was started first. 1WC CRTx had a higher toxicity profile compared to 3WC, and the ongoing use of this modified protocol in the absence of clear benefit is currently under review.

Christina Teng, Anew Killen, Charlotte Lemech 

Scientia Clinical Research, Randwick, Australia 

Aim: To identify and summarize the demographic factors of patients referred and enrolled to Scientia Clinical Research (SCR) Phase 1 oncology trials.

Background: Phase 1 trials are first-in-human studies evaluating the safety, efficacy, and pharmacokinetics of newly developed medications. SCR is a dedicated Phase 1 trial unit located in NSW, Australia.

Methods: Descriptive statistics were used to summarize de-identified data from the clinical records of patients referred to the SCR Phase 1 unit.

Results: In the 2022 calendar year, 168 patients were referred to SCR for an opinion about early phase clinical trials, from 51 referrers. Of those referred, 139 patients had a first consultation. Figure 1 summarizes the outcomes of the patients referred. The mean age at time of referral was 59 years, (standard deviation 12, median 61, range 12–83). The most common primary diagnoses were upper gastrointestinal (29%), colorectal (23%), and gynecological (23%) cancers. Of the patients where referral location was available, the majority resided in metropolitan Sydney (47%). Regional NSW residents comprised 29% and interstate referrals 9%. Five initial consultations were conducted with the assistance of an interpreter (three Mandarin, one Cambodian, one Auslan). Of these, two patients proceeded to trial participation. In 2022, 52 patients consented and commenced trial screening over 21 trials; 29 were female (56%). Median age was 71. Residents of metropolitan Sydney comprised 50% of screened patients. Four patients were ineligible at screening; 48 commenced trial treatment. Median time from referral to starting trial was 14 days. The most common diagnosis of recruited patients was gynecological cancer (31%), followed by upper gastrointestinal (21%) and colorectal (20%) cancers.

Conclusion: In 2022, referrals and trial participants to phase 1 oncology trials at SCR represented a diverse geographical population. Patients from culturally and linguistically diverse backgrounds were under-represented.

Lucy Porter1, Phillip Parente2,3, Grace Gard4,5, Benjamin Brady6, Wasek Faisal7,8, Dishan Herath4, Margaret Lee2,3, Peter Gibbs4,5, Ben Markman5,6, Rachel Wong2,3 

1School of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia

2Department of Medical Oncology, Eastern Health, Melbourne, Australia,

3Eastern Health Clinical School, Monash University, Melbourne, Australia

4Department of Medical Oncology, Western Health, Melbourne, Australia,

5Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia

6Department of Medical Oncology, Cabrini Health, Melbourne, Australia

7Grampians Integrated Cancer Service, Grampians Health, Ballarat, Australia

8School of Health, La Trobe University, Melbourne, Australia 

Background: The treatment of early-stage non-small cell lung cancer (NSCLC) is evolving, with data increasingly supporting the role of immunotherapy and targeted agents in adjuvant and neo-adjuvant settings. Atezolizumab, a PD-L1 inhibitor, has been PBS-subsidized since November 2022 for resected Stage II–IIIa NSCLC patients with PD-L1 ≥ 50 who are EGFR/ALK wild-type and treated with platinum-based chemotherapy. The proportion of patients who receive molecular testing and are eligible for PBS-subsidized atezolizumab treatment in Australian hospitals is unknown.

Methods: Patients enrolled in the multi-site INHALE lung cancer registry between February 2020 and January 2023 at four Australian sites (n = 448) were retrospectively analyzed. NSCLC patients who underwent surgical resection were included (n = 115) and analyzed for stage, molecular testing, treatment details, and outcomes.

Results: A total of 90/115 resected NSCLC patients were treated with curative intent. 63/90 received surgery alone, 25/90 received adjuvant systemic therapy and/or radiotherapy, and 2/90 received neo-adjuvant systemic therapy and/or radiotherapy. 7/90 experienced disease recurrence/progression.

A total of 70/90 curative-intent resected patients had PD-L1 testing. 62/70 within 30 days and 67/70 within 90 days of surgery. 56/90 underwent genomic testing. Testing results are shown in Table 1.

Of 70 PD-L1 tested patients, one was eligible for adjuvant atezolizumab as per current PBS criteria. 67/70 were ineligible (PD-L1 < 50%, ineligible-stage, no platinum-based chemotherapy), and 2/70 were PBS-eligibility unknown (genomic testing not performed). PD-L1 and genomic testing rates were higher in metropolitan versus regional settings (70%–90% vs. 40% and 15%–88% vs. 50%, respectively), and private versus public settings (90% vs. 70% and 88% vs. 15%–65%, respectively).

Conclusion: In this real-world analysis of resected NSCLC patients, very few routine care patients were eligible for PBS-subsidized atezolizumab. We anticipate that PD-L1 and genomic testing rates will increase in response to the availability of adjuvant atezolizumab on the PBS and as evidence supporting neo-adjuvant/adjuvant use of immunotherapy and/or targeted agents continues to emerge.

Joanne Tang1, Brian Stein2,3 

1Central Adelaide Local Health Network, Adelaide, Australia

2Icon Cancer Centre, Adelaide, Australia,

3University of Adelaide, Adelaide, Australia 

Background: EviQ is a consensus driven evidence-based repository of cancer treatment protocols widely used in Australia and serves as a de facto standard. A standardized assessment of the clinical benefit of the protocols published on EviQ has not been undertaken. Based on the references in EviQ we aimed to determine the feasibility of scoring all solid tumor protocols to assess the benefit of protocols and explore correlations between ESMO-MCBS scoring and treatment indication, tumor stream and trial endpoint.

Methods: We graded all solid tumor protocols published on EviQ until April 31, 2022 with ESMO-MBCS version 1.1, excluding functional duplicates (e.g., weight-based vs. flat dose immunotherapy). We only used the “Evidence” section for reference as this is the evidentiary justification for protocol publication. In addition to technically unevaluable protocols, we regarded protocols using phase II data in common scenarios, lacking a standard comparator, or based on conglomerated data as unevaluable.

Results: Of 478 included protocols, 301 (63%) were evaluable. Intent was curative in 97 and palliative in 204. Overall, 140 (47%) of evaluated protocols met the MCBS threshold for significant benefit; with 75% of curative but only 33% of palliative protocols being of “substantive benefit”. Of the 57 protocols that had toxicity or quality-of-life benefit, 88% were palliative and 47% were chemotherapy regimens. Of low benefit protocols, 49% were chemotherapy-based, and response-based outcomes were over-represented in palliative protocols. There was no difference in scores by tumor stream. Of the 177 unevaluable protocols, 31% lacked a standard comparator and 42% used phase II data in non-orphan settings.

