154 results on '"Palmer DH"'
Search Results
2. hENT1 Predicts Benefit from Gemcitabine in Pancreatic Cancer but Only with Low CDA mRNA
- Author
-
Aughton, K, Elander, NO, Evans, A, Jackson, R, Campbell, F, Costello, E, Halloran, CM, Mackey, JR, Scarfe, AG, Valle, JW, Carter, R, Cunningham, D, Tebbutt, NC, Goldstein, D, Shannon, J, Glimelius, B, Hackert, T, Charnley, RM, Anthoney, A, Lerch, MM, Mayerle, J, Palmer, DH, Buechler, MW, Ghaneh, P, Neoptolemos, JP, Greenhalf, W, Aughton, K, Elander, NO, Evans, A, Jackson, R, Campbell, F, Costello, E, Halloran, CM, Mackey, JR, Scarfe, AG, Valle, JW, Carter, R, Cunningham, D, Tebbutt, NC, Goldstein, D, Shannon, J, Glimelius, B, Hackert, T, Charnley, RM, Anthoney, A, Lerch, MM, Mayerle, J, Palmer, DH, Buechler, MW, Ghaneh, P, Neoptolemos, JP, and Greenhalf, W
- Abstract
Gemcitabine or 5-fluorouracil (5-FU) based treatments can be selected for pancreatic cancer. Equilibrative nucleoside transporter 1 (hENT1) predicts adjuvant gemcitabine treatment benefit over 5-FU. Cytidine deaminase (CDA), inside or outside of the cancer cell, will deaminate gemcitabine, altering transporter affinity. ESPAC-3(v2) was a pancreatic cancer trial comparing adjuvant gemcitabine and 5-FU. Tissue microarray sections underwent in situ hybridization and immunohistochemistry. Analysis of both CDA and hENT1 was possible with 277 patients. The transcript did not correlate with protein levels for either marker. High hENT1 protein was prognostic with gemcitabine; median overall survival was 26.0 v 16.8 months (p = 0.006). Low CDA transcript was prognostic regardless of arm; 24.8 v 21.2 months with gemcitabine (p = 0.02) and 26.4 v 14.6 months with 5-FU (p = 0.02). Patients with low hENT1 protein did better with 5-FU, but only if the CDA transcript was low (median survival of 5-FU v gemcitabine; 29.3 v 18.3 months, compared with 14.2 v 14.6 with high CDA). CDA mRNA is an independent prognostic biomarker. When added to hENT1 protein status, it may also provide treatment-specific predictive information and, within the frame of a personalized treatment strategy, guide to either gemcitabine or 5FU for the individual patient.
- Published
- 2021
3. The Nrf2 inhibitor brusatol is a potent antitumour agent in an orthotopic mouse model of colorectal cancer
- Author
-
Evans, JP, Winiarski, BK, Sutton, PA, Jones, RP, Ressel, L, Duckworth, CA, Pritchard, DM, Lin, ZX, Vicky, FL, Tweedle, EM, Costello, E, Goldring, CE, Copple, IM, Park, BK, Palmer, DH, and Kitteringham, NR
- Subjects
respiratory system ,neoplasms ,digestive system ,environment and public health ,digestive system diseases - Abstract
© Evans et al. Nrf2 is a transcription factor that regulates cellular stress response and irinotecan-metabolising pathways. Its aberrant activity has been reported in a number of cancers, although relatively few studies have explored a role for Nrf2 in colorectal cancer (CRC). This study assessed the expression of Nrf2 in patient CRC tissues and explored the effect of Nrf2 modulation alone, or in combination with irinotecan, in human (HCT116) and murine (CT26) cell lines in vitro and in an orthotopic syngeneic mouse model utilising bioluminescent imaging. Using a tissue microarray, Nrf2 was found to be overexpressed (p < 0.01) in primary CRC and metastatic tissue relative to normal colon, with a positive correlation between Nrf2 expression in matched primary and metastatic samples. In vitro experiments in CRC cell lines revealed that Nrf2 siRNA and brusatol, which is known to inhibit Nrf2, decreased viability and sensitised cells to irinotecan toxicity. Furthermore, brusatol effectively abrogated CRC tumour growth in subcutaneously and orthotopicallyallografted mice, resulting in an average 8-fold reduction in luminescence at the study end-point (p=0.02). Our results highlight Nrf2 as a promising drug target in the treatment of CRC.
- Published
- 2018
4. Intratumoural expression of deoxycytidylate deaminase or ribonuceotide reductase subunit M1 expression are not related to survival in patients with resected pancreatic cancer given adjuvant chemotherapy
- Author
-
Elander, NO, Aughton, K, Ghaneh, P, Neoptolemos, JP, Palmer, DH, Cox, TF, Campbell, F, Costello, E, Halloran, CM, Mackey, JR, Scarfe, AG, Valle, JW, McDonald, AC, Carter, R, Tebbutt, NC, Goldstein, D, Shannon, J, Dervenis, C, Glimelius, B, Deakin, M, Charnley, RM, Anthoney, A, Lerch, MM, Mayerle, J, Olah, A, Buchler, MW, Greenhalf, W, and Canc, European Study Grp Pancreatic
- Subjects
Adult ,0301 basic medicine ,Cancer Research ,Ribonucleoside Diphosphate Reductase ,medicine.medical_treatment ,Adenocarcinoma ,Article ,Disease-Free Survival ,03 medical and health sciences ,Folinic acid ,Pancreatectomy ,0302 clinical medicine ,Pancreatic cancer ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Humans ,Medicine ,DCMP Deaminase ,Randomized Controlled Trials as Topic ,Chemotherapy ,Tissue microarray ,Manchester Cancer Research Centre ,business.industry ,ResearchInstitutes_Networks_Beacons/mcrc ,Tumor Suppressor Proteins ,Prognosis ,medicine.disease ,Immunohistochemistry ,Gemcitabine ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,Chemotherapy, Adjuvant ,Tissue Array Analysis ,Deoxycytidylate Deaminase ,030220 oncology & carcinogenesis ,Cancer research ,business ,medicine.drug - Abstract
Background: Deoxycytidylate deaminase (DCTD) and ribonucleotide reductase subunit M1 (RRM1) are potential prognostic and predictive biomarkers for pyrimidine-based chemotherapy in pancreatic adenocarcinoma. Methods: Immunohistochemical staining of DCTD and RRM1 was performed on tissue microarrays representing tumour samples from 303 patients in European Study Group for Pancreatic Cancer (ESPAC)-randomised adjuvant trials following pancreatic resection, 272 of whom had received gemcitabine or 5-fluorouracil with folinic acid in ESPAC-3(v2), and 31 patients from the combined ESPAC-3(v1) and ESPAC-1 post-operative pure observational groups. Results: Neither log-rank testing on dichotomised strata or Cox proportional hazard regression showed any relationship of DCTD or RRM1 expression levels to survival overall or by treatment group. Conclusions: Expression of either DCTD or RRM1 was not prognostic or predictive in patients with pancreatic adenocarcinoma who had had post-operative chemotherapy with either gemcitabine or 5-fluorouracil with folinic acid.
- Published
- 2018
- Full Text
- View/download PDF
5. Expression of dihydropyrimidine dehydrogenase (DPD) and hENT1 predicts survival in pancreatic cancer
- Author
-
Elander, NO, Aughton, K, Ghaneh, P, Neoptolemos, JP, Palmer, DH, Cox, TF, Campbell, F, Costello, E, Halloran, CM, Mackey, JR, Scarfe, AG, Valle, JW, McDonald, AC, Carter, R, Tebbutt, NC, Goldstein, D, Shannon, J, Dervenis, C, Glimelius, B, Deakin, M, Charnley, RM, Anthoney, A, Lerch, MM, Mayerle, J, Oláh, A, Büchler, MW, Greenhalf, W, Elander, NO, Aughton, K, Ghaneh, P, Neoptolemos, JP, Palmer, DH, Cox, TF, Campbell, F, Costello, E, Halloran, CM, Mackey, JR, Scarfe, AG, Valle, JW, McDonald, AC, Carter, R, Tebbutt, NC, Goldstein, D, Shannon, J, Dervenis, C, Glimelius, B, Deakin, M, Charnley, RM, Anthoney, A, Lerch, MM, Mayerle, J, Oláh, A, Büchler, MW, and Greenhalf, W
- Abstract
Background: Dihydropyrimidine dehydrogenase (DPD) tumour expression may provide added value to human equilibrative nucleoside transporter-1 (hENT1) tumour expression in predicting survival following pyrimidine-based adjuvant chemotherapy. Methods: DPD and hENT1 immunohistochemistry and scoring was completed on tumour cores from 238 patients with pancreatic cancer in the ESPAC-3(v2) trial, randomised to either postoperative gemcitabine or 5-fluorouracil/folinic acid (5FU/FA). Results: DPD tumour expression was associated with reduced overall survival (hazard ratio, HR = 1.73 [95% confidence interval, CI = 1.21-2.49], p = 0.003). This was significant in the 5FU/FA arm (HR = 2.07 [95% CI = 1.22-3.53], p = 0.007), but not in the gemcitabine arm (HR = 1.47 [0.91-3.37], p = 0.119). High hENT1 tumour expression was associated with increased survival in gemcitabine treated (HR = 0.56 [0.38-0.82], p = 0.003) but not in 5FU/FA treated patients (HR = 1.19 [0.80-1.78], p = 0.390). In patients with low hENT1 tumour expression, high DPD tumour expression was associated with a worse median [95% CI] survival in the 5FU/FA arm (9.7 [5.3-30.4] vs 29.2 [19.5-41.9] months, p = 0.002) but not in the gemcitabine arm (14.0 [9.1-15.7] vs. 18.0 [7.6-15.3] months, p = 1.000). The interaction of treatment arm and DPD expression was not significant (p = 0.303), but the interaction of treatment arm and hENT1 expression was (p = 0.009). Conclusion: DPD tumour expression was a negative prognostic biomarker. Together with tumour expression of hENT1, DPD tumour expression defined patient subgroups that might benefit from either postoperative 5FU/FA or gemcitabine.
- Published
- 2018
6. Effect of adjuvant chemotherapy with fluorouracil plus folinic acid or gemcitabine vs observation on survival in patients with resected periampullary adenocarcinoma: the ESPAC-3 periampullary cancer randomized trial.