Conclusion: Within EviQ, 47% of evaluable protocols are ESMO-MCBS “substantial benefit”; most are curative. Few protocols gained points for less toxicity. We suggest adding ESMO-MCBS score to EviQ to aid in regimen consideration.

Sophie Tran1,2, Cassana Moore2, Danielle Roscoe2, Hieu Chau2, Sachin Joshi2,3, Quan Tran2, Rohan Nair2,3, Mahesh Iddawela2,3,4 

1Eastern Health, Box Hill, Australia

2Latrobe Regional Health, Traralgon, Australia

3Alfred Health, Melbourne, Australia

4Department of Rural Health, Monash University, Traralgon, Australia 

Background: Brain metastases (BM) is a common occurrence in non-small cell lung cancer (NSCLC) and is associated with high morbidity and mortality. The advent of new systemic treatments and radiation techniques has led to improved outcomes in this prognostically poor group. As such, early detection at diagnosis and surveillance is important. This project aims to review the characteristics, current practice, management and outcomes of lung cancer BM at a single regional center to help inform development of local protocols.

Methods: Patients with lung cancer BM were identified from the William Buckland Radiotherapy Centre medical records and Latrobe Regional Health lung cancer database. A total of 101 patients were identified between 2017 and 2022. Demographic and clinical information data were collected.

Results: The median age at lung cancer diagnosis was 68 years, 59% were male and 71% ECOG 0 or 1. At initial diagnosis, 66% (67/101) had metastatic disease and of these patients, 60% (40/67) had BM. Initial brain imaging was performed in 68% (67/101). Targetable oncogenic aberrations were found in 13% (13/101). The median time to BM was 10 months and median survival from BM diagnosis was 4 months. 37% (37/101) of patients had surgical resection, 20% (20/101) had stereotactic radiation (SRS) alone, 32% (32/101) had whole brain radiation therapy (WBRT) and 12% (12/101) had no local therapy. The surgery or SRS group had better survival outcomes compared to those who had WBRT or no local treatment following BM diagnosis (log rank χ2(2) = 7.590, p = 0.02) (Figure 1).

Conclusion: The incidence of BM at initial diagnosis is higher than published data with prognosis following BM diagnosis poor. This may be reflective of the regional demographic of patients who present more advanced. This underlines the importance of establishing local guidelines in intracranial disease detection and screening for earlier diagnosis and timely management to improve outcomes.

Niamh Walsh1,2, Rosalie Stephens3, Alvin Tan4, Vanessa Durandt5, Jennifer McLachlan6, Jody Jordan7, Kate Gregory8, Sean Sutton2, Catherine Barrow2, Annie Wong1,2 

1University of Otago, Wellington, New Zealand

2Wellington Blood & Cancer Centre, Wellington, New Zealand

3Regional Blood and Cancer Service, Auckland, New Zealand

4Medical Oncology, Waikato, New Zealand

5Oncology and Haematology, Dunedin, New Zealand

6Canterbury Regional Cancer and Haematology Service, Christchurch, New Zealand

7Midcentral District Cancer, Hawkes Bay, New Zealand

8Nelson Cancer Centre, Nelson, New Zealand 

Background: Most patients with metastatic melanoma will develop brain metastases (BM), with poor prognosis. Immune checkpoint inhibitors (ICI) are active for melanoma BM. However, landmark clinical trials are not representative of the “real-world” population, due to exclusion of patients with symptomatic BM, poor performance status or steroid use.1,2 This study reports the real-world outcomes of ICI for melanoma BM in New Zealand (NZ).

Methods: A retrospective audit of patients with melanoma BM treated with ICI between September 1, 2016, and September 1, 2020 was conducted across seven NZ cancer centers. Baseline demographics, tumor characteristics, treatment, intracranial tumor response rates, intracranial progression-free survival (iPFS), and overall survival (OS) were recorded.

Results: A total of 144 patients received at least one dose of ICI. Most patients (93%) received anti-PD1 monotherapy. Almost a quarter of patients had baseline performance status (ECOG 2–3), 56% were symptomatic and 33% had corticosteroids. Intracranial response was seen in 36%, and median-iPFS was 9 months. The 6- and 12-month-iPFS rates were 55% and 45%, respectively. Seventy-seven (53%) patients received local therapies, for example, surgery or radiation. The median OS was 15 months, and a third of patients were alive at 2 years (Figure 1). The toxicity of ICI was 28% and 15% for grade 1–2 and 3–4 events, respectively. Of those patients still alive at 2 years, 23% were asymptomatic.

Conclusions: Most patients had symptomatic disease and received anti-PD1 monotherapy. The intracranial tumor response rate for anti-PD1 monotherapy was higher than expected in this symptomatic population. However, this must be interpreted with caution given the retrospective study design. Nonetheless, this study provides real-world data for melanoma BM in NZ.

Acknowledgments: Cancer Society Central Districts and Graduate Women Otago Scholarship.

References:

1. Tawbi HA, Forsyth PA, Algazi A, et al. Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain. N Engl J Med. 2018;379(8):722–730. https://doi.org/10.1056/NEJMoa1805453

2. Long GV, Atkinson V, Lo S, Sandhu, et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol. 2018;19(5):672–681. https://doi.org/10.1016/S1470-2045(18)30139-6

Isabella Wilson1, Madeleine Strach1,2, Vivek Bhai1, Michelle Harrison1, Whiter Tang1, Peter Grimison

1Chris O'Brien Lifehouse, Sydney, Australia

2The University of Sydney Faculty of Medicine and Health, Sydney, Australia 

Background: Gemcitabine and docetaxel (GD) is a standard chemotherapy regimen for soft tissue sarcoma (STS). The GeDDiS phase 3 trial compared GD with doxorubicin in the 1st-line setting, using a 90-min gemcitabine infusion time, reporting a response rate of 20% and median progression-free survival (PFS) of 5.4 months.1 We examined the real-world efficacy and toxicity of GD in the 1st and later line using a 30-min gemcitabine infusion time.

Methods: A retrospective analysis was conducted of patients with unresectable or metastatic STS receiving ≥1 cycle of GD between July 2018 and October 2022. Data collected included demographics, tumor characteristics, dose intensity, toxicity, response, PFS, and overall survival (OS).

Results: Thirty-eight patients were included for analysis. Median follow-up was 19 months (range 3–30). Histologic subtypes (n) were leiomyosarcoma (20), synovial sarcoma (4), undifferentiated pleomorphic sarcoma (4), other (10). Line of treatment (n) was 1st line (10), 2nd line (13), and ≥3rd line (15). Median number of cycles was 6 (range 1–12). Sixteen patients (42%) required dose modification during the 1st three cycles. Response rate was 42%, including 5% with complete response. At the time of data collection, 33 patients had disease progression and 24 patients had died. PFS (median, 6-month rate) was 4.5 months and 44%. OS (median, 12-month rate) was 15 months and 65% (Figure 1). Grade 3/4 toxicity included neutropenia (8%), febrile neutropenia (3%), and anemia (21%). There was one death possibly attributable to GD.