- Author
-
Neoptolemos JP, Moore MJ, Cox TF, Valle JW, Palmer DH, McDonald AC, Carter R, Tebbutt NC, Dervenis C, Smith D, Glimelius B, Charnley RM, Lacaine F, Scarfe AG, Middleton MR, Anthoney A, Ghaneh P, Halloran CM, Lerch MM, and Oláh A
- Abstract
Context: Patients with periampullary adenocarcinomas undergo the same resectional surgery as that of patients with pancreatic ductal adenocarcinoma. Although adjuvant chemotherapy has been shown to have a survival benefit for pancreatic cancer, there have been no randomized trials for periampullary adenocarcinomas.Objective: To determine whether adjuvant chemotherapy (fluorouracil or gemcitabine) provides improved overall survival following resection.Design, Setting, and Patients: The European Study Group for Pancreatic Cancer (ESPAC)-3 periampullary trial, an open-label, phase 3, randomized controlled trial (July 2000-May 2008) in 100 centers in Europe, Australia, Japan, and Canada. Of the 428 patients included in the primary analysis, 297 had ampullary, 96 had bile duct, and 35 had other cancers.Interventions: One hundred forty-four patients were assigned to the observation group, 143 patients to receive 20 mg/m2 of folinic acid via intravenous bolus injection followed by 425 mg/m2 of fluorouracil via intravenous bolus injection administered 1 to 5 days every 28 days, and 141 patients to receive 1000 mg/m2 of intravenous infusion of gemcitabine once a week for 3 of every 4 weeks for 6 months.Main Outcome Measures: The primary outcome measure was overall survival with chemotherapy vs no chemotherapy; secondary measures were chemotherapy type, toxic effects, progression-free survival, and quality of life.Results: Eighty-eight patients (61%) in the observation group, 83 (58%) in the fluorouracil plus folinic acid group, and 73 (52%) in the gemcitabine group died. In the observation group, the median survival was 35.2 months (95%% CI, 27.2-43.0 months) and was 43.1 (95%, CI, 34.0-56.0) in the 2 chemotherapy groups (hazard ratio, 0.86; (95% CI, 0.66-1.11; χ2 = 1.33; P = .25). After adjusting for independent prognostic variables of age, bile duct cancer, poor tumor differentiation, and positive lymph nodes and after conducting multiple regression analysis, the hazard ratio for chemotherapy compared with observation was 0.75 (95% CI, 0.57-0.98; Wald χ2 = 4.53, P = .03).Conclusions: Among patients with resected periampullary adenocarcinoma, adjuvant chemotherapy, compared with observation, was not associated with a significant survival benefit in the primary analysis; however, multivariable analysis adjusting for prognostic variables demonstrated a statistically significant survival benefit associated with adjuvant chemotherapy.Trial Registration: clinicaltrials.gov Identifier: NCT00058201. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
7. Total knee arthroplasty in juvenile rheumatoid arthritis.
- Author
-
Palmer DH, Mulhall KJ, Thompson CA, Severson EP, Santos ERG, Saleh KJ, Palmer, David H, Mulhall, Kevin J, Thompson, Corey A, Severson, Erik P, Santos, Edward R G, and Saleh, Khaled J
- Abstract
Background: There is a paucity of reports regarding the long-term results of total knee arthroplasty in patients with juvenile rheumatoid arthritis. The purpose of this study was to evaluate the outcome of total knee arthroplasty in patients with juvenile rheumatoid arthritis who had been followed for a minimum of twelve years.Methods: Eight consecutive patients (fifteen knees) with juvenile rheumatoid arthritis underwent total knee arthroplasty at an average age of 16.8 years. Clinical evaluation of pain status, range of motion, and the ability to walk and radiographic evaluation of the alignment of the knees and component loosening were performed preoperatively and at a mean of 15.5 years postoperatively.Results: All patients had substantial pain and functional limitation before the surgery, and seven of the eight patients used a wheelchair. At the time of the latest follow-up, which was after revision surgery in three patients, all of the knees were pain-free and six patients were able to walk about the community. The mean arc of motion had increased from 36 degrees to 79 degrees . The final radiographic evaluation showed that thirteen of the fifteen knees were in neutral alignment and two were in valgus. Failure, defined as revision of any of the components or definite loosening as seen radiographically, occurred in three knees.Conclusions: Good results, in terms of pain relief and restoration of function, were seen at a minimum of twelve years following total knee arthroplasty in our series of patients with juvenile rheumatoid arthritis. This procedure is a reasonable option when nonoperative therapy has been inadequate for patients with severe disability and pain in this relatively young population. [ABSTRACT FROM AUTHOR]- Published
- 2005
8. Acute toxicity of selenium to three species of marine invertabrates, with notes on a continuous-flow test system
- Author
-
Ahsanullah, M and Palmer, DH
- Abstract
Acute toxicity of selenium (as sodium selenite) to three invertebrates common in Victorian coastal waters was determined using a continuous-flow test system. A description of the system is given. The three species were a cumacean, Cyclaspis usitata; juveniles of the bivalve Notocallista sp.; and two stocks of the laboratory-cultured amphipod Allorchestes compressa. Stock 1 of A. compressa was originally collected from Middle Spit, Western Port, and stock 11 from Somers, Western Port. The 96-h LC50 values for C. usitata, Notocallista sp. and stocks 1 and 11 of A. compressa were 6.12, 2.88, 6.17 and 4.77 mg selenium per litre respectively. The results indicated that juveniles of Notocallista sp. were more sensitive to selenium than adult A. compressa. The lack of information on the toxicity of selenium to invertebrates is indicated.
- Published
- 1980
- Full Text
- View/download PDF
9. Helicopter skiing wrist injuries: a case report of 'Bugaboo forearm'.
- Author
-
Palmer DH and Lane-Larsen CL
- Published
- 1994
- Full Text
- View/download PDF
10. A phase Ib/IIa trial to evaluate the CCK2 receptor antagonist Z-360 in combination with gemcitabine in patients with advanced pancreatic cancer.
- Author
-
Meyer T, Caplin ME, Palmer DH, Valle JW, Larvin M, Waters JS, Coxon F, Borbath I, Peeters M, Nagano E, and Kato H
- Abstract
AIM: To evaluate the combination of the gastrin antagonist Z-360 and gemcitabine for advanced pancreatic cancer. METHODS: Previously untreated patients with PC were randomly allocated to Z-360 120 mg, 240 mg or placebo. Z-360/placebo was given on day -3 and gemcitabine 1000 mg/m(2) commenced on day 1 followed by Z-360 on day 2. Thereafter Z-360/placebo was given twice daily concurrently with standard dose of gemcitabine. Pharmacokinetics for both drugs was measured alone and in combination. Toxicity, response and quality of life were also recorded. RESULTS: Thirty-three patients with a median age of 62 years were randomised of which six had locally advanced disease and 26 had metastatic disease. Analysis of the area under the plasma concentration versus time curve (AUC), the maximum observed concentration (Cmax(obs)) and the time of the maximum observed concentration (Tmax(obs)) for Z-360, gemcitabine and 2,2-difluorodeoxyuridine (dFdU), could not exclude an effect on the systemic exposure to Z-360, gemcitabine and dFdU when co-administration of Z-360 and gemcitabine was compared with single agent administration. The most commonly reported adverse events were nausea, abdominal pain, vomiting and fatigue. At the end of the study, 62.5%, 25% and 60% had stable disease in the 120 mg, 240 mg and placebo group, respectively. A higher proportion of patients in Z-360 groups reported improvement in pain. CONCLUSIONS: Z-360 is safe and well tolerated when combined with gemcitabine. A Phase III trial is needed to determine whether the combination of Z-360 and gemcitabine is superior to gemcitabine alone in advanced PC. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
11. Novel approaches in the management of bladder cancer
- Author
-
Greensmith, RMD, Hussain, SA, Copple, IM, and Palmer, DH
- Abstract
Background: In recent decades, despite survival improvements in the majority of high incidence cancers, survival rates for patients with bladder cancers have remained static. Key challenges in the treatment of bladder cancer are the acquisition of chemotherapy resistance, and progression to lethal muscle- invasive bladder cancer. Aims: This thesis aims to explore methods of pharmacological inhibition of chemotherapy resistance, mechanisms of bladder tumour invasion, and gene expression in bladder cancer. Rationale: Nuclear factor (erythroid-derived 2)-like 2 (NRF2) overexpression has been associated with chemotherapy resistance in a number of cancers including bladder. Therefore, the utility of the purported specific inhibitor of NRF2, brusatol in the attenuation of cisplatin chemotherapy resistance was investigated. In bladder cancer, cluster of differentiation-40 (CD40)-ligation with membrane bound CD40-Ligand (CD40L) has been shown to induce marked cell death. However, little is known about the role of CD40-ligation in tumour cell invasion. With CD40 currently under investigation as a putative target for anti-cancer therapy, it was a timely goal of this thesis to understand more about its role in tumour cell invasion. Until recently, knowledge of gene expression profiles in bladder cancer has been limited. Understanding of gene expression patterns in cancer may allow for enhanced prognostication, identification of potential drug targets and move the management of bladder cancer towards the ideal of personalised medicine. Methods: In vitro cytotoxicity assays were employed to determine the effect of brusatol on cisplatin sensitivity. Western blotting was employed to determine the effects of brusatol on proteins downstream of NRF2. An organotypic model of bladder cancer was used to investigate the effect of CD40-ligation on bladder tumour cell invasion. Non-cleavable membrane bound CD40-ligand delivered by adenoviral vector was used in CD40 expressing cell lines of invasive and non-invasive origin. The effect of CD40-ligation was investigated downstream by western analysis and RNA microarray. Whole-transcript based microarray analysis was performed on bladder biopsies obtained from 19 patients, and 10 controls. Unsupervised hierarchical clustering was used to identify samples with similar expression profiles. Hypergeometric analysis was used to identify canonical pathways and curated networks with statistically significant enrichment of differentially expressed genes. Samples from a further 6 patients (muscle invasive bladder cancer (MIBC) n = 3, normal tissue n = 3) were tested for osteopontin expression by immunohistochemistry. Results: Brusatol increased the sensitivity