Conclusion: This data demonstrates efficacy of GD comparable to other published data and favorable toxicity in a real-world population despite shorter gemcitabine infusion time and heavily pre-treated population.

Reference:

1. Seddon B, Strauss SJ, Whelan J, et al. Gemcitabine and docetaxel versus doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): a randomized controlled phase 3 trial. Lancet Oncol. 2017;18(10):1397-1410.

Vanessa Wong1,2,3, Michael Harold1, Bill Karanatsios2, Eleonora Kay4, Duncan Colyer4, Peter Gibbs1,2, Shehara Mendis1,2 

1Walter & Eliza Hall Institute of Medical Research, Parkville, Australia

2Western Health, St Albans, Australia

3Ballarat Health Services, Ballarat, Australia

4Victorian Comprehensive Cancer Centre (VCCC) Alliance, Parkville, Australia

Background: Cancer clinical trials are becoming progressively more complicated and expensive to run, making pragmatic trial designs that limit complexity and/or cost an attractive alternative. One such pragmatic trial design is to conduct a clinical trial using an existing prospective clinical registry to collect patient, treatment, and trial endpoint data. As such, investigator-initiated registry-based clinical trials (Reg-CTs) in cancer populations are emerging in Victoria, supported by the Victorian Comprehensive Cancer Centre (VCCC) Alliance. The perspectives of clinicians and study staff actively involved in this novel trial methodology is of interest.

Methods: A survey was constructed by a panel of clinicians and trials staff. The key focus was barriers and enablers of Reg-CTs. Invitations to participate in the survey were sent to clinical investigators, study coordinators, research nurses, research officers and clinical trials assistants at Victorian sites involved in Reg-CTs.

Results: A total of 42/100 (42%) potential respondents replied, including 18 (43%) investigators and 16 (38%) study coordinators. Thirty-eight (90%) were actively recruiting to Reg-CTs and 20 (48%) had been involved in Reg-CTs for over 2 years. The most important benefits perceived were involvement in trials asking relevant unanswered clinical questions and reduced study-related activity for site staff and patients. Funding was seen as the most important barrier to initiating Reg-CTs, and patient eligibility the largest barrier to enrollment. The single greatest challenge perceived in running Reg-CTs was having clear demarcation of staff responsibility for trial data entry versus registry data entry.

Conclusion: Clinical trial teams overall appreciate multiple benefits of involvement in Reg-CTs, including reduced burden of trial-related procedures. Identifying sufficient patient candidates remains a challenge in Reg-CTs. Though generally considered cost-effective in design and conduct, adequate funding for sites to run these trials was perceived as the single most important barrier to increasing uptake of Reg-CT trials.

Nicholas Yeo1, Sayeda Naher

1Sutherland Hospital Day Oncology Unit, Caringbah, Australia

2Illawarra Cancer Care Centre, Wollongong, Australia 

Background: Metastatic breast cancer represents a heterogenous group of patients. Whilst treatment and survival have improved, it remains the 5th most common cause of cancer death in Australia. There is a paucity of data describing the outcomes for patients with metastatic breast cancer treated in regional centers. We aim to describe the experiences within the Illawarra Shoalhaven Local Health District (ISLHD) and provide real-world data on the treatment patterns for these patients, including any factors influencing treatment response.

Methods: We retrospectively reviewed patients with metastatic breast cancer with available electronic medical records from 2011 to 2020, in the ISLHD. Standard demographic, clinicopathological characteristics, treatment data, and outcomes were assessed.

Results: We identified 169 patients with a mean age of 65 (interquartile range [IQR] 53–79). Most women had hormone-receptor positive breast cancers number (n = 104, 62%), followed by HER2-positive (n = 45, 26%), and triple negative breast cancers (TNBC) (n = 20, 12%). Amongst them, 125 (73.5%) of patients had de novo metastatic disease. The average number of lines of treatment received was 2 (IQR 1–3). The most prescribed chemotherapy regimens were taxanes (38.2%), followed by capecitabine (13.2%). Number of metastases (odds ratio [OR] 1.33, 95% CI 1.03–1.71, p = 0.03) and triple negative disease (OR 8.17, 95% CI 4.6–14.5, p < 0.001) were associated with poorer survival. There were no significant differences between de novo and recurrent disease (OR .80, 95% CI .53–1.22, p = 0.30).

Conclusion: This is the first retrospective review describing the treatment patterns and outcomes for patients with metastatic breast cancer within the Illawarra Shoalhaven Local Health District. Whilst treatment and survival have improved, it is evident that triple negative breast cancers remain an aggressive subgroup. There is a need for better treatments for this subgroup of patients.