of bladder cancer cell lines to cisplatin in a dosage and cell line dependent manner. Furthermore, brusatol exhibited marked toxicity as a standalone treatment. However, evidence suggests that brusatol does not act as a specific inhibitor of NRF2. CD40-ligation with non-cleavable membrane-bound CD40-ligand led to marked cell death in CD40-positive non-muscle invasive cell lines. However, in muscle- invasive CD40-positive cell lines marked inhibition of tumour invasion was also observed. Expression analysis of family with sequence similarity member 83 d (FAM83D), fibronectin (FN1), matrix metalloproteinase 1 (MMP1) and anillin (ANLN) revealed downregulation of FN1, FAM83D, and ANLN, and upregulation of MMP1, with a concurrent upregulation of MMP1 and a paradoxical increase of FN1 in EJ cells, with no change in FN1 and MMP1 expression in RT112 cells. xiii Pathways associated with cell cycle and proliferation were markedly upregulated in muscle-invasive and grade 3 cancers. Genes associated with the classical complement pathway were downregulated in non-muscle invasive cancer. Osteopontin was significantly overexpressed in invasive cancer compared to healthy tissue. Conclusions: It is unlikely that brusatol is a specific inhibitor of NRF2 however its marked cytotoxicity as a stand-alone agent and potentiating effects in combination with cisplatin are rationale for further investigation. CD40-ligation attenuates the invasion of MIBC cells in an organotypic model of bladder cancer. However, it is unclear whether the attenuation of invasion is due to the inhibition of invasive pathways, or direct induction of apoptosis. Nevertheless, with the usage of CD40-agnoistic antibodies currently in clinical trials, attenuation of tumour invasion is a promising effect. Gene expression analysis identifies divergent pathways that may be useful in the stratification of bladder cancers, and the identification of drug targets. Furthermore, this study supports a key role for osteopontin in bladder cancer, which warrants further investigation as a marker prognostic of muscle invasive disease.
- Published
- 2017
12. Metastasis-associated Macrophages Orchestrate the Formation of a Hospitable Metastatic Niche in Pancreatic Cancer
- Author
-
Nielsen, SR, Schmid, MC, and Palmer, DH
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers with an overall 5-year survival rate
- Published
- 2017
- Full Text
- View/download PDF
13. Sorafenib in advanced hepatocellular carcinoma.
- Author
-
Spinzi G, Paggi S, Copur MS, Palmer DH, Llovet JM, Bruix J, and Roberts LR
- Published
- 2008
14. Pancreatic Adenocarcinoma: Long-Term Outcomes of Adjuvant Therapy in the ESPAC4 Phase III Trial.
- Author
-
Palmer DH, Jackson R, Springfeld C, Ghaneh P, Rawcliffe C, Halloran CM, Faluyi O, Cunningham D, Wadsley J, Darby S, Meyer T, Gillmore R, Lind P, Glimelius B, Falk S, Ma YT, Middleton GW, Cummins S, Ross PJ, Wasan H, McDonald A, Crosby T, Hammel P, Borg D, Sothi S, Valle JW, Mehrabi A, Bailey P, Tjaden C, Michalski C, Hackert T, Büchler MW, and Neoptolemos JP
- Abstract
Purpose: The ESPAC4 trial showed that adjuvant chemotherapy with gemcitabine plus capecitabine (GemCap) produced longer overall survival (OS) than gemcitabine monotherapy. Subsequently, the PRODIGE24-CCTG PA.6 trial showed even longer survival for modified fluorouracil, folinic acid, irinotecan, and oxaliplatin (mFOLFIRINOX) than gemcitabine but had more restrictive eligibility criteria. Our aim was to analyze the ESPAC4 survival on long-term follow-up., Methods: The OS of 732 ESPAC4 patients comparing 367 randomly assigned to gemcitabine and 365 to GemCap was previously reported after a median follow-up time of 43.2 months (95% CI, 39.7 to 45.5) and 458 deaths. Analysis was now carried out after a median follow-up of 104 months (101-108) and 566 deaths., Results: The median OS was 29.5 months (27.5-32.1) for all patients, 28.4 months (25.2-32.0) in the gemcitabine group and 31.6 months (26.5-38.0) in the GemCap group (hazard ratio [HR], 0.83 [0.71 to 0.98]; P = .031). R0 patients given gemcitabine had a median survival of 32.2 months (27.9-41.6) compared with 49.9 months (39.0-82.3) for those given GemCap (HR, 0.63 [0.47 to 0.84]; P = .002). Lymph node-negative patients had significantly higher 5 year OS rates on GemCap (59% [49%-71%]) than gemcitabine (53% [42%-66%]; HR, 0.63 [0.41 to 0.98]; P = .04) but not those with positive lymph nodes ( P = .225). The OS advantage for GemCap was retained in the PRODIGE24 subgroup of 193 (26.4%) ESPAC4 patients not eligible for PRODIGE24 with a median survival of 20.7 (16.2-27.3) months in patients allocated to gemcitabine compared with 25.9 (22.3-30.2) months for ineligible patients allocated to GemCap (HR, 0.71 [95% CI, 0.52 to 0.98]; χ
2 log-rank-1df = 4.31; P = .038)., Conclusion: GemCap is a standard option for patients not eligible for mFOLFIRINOX. Exploratory evidence suggests that GemCap may be particularly efficacious in R0 patients and also in lymph node-negative patients.- Published
- 2024
- Full Text
- View/download PDF
15. Macrophage-fibroblast JAK/STAT dependent crosstalk promotes liver metastatic outgrowth in pancreatic cancer.
- Author
-
Raymant M, Astuti Y, Alvaro-Espinosa L, Green D, Quaranta V, Bellomo G, Glenn M, Chandran-Gorner V, Palmer DH, Halloran C, Ghaneh P, Henderson NC, Morton JP, Valiente M, Mielgo A, and Schmid MC
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Signal Transduction, Janus Kinases metabolism, Mice, Inbred C57BL, Fibroblasts metabolism, Fibroblasts pathology, Male, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Female, Pancreatic Neoplasms pathology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms immunology, Liver Neoplasms secondary, Liver Neoplasms metabolism, Liver Neoplasms genetics, Liver Neoplasms pathology, Liver Neoplasms immunology, STAT3 Transcription Factor metabolism, STAT3 Transcription Factor genetics, Macrophages metabolism, Macrophages immunology, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal immunology
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease for which better therapies are urgently needed. Fibroblasts and macrophages are heterogeneous cell populations able to enhance metastasis, but the role of a macrophage-fibroblast crosstalk in regulating their pro-metastatic functions remains poorly understood. Here we deconvolve how macrophages regulate metastasis-associated fibroblast (MAF) heterogeneity in the liver. We identify three functionally distinct MAF populations, among which the generation of pro-metastatic and immunoregulatory myofibroblastic-MAFs (myMAFs) critically depends on macrophages. Mechanistically, myMAFs are induced through a STAT3-dependent mechanism driven by macrophage-derived progranulin and cancer cell-secreted leukaemia inhibitory factor (LIF). In a reciprocal manner, myMAF secreted osteopontin promotes an immunosuppressive macrophage phenotype resulting in the inhibition of cytotoxic T cell functions. Pharmacological blockade of STAT3 or myMAF-specific genetic depletion of STAT3 restores an anti-tumour immune response and reduces metastases. Our findings provide molecular insights into the complex macrophage-fibroblast interactions in tumours and reveal potential targets to inhibit PDAC liver metastasis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Molecular biology of cholangiocarcinoma and its implications for targeted therapy in patient management.
- Author
-
Gilbert TM, Randle L, Quinn M, McGreevy O, O'leary L, Young R, Diaz-Neito R, Jones RP, Greenhalf B, Goldring C, Fenwick S, Malik H, and Palmer DH
- Abstract
Cholangiocarcinoma (CCA) remains a devastating malignancy and a significant challenge to treat. The majority of CCA patients are diagnosed at an advanced stage, making the disease incurable in most cases. The advent of high-throughput genetic sequencing has significantly improved our understanding of the molecular biology underpinning cancer. The identification of 'druggable' genetic aberrations and the development of novel targeted therapies against them is opening up new treatment strategies. Currently, 3 targeted therapies are approved for use in CCA; Ivosidenib in patients with IDH1 mutations and Infigratinib/Pemigatinib in those with FGFR2 fusions. As our understanding of the biology underpinning CCA continues to improve it is highly likely that additional targeted therapies will become available in the near future. This is important, as it is thought up to 40 % of CCA patients harbour a potentially actionable mutation. In this review we provide an overview of the molecular pathogenesis of CCA and highlight currently available and potential future targeted treatments., Competing Interests: Declaration of competing interest Palmer has grant funding from BMS, Nucana, Astra Zeneca, Sirtex, honoraria from Boston Scientific and Sirtex, and support for travel from Nucana. Randle reports a relationship with National Centre for the Replacement Refinement and Reduction of Animals in Research (NC3Rs) that includes grant funding and membership of the NC3RS PhD studentship assessment board. Goldring reports financial support by the University of Liverpool. Jones, Young, Diaz-Neito, O’leary, McGreevy, Gilbert, Quinn, Fenwick and Malik declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2024 Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
17. Treatment sequences and prognostic/predictive factors in metastatic pancreatic ductal adenocarcinoma: univariate and multivariate analyses of a real-world study in Europe.