Abstract Image

海报摘要
Dianheng Bu1, Javier torres1,2, Arvind Sahu1 1Goulburn Valley Health, Shepperton, Australia2墨尔本大学Shepperton医学院农村卫生部,Shepperton, Australia背景:长期坚持内分泌治疗是具有挑战性的,即使在临床试验环境中也是如此。不坚持治疗会增加癌症复发的风险和较差的生存率总体而言,关于内分泌治疗依从率的证据很少。该研究旨在衡量早期乳腺癌辅助内分泌治疗的依从率以及导致区域环境中不依从性的因素。方法:于2023年1 - 6月在Goulburn Valley Health进行横断面调查。女性患者接受辅助口服内分泌治疗。依从性采用经过验证的6项简化药物依从性问卷(SMAQ)进行测量。结果:51例患者参与,平均年龄60.1岁。由SMAQ确定的依从率为64.7%(33 / 51)。年龄和副作用数量显著影响依从性。依从性患者的平均年龄为64.3岁,非依从性患者的平均年龄为52.4岁(p &lt;0.001)(图1)。依从治疗患者的平均副作用数为1.8,非依从治疗患者的平均副作用数为3.8 (p &lt;0.01)。没有发现基于癌症分期、ECOG状态或内分泌治疗类型的依从性有显著差异。结论:该地区早期乳腺癌患者口服辅助内分泌治疗的依从率低于65%。年龄越小,副作用越多,依从率越低。这些发现强调了早期干预的必要性,以提高这一人群的依从性和优化治疗结果。李丽娟,李丽娟。内分泌治疗在乳腺癌预防中的临床应用。中华癌症杂志,2014;7(4):378-387.2。Ortega Suarez FJ, Sanchez Plumed J, Lorenzo AD。他克莫司肾移植患者简化服药依从性问卷(SMAQ)验证。Nefrologia。2011;31(6):690 - 696。Caitlin cock1, Sowmya Cheruvu1, Hilary L. martin11背景:在获得抗病毒药物和疫苗接种之前,在肿瘤患者队列中报道的COVID-19死亡率高达24%西澳大利亚州通过严格的疫苗接种规定和边境限制对COVID-19大流行进行了管理。本研究旨在评估2022年3月西澳边境开放后COVID-19感染对肿瘤患者的影响。方法:对2022年3月至5月期间在西澳某三级医院进行COVID-19检测呈阳性的肿瘤内科患者进行前瞻性筛查。回顾病例记录以收集数据,包括治疗延误、癌症结局以及感染后30天、60天和12个月的死亡率。结果:新冠肺炎阳性37例,症状率64.9%。78.4%的患者在感染前至少接受过两次COVID-19免疫接种。乳腺癌是最常见的恶性肿瘤(40.5%),其次是非小细胞肺癌(21.6%)。29名患者接受积极治疗,9名接受化疗,3名接受免疫治疗。20例患者接受抗病毒治疗。8名患者住院治疗。治疗延迟影响了76.9%的幸存者,中位延迟为2周。10例患者在随后的12个月内出现疾病进展。5名患者报告了第二次COVID-19感染,并无并发症康复。共有13例患者在随访期间死亡:8例在30天内死亡,3例在30天至60天死亡,2例在60天至12个月死亡。无与COVID-19相关的直接死亡报告。结论:令人欣慰的是,没有与COVID-19直接相关的死亡。这些发现支持在这一癌症患者群体中使用抗病毒药物和疫苗接种,并与COVID-19早期的高死亡率形成对比。参考文献:Russell B, Moss CL, Shah V等。癌症患者COVID-19死亡风险:来自伦敦盖伊癌症中心和国王学院医院的分析中国生物医学工程杂志,2013;31(7):349 - 349。https://doi.org/10.1038/s41416-021-01500-z.Andrew Fantoni1, Brandon Lau2, Jess McNeill3, Christine Henneker3, Shaouli Shahid4, Wei-Sen Lam1 1澳大利亚莫道克Fiona Stanley医院肿瘤内科2澳大利亚内德兰兹Charles Gairdner爵士医院肿瘤内科3澳大利亚东珀斯乡村卫生服务中心4澳大利亚本特利科廷科技大学土著研究中心远程医疗可以提供更多的肿瘤专科护理,同时保持病人离家近。 方法:使用患者电子病历(eMR)对2015 - 2022年SMHS内所有接受FOLFIRINOX或GnP(纳米颗粒白蛋白结合紫杉醇)作为NAT治疗BR或LA胰腺癌的患者进行识别。结果:19例接受NAT治疗的患者中,15例接受FOLFIRINOX治疗,4例接受GnP治疗。在接受FOLFIRINIX治疗的患者中,40.0% (n = 6)成功行胰腺切除术,53.3% (n = 8)不符合手术条件,6.7% (n = 1)失去随访。在接受GnP的患者中,只有一名患者接受了胰腺切除术。排除NAT后胰切除术的原因包括NAT反应不足/疾病进展(63.6%,n = 7)和表现不佳(36.4%,n = 11)。胰腺切除术标本从IA-IIB分期,多数为2级(85.7%;n = 6)和切除阴性边缘(R0) (57.1%, n = 4)。接受胰腺切除术的患者的中位生存期(30个月对13个月)和中位无进展生存期(20个月对7个月)分别高于未接受胰腺切除术的患者。结论:五分之二的FOLFIRINOX NAT患者和四分之一的GnP NAT患者进行了胰腺切除术。这些比率反映了与已发表的试验数据(如CONKO-007.7)相比,现实世界中不太可能适合手术的患者。Ek Leone Oh, Elizabeth Steinepreis, Hilary Martin Fiona Stanley医院,Murdoch,澳大利亚背景:小叶乳腺癌约占乳腺癌的10%,其治疗通常是从主要由导管性乳腺癌患者组成的试验中推断出来的。本研究旨在探讨转移性小叶性乳腺癌患者的治疗效果。方法:我们对2017年1月至2023年4月31日在单一中心诊断为转移性小叶性乳腺癌的患者进行回顾性研究。治疗的临床获益定义为疾病稳定、部分缓解或完全缓解。结果:21例转移性小叶性乳腺癌。16例(76.2%)为激素受体阳性HER2阴性乳腺癌(HR+ HER2−),2例(9.5%)为三阴性乳腺癌,3例(14.3%)为HER2阳性激素受体阳性(HER2+HR+)。HR+HER2−亚组的治疗和结果见表1。在HR+HER2−亚组患者中,14例(87.5%)接受了CDK4/6抑制剂和非甾体芳香化酶抑制剂(NSAI)的联合治疗,其中13例作为转移性疾病的一线治疗开始,6例(42.9%)在数据截止时仍在使用该治疗。7例(43.8%)HR+HER−患者在治疗的某个阶段接受了化疗。两名患有三阴性小叶乳腺癌的患者在所有试验药物(奥拉帕尼、紫杉醇、卡铂/吉西他滨和伊瑞布林)的治疗下都经历了快速进展的疾病。3例HER2+HR+疾病患者接受紫杉烷或长春瑞滨、帕妥珠单抗和曲妥珠单抗联合一线治疗。在数据截止时,所有患者仍在接受这种治疗,中位持续时间为34个月(范围19-68个月)。结论:与支持使用CDK4/6抑制剂和NSAI联合治疗小叶癌的注册试验数据相比,HR+ HER2−小叶乳腺癌的PFS相似。化疗也有一些反应,所有接受卡培他滨治疗的患者都获得了临床获益。her2阳性患者预后良好,而三阴性小叶乳腺癌患者预后较差。两名患有三阴性小叶乳腺癌的患者在所有药物(奥拉帕尼、紫杉醇、卡铂/吉西他滨和伊瑞布林)的试验中都经历了快速进展的疾病。3例HER2+HR+疾病患者接受紫杉烷或长春瑞滨、帕妥珠单抗和曲妥珠单抗联合一线治疗。在数据截止时,所有患者仍在接受这种治疗,中位持续时间为34个月(范围19-68个月)。Hayley Roberts1,2, Benjamin Solomon2, Susan Harden2, Senthil Lingaratnam2, Marliese Alexander2 1Alfred Health, Melbourne, Australia2 2 peter MacCallum Cancer Centre, Melbourne, Australia背景:尽管最近在免疫治疗、酪氨酸激酶抑制剂(TKIs)和放疗方面取得了进展,但肺癌仍然是澳大利亚癌症死亡的主要原因。全身抗癌治疗后30天死亡率(SACT)已被建议作为肺癌的质量指标,并在英国作为基准引入。然而,尽管多个国际研究人员报告了30天死亡率,但计算这些死亡率的确切方法在不同的研究中差别很大。 4例患者不符合筛查条件;48例开始试验性治疗。从转诊到开始试验的中位时间为14天。入选患者最常见的诊断是妇科癌症(31%),其次是上胃肠道癌症(21%)和结直肠癌(20%)。结论:2022年,SCR一期肿瘤学试验的转诊患者和试验参与者代表了不同的地理人群。来自不同文化和语言背景的患者人数不足。Lucy Porter1, Phillip parente2,3, Grace gard4,5, Benjamin Brady6, Wasek faisal7,8, Dishan Herath4, Margaret le2,3, Peter gibb4,5, Ben markman5,6, Rachel wong 2,3 1澳大利亚墨尔本莫纳什大学医学院、护理与健康科学学院2澳大利亚墨尔本东部健康医学肿瘤科3澳大利亚墨尔本莫纳什大学东部健康临床医学院4墨尔本西部健康医学肿瘤科澳大利亚,5沃尔特和伊莱扎·霍尔医学研究所,墨尔本,澳大利亚;6卡布里尼健康中心,墨尔本,澳大利亚;7格兰屏综合癌症服务中心,格兰屏健康中心,巴拉瑞特,澳大利亚;8拉特伯大学健康学院,墨尔本,澳大利亚背景:早期非小细胞肺癌(NSCLC)的治疗正在发展,越来越多的数据支持免疫治疗和靶向药物在辅助和新辅助环境中的作用。Atezolizumab是一种PD-L1抑制剂,自2022年11月以来一直获得pbs补贴,用于切除的PD-L1≥50的II-IIIa期非小细胞肺癌患者,这些患者是EGFR/ALK野生型,接受铂基化疗。在澳大利亚医院接受分子检测并有资格接受pbs补贴的atezolizumab治疗的患者比例尚不清楚。方法:回顾性分析2020年2月至2023年1月在澳大利亚四个地点(n = 448)登记的多地点吸气性肺癌登记的患者。接受手术切除的NSCLC患者(n = 115)被纳入研究,并对分期、分子检测、治疗细节和结果进行分析。结果:115例非小细胞肺癌患者中有90例获得了治疗目的。63/90接受单纯手术治疗,25/90接受辅助全身治疗和/或放疗,2/90接受新辅助全身治疗和/或放疗。7/90出现疾病复发/进展。共有70/90名治疗意向切除患者进行了PD-L1检测。30天内62/70,90天内67/70。56/90进行了基因组检测。测试结果如表1所示。在70名PD-L1测试患者中,根据目前的PBS标准,1名患者符合辅助atezolizumab的条件。67/70不合格(PD-L1 &lt;50%(不合格期,无铂类化疗)和2/70的患者不知道是否适合pbs(未进行基因组检测)。PD-L1和基因组检测率在大都市比在区域环境更高(分别为70% - 90%比40%和15%-88%比50%),在私人环境比在公共环境更高(分别为90%比70%和88%比15%-65%)。结论:在对切除的非小细胞肺癌患者的实际分析中,很少有常规护理患者符合pbs补贴的atezolizumab的条件。我们预计,随着辅助atezolizumab在PBS上的可用性,以及支持新辅助/辅助使用免疫治疗和/或靶向药物的证据不断出现,PD-L1和基因组检测率将会增加。Joanne Tang1, Brian Stein2, 31 1澳大利亚阿德莱德中央地方卫生网络2澳大利亚阿德莱德icon癌症中心3澳大利亚阿德莱德大学背景:EviQ是一个共识驱动的基于证据的癌症治疗方案存储库,在澳大利亚广泛使用,并作为事实上的标准。尚未对在EviQ上发表的方案的临床益处进行标准化评估。基于EviQ中的文献,我们旨在确定对所有实体瘤方案进行评分的可行性,以评估方案的益处,并探讨ESMO-MCBS评分与治疗指征、肿瘤流和试验终点之间的相关性。方法:我们使用ESMO-MBCS 1.1版本对截至2022年4月31日在EviQ上发表的所有实体瘤方案进行分级,排除功能重复(例如,基于体重的免疫治疗与平剂量免疫治疗)。我们仅使用“证据”部分作为参考,因为这是协议发布的证据理由。除了技术上不可评估的协议之外,我们还认为在常见场景中使用阶段II数据的协议,缺乏标准比较器,或者基于集中的数据是不可评估的。结果:纳入的478个方案中,301个(63%)可评价。97例为治疗目的,204例为缓解目的。 总体而言,140个(47%)评估方案达到了显著获益的MCBS阈值;75%的治疗性方案,但只有33%的姑息性方案具有“实质性益处”。在57个具有毒性或生活质量益处的方案中,88%是姑息治疗方案,47%是化疗方案。在低效益方案中,49%是基于化疗的,而基于反应的结果在姑息治疗方案中被过度代表。肿瘤分类评分无差异。在177个不可评估的方案中,31%缺乏标准比较物,42%使用非孤儿环境的II期数据。结论:在EviQ中,47%的可评估方案为ESMO-MCBS“实质性获益”;大多数是可以治愈的。很少有方案因毒性较低而得分。我们建议在EviQ中加入ESMO-MCBS评分,以辅助方案的考虑。Sophie Tran1,2, Cassana Moore2, Danielle rosco2, Hieu Chau2, Sachin Joshi2,3, Quan Tran2, Rohan Nair2,3, Mahesh iddawel2,3,4 1澳大利亚Box Hill东部卫生部2澳大利亚Traralgon拉特罗布地区卫生部3澳大利亚墨尔本alfred Health 4澳大利亚Traralgon莫纳什大学农村卫生部背景:脑转移(BM)在非小细胞肺癌(NSCLC)中常见,具有高发病率和死亡率。新的全身治疗和放射技术的出现改善了这一预后不良群体的预后。因此,诊断和监测的早期发现是重要的。本项目旨在回顾单个区域中心肺癌BM的特点、当前实践、管理和结果,以帮助制定当地方案。方法:从威廉巴克兰放射治疗中心病历和拉特罗布地区健康肺癌数据库中确定肺癌BM患者。2017年至2022年间,共有101名患者被确诊。收集了人口统计学和临床信息数据。结果:肺癌诊断的中位年龄为68岁,59%为男性,71%为ECOG 0或1。在最初诊断时,66%(67/101)的患者有转移性疾病,其中60%(40/67)的患者有脑转移。