- Author
-
Taieb J, Seufferlein T, Reni M, Palmer DH, Bridgewater JA, Cubillo A, Prager GW, Vermeire A, Hédouin-Biville F, Teng Z, and Macarulla T
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Europe epidemiology, Gemcitabine, Multivariate Analysis, Prognosis, Retrospective Studies, Pancreatic Neoplasms, Adenocarcinoma, Pancreatic Neoplasms drug therapy
- Abstract
Background: Real-world data on treatment patterns/outcomes for metastatic pancreatic cancer (mPAC) are limited. This study aims to assess real-world treatment patterns, survival outcomes, and prognostic/predictive factors in patients with mPAC., Methods: Retrospective, observational, chart-review involving medical oncologists and gastroenterologists from five European countries. Physicians reported information on disease and patient characteristics, diagnosis, and treatment for patients diagnosed with mPAC from January-October 2016. Outcomes included median progression-free survival (mPFS), median overall survival (mOS), and the impact of baseline performance status on survival. Univariate/multivariate regression analyses were undertaken to identify prognostic/predictive factors., Results: Three hundred four physicians and 3432 patients were included. First-line therapies included modified (m)FOLFIRINOX (28.4%), gemcitabine + nab-paclitaxel (28.0%), and gemcitabine monotherapy (23.0%). Frequent second-line therapies were gemcitabine monotherapy (25.0%), fluorouracil (5-FU) + oxaliplatin (21.8%), and gemcitabine + nab-paclitaxel (16.7%). Most frequent first- to second-line treatment sequences were gemcitabine + nab-paclitaxel followed by fluoropyrimidine combinations. Longest unadjusted estimated mOS was observed with (m)FOLFIRINOX followed by gemcitabine-based combinations (19.1 months). Multivariate analysis identified significant prognostic/predictive factors for OS and PFS including performance status and carbohydrate antigen 19-9 (CA 19-9) levels., Conclusions: Treatment and treatment sequences were generally in accordance with guidelines at the time of the study. Identification of prognostic/predictive factors for survival may help inform the individualised management of mPAC patients in the future., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
18. PINCER (A Platform Study for solId orgaN CancERs): an agile pan-network platform study to deliver high-quality translational research.
- Author
-
Jones RP, Mielgo A, Schmid M, Bury D, Andrews T, Burdak-Rothkamm S, Shackcloth M, J S Cross T, Fenwick S, Malik HZ, Diaz-Nieto R, Ottensmeier C, Palmer DH, and Vimalachandran D
- Subjects
- Humans, Translational Research, Biomedical, Neoplasms genetics, Neoplasms therapy
- Published
- 2023
- Full Text
- View/download PDF
19. The Potent G-Quadruplex-Binding Compound QN-302 Downregulates S100P Gene Expression in Cells and in an In Vivo Model of Pancreatic Cancer.
- Author
-
Ahmed AA, Greenhalf W, Palmer DH, Williams N, Worthington J, Arshad T, Haider S, Alexandrou E, Guneri D, Waller ZAE, and Neidle S
- Subjects
- Humans, Cell Line, Tumor, Calcium-Binding Proteins metabolism, Gene Expression, Neoplasm Proteins metabolism, Pancreatic Neoplasms, G-Quadruplexes, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism
- Abstract
The naphthalene diimide compound QN-302, designed to bind to G-quadruplex DNA sequences within the promoter regions of cancer-related genes, has high anti-proliferative activity in pancreatic cancer cell lines and anti-tumor activity in several experimental models for the disease. We show here that QN-302 also causes downregulation of the expression of the S100P gene and the S100P protein in cells and in vivo. This protein is well established as being involved in key proliferation and motility pathways in several human cancers and has been identified as a potential biomarker in pancreatic cancer. The S100P gene contains 60 putative quadruplex-forming sequences, one of which is in the promoter region, 48 nucleotides upstream from the transcription start site. We report biophysical and molecular modeling studies showing that this sequence forms a highly stable G-quadruplex in vitro, which is further stabilized by QN-302. We also report transcriptome analyses showing that S100P expression is highly upregulated in tissues from human pancreatic cancer tumors, compared to normal pancreas material. The extent of upregulation is dependent on the degree of differentiation of tumor cells, with the most poorly differentiated, from more advanced disease, having the highest level of S100P expression. The experimental drug QN-302 is currently in pre-IND development (as of Q1 2023), and its ability to downregulate S100P protein expression supports a role for this protein as a marker of therapeutic response in pancreatic cancer. These results are also consistent with the hypothesis that the S100P promoter G-quadruplex is a potential therapeutic target in pancreatic cancer at the transcriptional level for QN-302.
- Published
- 2023
- Full Text
- View/download PDF
20. GATA4 and GATA6 loss-of-expression is associated with extinction of the classical programme and poor outcome in pancreatic ductal adenocarcinoma.
- Author
-
de Andrés MP, Jackson RJ, Felipe I, Zagorac S, Pilarsky C, Schlitter AM, Martinez de Villareal J, Jang GH, Costello E, Gallinger S, Ghaneh P, Greenhalf W, Knösel T, Palmer DH, Ruemmele P, Weichert W, Buechler M, Hackert T, Neoptolemos JP, Notta F, Malats N, Martinelli P, and Real FX
- Subjects
- Humans, Pancreas pathology, Gene Expression Profiling, GATA6 Transcription Factor genetics, GATA4 Transcription Factor genetics, GATA4 Transcription Factor metabolism, Pancreatic Neoplasms pathology, Carcinoma, Pancreatic Ductal pathology
- Abstract
Objective: GATA6 is a key regulator of the classical phenotype in pancreatic ductal adenocarcinoma (PDAC). Low GATA6 expression associates with poor patient outcome. GATA4 is the second most expressed GATA factor in the pancreas. We assessed whether, and how, GATA4 contributes to PDAC phenotype and analysed the association of expression with outcome and response to chemotherapy., Design: We analysed PDAC transcriptomic data, stratifying cases according to GATA4 and GATA6 expression and identified differentially expressed genes and pathways. The genome-wide distribution of GATA4 was assessed, as well as the effects of GATA4 knockdown. A multicentre tissue microarray study to assess GATA4 and GATA6 expression in samples (n=745) from patients with resectable was performed. GATA4 and GATA6 levels were dichotomised into high/low categorical variables; association with outcome was assessed using univariable and multivariable Cox regression models., Results: GATA4 messenger RNA is enriched in classical, compared with basal-like tumours. We classified samples in 4 groups as high/low for GATA4 and GATA6 . Reduced expression of GATA4 had a minor transcriptional impact but low expression of GATA4 enhanced the effects of GATA6 low expression. GATA4 and GATA6 display a partially overlapping genome-wide distribution, mainly at promoters. Reduced expression of both proteins in tumours was associated with the worst patient survival. GATA4 and GATA6 expression significantly decreased in metastases and negatively correlated with basal markers., Conclusions: GATA4 and GATA6 cooperate to maintain the classical phenotype. Our findings provide compelling rationale to assess their expression as biomarkers of poor prognosis and therapeutic response., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