68%(67/101)的患者进行了初始脑成像。13%(13/101)发现了可靶向的致癌畸变。到脑转移的中位时间为10个月,诊断为脑转移后的中位生存期为4个月。37%(37/101)的患者行手术切除,20%(20/101)的患者单独行立体定向放疗(SRS), 32%(32/101)的患者行全脑放疗(WBRT), 12%(12/101)的患者未行局部治疗。手术组或SRS组的生存结果优于行WBRT或未行局部治疗的BM组(log rank χ2(2) = 7.590, p = 0.02)(图1)。结论:初诊时BM的发病率高于已发表的数据,BM诊断后预后较差。这可能反映了呈现更晚期的患者的区域人口统计。这强调了建立颅内疾病检测和筛查指南的重要性,以便早期诊断和及时管理以改善结果。Niamh walsh 1,2, Rosalie Stephens3, Alvin Tan4, Vanessa durand5, Jennifer McLachlan6, Jody Jordan7, Kate Gregory8, Sean Sutton2, Catherine Barrow2, Annie wong 1,2新西兰惠灵顿奥塔哥大学癌症中心,惠灵顿,新西兰3区域血液和癌症服务中心,奥克兰,新西兰4医学肿瘤学,怀卡托,新西兰5肿瘤和血液学,达尼丁,新西兰6坎特伯雷区域癌症和血液学服务中心,新西兰克赖斯特彻奇,新西兰7中中区癌症,新西兰霍克斯湾,新西兰8纳尔逊癌症中心,新西兰尼尔森背景:大多数转移性黑色素瘤患者会发展为脑转移(BM),预后差。免疫检查点抑制剂(ICI)对黑色素瘤脑转移有活性。然而,具有里程碑意义的临床试验并不代表“现实世界”的人群,因为排除了有症状性脑脊髓炎、表现不佳或使用类固醇的患者。1,2本研究报告了在新西兰(NZ) ICI治疗黑素瘤的实际结果。方法:对2016年9月1日至2020年9月1日期间在新西兰7个癌症中心接受ICI治疗的黑色素瘤BM患者进行回顾性审计。记录基线人口统计学、肿瘤特征、治疗、颅内肿瘤反应率、颅内无进展生存期(iPFS)和总生存期(OS)。结果:共有144例患者接受了至少一剂ICI。大多数患者(93%)接受抗pd1单药治疗。几乎四分之一的患者有基线表现状态(ECOG 2-3), 56%有症状,33%有皮质类固醇。颅内缓解率为36%,中位ipfs为9个月。6个月和12个月的ipfs分别为55%和45%。77名(53%)患者接受了局部治疗,例如手术或放疗。 西澳大利亚国家卫生服务(WACHS)远程血液治疗模式于2019年9月在皮尔巴拉(Karratha)实施,并于2020年3月在金伯利(Broome)和Wheatbelt (nargin)实施。我们检查了该模型在治疗利用和毒性结果方面的安全性。方法:收集2019年9月至2022年2月期间所有就诊于WACHS远程血液治疗服务的患者的回顾性数据;包括患者人口统计、癌症诊断、治疗意图、接受的治疗周期和治疗线、治疗延误和毒性(CTCAEv5.0的不良事件[ae]、死亡和住院情况)。描述性统计用于报告安全性结果。结果:159例患者转介治疗,154例(97%)接受治疗。21例因病情恶化或倾向于大都市治疗而退出。最终队列(n = 133)的中位年龄为61岁(范围24-90岁),其中54%为男性,23%为原住民,94%为ECOG≤1。总共提供了1034个治疗周期,代表171个治疗线。74%的患者为姑息治疗。平均剂量强度维持在90%(±14%)。47条治疗线(27%)接受了剂量减少,62%在远程血液治疗之前。58个品系(34%)由于毒性(53%)或患者偏好(38%)而存在显著的剂量延迟。63条线(37%)与≥3级ae相关。ae的发生率和治疗方案之间没有明显的关联。32% (n = 54)的治疗线发生≥1次住院。11人死亡,其中一人死于中毒。48个含免疫治疗的品系中,4个品系(8%)的免疫相关不良事件≥3级,无死亡病例。结论:本研究证实了WACHS远程血液治疗模型的安全性,包括低免疫介导的毒性。剂量强度没有受到影响,治疗延误是由于预期的毒性或患者的偏好,而不是后勤问题。Jocelyn Finney1, Isabella Wilson1, Ben felmingham 2,3,4, Sanjeev Kumar1, Alina Mahmood1, Lisa horvath1,5, David Celermajer6, Rachel Conyers7 1澳大利亚悉尼,chris O'Brien生命之家2澳大利亚墨尔本,默多克儿童研究所3澳大利亚墨尔本,皇家儿童医院儿童癌症中心4澳大利亚墨尔本,默多克儿童研究所心脏再生实验室5悉尼大学,悉尼,悉尼背景:癌症治疗相关心功能障碍(CTRCD)是多种化疗、靶向药物和免疫疗法的重要副作用,是治疗的一部分。蒽环类药物是一种常用的化疗药物,可引起不可逆的早发性和晚发性心力衰竭,目前文献记载不足。在澳大利亚,目前还没有其他针对成人CTRCD的多中心纵向研究。COBLH旨在利用澳大利亚心脏肿瘤学登记处(ACOR)来探讨接受心脏毒性治疗的成人中CTRCD的流行病学、临床表现和管理。此外,一项生物银行子研究将试图确定基因多态性,使个体易患蒽环类药物心脏毒性(ACT)。方法:ACOR是一项前瞻性观察性研究,涉及澳大利亚14个地点。COBLH患者标准包括过去5年内接受心脏毒性治疗的非姑息性高风险成人患者。信息从医疗记录中获取,并在进入ACOR时去识别。ACOR收集患者人口统计、病史、癌症诊断和治疗、心脏评估和CTRCD管理方面的数据。这些仪器在治疗前基线收集,之后每隔6个月收集一次。患有ACT的患者将被要求参加生物银行研究,该研究将涉及一次性抽取12ml外周血,在外部部位进行全基因组外子组测序和多能干细胞衍生心肌细胞的培养。结果:在提交时,COBLH招募的220例患者中有19例发生了CTRCD。这包括10例ACT患者,3例左心室功能障碍患者,3例疑似蒽环类药物相关心肌病患者,3例症状性心力衰竭患者。ACOR预计全部注册站点的容量为每年捕获600名患者。结论:COBLH将使用ACOR研究成人患者的长期CTRCD,生物库研究旨在证明遗传易感性与ACT之间的因果关系。鸣谢:2、3。 背景:间变性甲状腺癌(ATCS)是一种罕见的侵袭性肿瘤,尽管采用多种治疗方法,但预后极差。最近的数据显示,lenvatinib(一种酪氨酸激酶抑制剂)和pembrolizumab(一种通过抑制PD-1起作用的检查点抑制剂)联合使用可以提高atc患者的应答率和生存率。方法和结果:该病例描述了一名67岁女性,于2021年9月首次诊断,表现为颈部肿块增大。甲状腺切除术及双侧颈部清扫示42 mm间变性甲状腺癌,血管浸润,甲状腺带肌外展,0/25淋巴结受累,边缘清晰。该患者于2021年12月完成了低剂量阿霉素辅助放化疗。2022年8月局部复发,认为不适合切除。她于2022年9月9日开始使用派姆单抗和lenvatinib (20mg), 4周后症状有所改善。治疗8周后的PET扫描显示几乎完全缓解,治疗6个月后的PET继续显示没有令人信服的复发证据(图1)。结论:在预后极差的疾病背景下,该病例表明lenvatinib和pembrolizumab联合治疗ATCs是一个改变游戏规则的方法。[15] lenvatinib/pembrolizumab联合治疗转移性间变性或低分化甲状腺癌患者可诱导长期完全缓解:一项回顾性研究结果和前瞻性II期ATLEP试验的初步结果。中国生物医学工程学报,2020;31(1):1085.2。刘建军,李建军,李建军,等。lenvatinib联合派姆单抗抢救治疗间变性甲状腺癌少细胞变异1例中国生物医学工程学报,2011;28(6):5401-5407。Grace Gard, Joanna Oakley, Vanessa Wong, Kelsey Serena, Michael Harold, Jo Cockwill, Katya Gray, Helen Anderson, Graeme Down, Judi Price, Peter Gibbs WEHI, Australia Parkville背景/目标:在癌症研究中,消费者和研究人员合作合作的兴趣越来越大,但合作的可行性以及如何优化共同设计过程尚未得到很好的确立。一个由卫生保健专业人员和消费者组成的研究小组旨在为新诊断的局部晚期直肠癌患者制定个性化护理计划。该小组反思并记录了他们的共同设计经验。方法:我们的研究组包括来自WEHI消费者项目的消费者、多学科结直肠癌临床医生和项目官员。他们一起创建了个性化护理计划模板,提供给病人和他们的全科医生。团队对共同设计过程的反思已经被Gibbs的反思周期所捕捉。患者目前正在纳入一个前瞻性队列,以评估个性化护理计划的影响。结果:在2022年,经过六次会议,我们创建了一个两页的个性化护理计划,并将其嵌入到WEHI-CRC数据库中。个别患者的个性化信息包括疾病分期、计划治疗和预定随访。对共同设计过程的反思强调了建立期望、在消费者群体中拥有专业知识、开放沟通和尊重的重要性。挑战包括时间承诺、权力动态、代表的多样性以及由于健康原因和时间可用性而失去消费者。调查结果反映出研究人员对消费者投入价值的态度发生了积极的变化。消费者和研究人员都传达了为项目做出贡献的高度客观和情感价值。结论:反思患者教育单的共同设计过程,我们发现共同设计小组的基本组成部分有明确的期望,以及应对时间承诺和根深蒂固的权力动态等挑战的策略。来自消费者的有意义的投入可以客观上改善项目的结果,同时为消费者和研究人员提供积极的个人价值。背景:在从静脉注射到口服治疗的过渡中,抗生素的选择和时机越来越受到关注和变化。我们的目的是了解在我们的机构中性粒细胞减少脓毒症的治疗和处方做法。方法:在墨尔本的一家三级医院进行回顾性研究。2020年1月至2021年12月期间符合中性粒细胞减少性败血症标准的实体器官恶性肿瘤成年患者符合条件。 背景:辅助派姆单抗单药治疗可改善高风险切除黑色素瘤患者的无复发生存期(RFS)和远处转移生存期(DMFS)。Vibostolimab是一种抗tigit抗体,在1期KEYVIBE-001研究中,Vibostolimab与pembrolizumab联合用于实体瘤的抗肿瘤活性和可管理的安全性。在双盲、随机、3期KEYVIBE-010研究(NCT05665595)中,佐剂共配制vibostolimab与pembrolizumab在切除高风险黑色素瘤患者中的疗效和安全性。方法:患者年龄≥12岁,手术切除的IIB或IIC期(病理或临床),III或IV期皮肤黑色素瘤,ECOG PS为0或1(18岁),Karnofsky表现状态≥70(≥16 - 18岁),或Lansky游戏表现量表≥70(16岁),无转移性疾病的证据将被纳入研究。患有眼部、粘膜或结膜黑色素瘤的患者将被排除在外。切除必须在随机分组前12周内进行。患者将需要提供档案肿瘤组织样本或新获得的肿瘤病变活检以前没有照射。患者将根据风险(IIB/IIC/临床IIB期和IIC/IIIA/IIIB期vs IIIC/IIID/IV期)和地区(亚洲vs世界其他地区)进行分层。大约1560名患者将按1:1的比例随机分配到静脉共配制vibostolimab 200 mg与pembrolizumab 200 mg或pembrolizumab 200 mg(青少年2 mg/kg至200 mg),每3周最多17个周期。主要终点是研究者评审的RFS。次要终点是研究者评价的DMFS、总生存期、安全性和生活质量。风险比和95% ci将使用分层Cox回归模型进行估计,采用Efron方法处理联系,治疗作为协变量。采用分层对数秩检验评估治疗间差异。报名正在进行中。Chee le1, Sandy Simon1, Danka Zebic1, Yeh Chen le1, Katrina Diamante1, Lenna Lai1, Rachel O'Connell1, Anna deFazio2, Sonia Yip1, Lyndal Anderson3, Bo Gao4, Jeffrey C. Goh5, Kate Webber6, Sally Baron-Hay7, Christopher Steer8, Linda Mileshkin9, Tarek meniaw10, Janine Lombard11, Martin Stockler1, Michael Friedlander12 1NHMRC临床试验中心,悉尼大学,坎普尔当,悉尼大学,韦斯特米德医学研究所和韦斯特米德医院,澳大利亚韦斯特米德3澳大利亚坎珀当皇家阿尔弗雷德王子医院4澳大利亚韦斯特米德医院5澳大利亚布里斯班皇家布里斯班和妇女医院6澳大利亚克莱顿莫纳什医疗中心7澳大利亚圣伦纳兹皇家北岸医院8澳大利亚奥尔伯里边境医学肿瘤研究中心9澳大利亚墨尔本彼得麦卡勒姆癌症中心10澳大利亚内德兰兹查尔斯盖德纳爵士医院11纽卡斯尔私立医院新兰姆顿高地澳大利亚12威尔士亲王医院和皇家妇女医院,兰德威克,澳大利亚芳香酶抑制剂(AIs)用于治疗激素受体阳性(HR+)晚期妇科癌症。然而,反应率明显低于用AIs治疗的HR+乳腺癌。CDK4/6通路的改变和磷脂酰肌醇-4,5-二磷酸3-激酶催化亚基α (PIK3CA)的突变被认为是导致AI耐药的原因。在HR+转移性乳腺癌中,AI联合CDK4/6抑制剂(ribociclib)或PIK3CA抑制剂(alpelisib)可改善临床结果。PARAGON-II试验(ACTRN12621000639820)旨在评估来曲唑加alpelisib或ribociclib在PIK3CA突变型(mut)和PIK3CA野生型(wt) HR+晚期妇科癌症中的活性。这是一项II期、开放标签、一揽子试验,包括6组常见和罕见的HR+妇科癌症。目标人群为复发/转移性癌症患者,表现良好,不需要化疗。PIK3CA突变状态是通过组织或ctDNA分析确定的。该研究包括PIK3CA mut子宫内膜癌(队列A)和任何其他PIK3CA mut肿瘤(F)。PIK3CA wt/未知队列包括子宫内膜癌(B)、铂耐药高级别上皮性卵巢癌(C)、子宫内膜间质肉瘤、平滑肌肉瘤、颗粒细胞肿瘤(D)和低级别浆液性卵巢癌(E)。在PIK3CA mut队列(A和F)中,参与者将每天接受来曲唑2.5 mg加阿派西布300 mg,直到疾病进展或不耐受。在PIK3CA wt/未知队列(B-E)中,参与者将接受来曲唑2.5 mg /天加ribociclib 600 mg /天(3周开/1周停)。总共将招募182名参与者。主要终点为客观有效率。 次要终点包括疾病控制率、缓解持续时间、6个月无进展生存期(PFS)、中位PFS、患者报告的结局和不良事件。正在收集肿瘤和血液样本用于转译研究。PARAGON-II试验于2022年5月开始招募。截至2023年6月30日,已从13个站点招募了64名参与者。招聘预计将于2024年12月完成。人工智能用于治疗激素受体阳性(HR+)晚期妇科癌症。然而,反应率明显低于用AIs治疗的HR+乳腺癌。CDK4/6通路的改变和磷脂酰肌醇-4,5-二磷酸3-激酶催化亚基α (PIK3CA)的突变被认为是导致AI耐药的原因。在HR+转移性乳腺癌中,AI联合CDK4/6抑制剂(ribociclib)或PIK3CA抑制剂(alpelisib)可改善临床结果。Su Saint le1,2,3, Wei Chua1,4,5, Nancy huang 1,2,3, ng Weng 1,4,5, Aflah roohullah 1,2,2, Annette Tognela2, Ray Asghari3, Mahtab Farzin6, Tara roberts4,5,7, Therese becker4,5,7, Bin Wang6, Soon lee5,6,7, Stephanie lim3,4,51澳大利亚利物浦利物浦医院肿瘤内科2澳大利亚坎贝尔镇坎贝尔镇医院麦克阿瑟癌症治疗中心肿瘤内科3西悉尼大学医学院坎贝尔镇澳大利亚4班克斯敦-利德库姆医院肿瘤科5利物浦医院解剖病理学科6利物浦英厄姆应用医学研究所7利物浦新南威尔士大学西南临床学院背景:肿瘤突变谱影响转移性结直肠癌(mCRC)的预后,但目前尚不清楚为什么一些肿瘤表现异常好或异常差。我们的目标是使用靶向下一代测序(NGS)识别与特殊病例相关的基因。方法:从2010年至2018年来自西南悉尼解剖病理组织库的RAS/ braf野生型和mmr熟练的mCRC患者中筛选出总生存期为6个月或3年或治疗反应不典型的患者。获得临床病理变量和生存结果。随机抽取8例患者进行30基因(Qiagen)、50基因或161基因(ThermoFisher) NGS面板测试,以建立工作流程、可行性并获得初步数据。根据ESCAT显著性等级对鉴定出的突变进行分类。所有病例都将进行161基因面板NGS。结果:鉴定出52例优秀者(GP)和14例优秀者(PP)。GP患者的中位年龄为65岁,PP患者的中位年龄为51岁。GP患者中位肿瘤为左侧,PP患者中位肿瘤为77%。从诊断转移性疾病开始,GP患者的中位总生存期为56.4个月(20.6-127.1个月),PP患者中位总生存期为4.8个月(1.4-15.4个月)。迄今为止,我们有6例GP和2例PP患者的NGS数据(4例30基因,2例50基因,2例161基因)。两名GP患者有TP53突变,另一名患者有PIK3CA突变(均为II级),而第四名患者有EGFR突变,其意义未知。2GP和2例PP患者未检测到突变。基因小组检测正在对迄今为止检测有限的患者进行。结论:靶向NGS面板可以识别RAS/ braf野生型mCRC中表现异常的基因,其总生存率差异超过10倍。初步数据显示GP中TP53和PIK3CA存在差异。所有病例的161个基因的最终NGS结果正在进行中。Yeh Chen le1, Elizabeth Barnes1, Katherine Francis1, Danka Zebic1,2, Sandy Simon1, Gemma blun1, peararly Khaw3, Viet Do4, Elizabeth christi3, Rachel Delahunty3, Rebecca Mercieca-Bebber1, Sonia Yip1, Jeffrey Goh5, Janine Lombard6, Kate Webber7, Sudarshan Selva-Nayagam8, Ganessan Kichenadasse9, Tarek Meniawy10, Michelle Harrison11, Michelle Wilson12, Martin Stockler1, Linda Mileshkin3悉尼大学nhmrc临床试验中心,Camperdown,澳大利亚2澳大利亚兰德威克威尔士亲王医院和皇家妇女医院3澳大利亚墨尔本彼得麦卡勒姆癌症中心4澳大利亚利物浦利物浦癌症治疗中心5澳大利亚布里斯班皇家布里斯班妇女医院6澳大利亚纽卡斯尔纽卡斯尔7澳大利亚克莱顿莫纳什医疗中心8澳大利亚阿德莱德皇家阿德莱德医院9澳大利亚贝德福德公园弗林德斯医疗中心10内德兰查尔斯加德纳爵士医院背景:尽管进行了初次手术、辅助放化疗和化疗,高危子宫内膜癌(EC)患者仍有显著的复发风险,此后的中位生存时间约为12个月。免疫检查点抑制剂在晚期EC中显示出活性。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
3.40
自引率
0.00%
发文量
175
审稿时长
6-12 weeks
期刊介绍: Asia–Pacific Journal of Clinical Oncology is a multidisciplinary journal of oncology that aims to be a forum for facilitating collaboration and exchanging information on what is happening in different countries of the Asia–Pacific region in relation to cancer treatment and care. The Journal is ideally positioned to receive publications that deal with diversity in cancer behavior, management and outcome related to ethnic, cultural, economic and other differences between populations. In addition to original articles, the Journal publishes reviews, editorials, letters to the Editor and short communications. Case reports are generally not considered for publication, only exceptional papers in which Editors find extraordinary oncological value may be considered for review. The Journal encourages clinical studies, particularly prospectively designed clinical trials.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信