21. Long-Term Outcomes and Exploratory Analyses of the Randomized Phase III BILCAP Study.
- Author
-
Bridgewater J, Fletcher P, Palmer DH, Malik HZ, Prasad R, Mirza D, Anthony A, Corrie P, Falk S, Finch-Jones M, Wasan H, Ross P, Wall L, Wadsley J, Evans TR, Stocken D, Stubbs C, Praseedom R, Ma YT, Davidson B, Neoptolemos J, Iveson T, Cunningham D, Garden OJ, Valle JW, and Primrose J
- Subjects
- Adolescent, Capecitabine, Chemotherapy, Adjuvant, Humans, Prognosis, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biliary Tract Neoplasms drug therapy
- Abstract
Purpose: The BILCAP study described a modest benefit for capecitabine as adjuvant therapy for curatively resected biliary tract cancer (BTC), and capecitabine has become the standard of care. We present the long-term data and novel exploratory subgroup analyses., Methods: This randomized, controlled, multicenter, phase III study recruited patients age 18 years or older with histologically confirmed cholangiocarcinoma or muscle-invasive gallbladder cancer after resection with curative intent and an Eastern Cooperative Oncology Group performance status of < 2. Patients were randomly assigned 1:1 to receive oral capecitabine (1,250 mg/m
2 twice daily on days 1-14 of a 21-day cycle, for eight cycles) or observation. The primary outcome was overall survival (OS). This study is registered with EudraCT 2005-003318-13., Results: Between March 15, 2006, and December 4, 2014, 447 patients were enrolled; 223 patients with BTC resected with curative intent were randomly assigned to the capecitabine group and 224 to the observation group. At the data cutoff of January 21, 2021, the median follow-up for all patients was 106 months (95% CI, 98 to 108). In the intention-to-treat analysis, the median OS was 49.6 months (95% CI, 35.1 to 59.1) in the capecitabine group compared with 36.1 months (95% CI, 29.7 to 44.2) in the observation group (adjusted hazard ratio 0.84; 95% CI, 0.67 to 1.06). In a protocol-specified sensitivity analysis, adjusting for minimization factors, nodal status, grade, and sex, the OS hazard ratio was 0.74 (95% CI, 0.59 to 0.94). We further describe the prognostic impact of R status, grade, nodal status, and sex., Conclusion: This long-term analysis supports the previous analysis, suggesting that capecitabine can improve OS in patients with resected BTC when used as adjuvant chemotherapy after surgery and should be considered as the standard of care., Competing Interests: John BridgewaterHonoraria: Merck Serono, ServierConsulting or Advisory Role: Merck Serono, Servier, Roche, Bayer, AstraZeneca, Incyte, Basilea, Taiho OncologySpeakers' Bureau: Celgene, Amgen, Bristol Myers SquibbResearch Funding: Amgen, Incyte (Inst)Travel, Accommodations, Expenses: MSD Oncology, Merck Serono, Servier, Bristol Myers Squibb/Medarex, Bristol Myers Squibb Daniel H. PalmerConsulting or Advisory Role: BMS (Inst), MSD, AstraZeneca, Eisai, Sirtex Medical, Roche, Boston Scientific, ViatrisResearch Funding: BMS, Bayer, Sirtex Medical, AstraZeneca, NuCana Darius MirzaStock and Other Ownership Interests: OrganOx Ltd Pippa CorrieHonoraria: Novartis, Merck Sharp & Dohme, Pierre Fabre, Bristol Myers SquibbConsulting or Advisory Role: Novartis, Bristol Myers Squibb, Merck Sharp & Dohme, Incyte, Pierre Fabre, Roche, MicrobioticaSpeakers' Bureau: Merck Sharp & Dohme, Novartis, Bristol Myers SquibbResearch Funding: MSD (Inst), Bristol Myers Squibb (Inst), Novartis (Inst), Array BioPharma (Inst), Celgene (Inst), Halozyme (Inst), Iovance Biotherapeutics (Inst), Lilly (Inst)Travel, Accommodations, Expenses: Bristol Myers Squibb, Merck Sharp & Dohme Stephen FalkStock and Other Ownership Interests: GlaxoSmithKlineConsulting or Advisory Role: BMSSpeakers' Bureau: Merck Serono, Servier, AmgenResearch Funding: Gilead Sciences (Inst), AstraZeneca (Inst), Novartis (Inst)Travel, Accommodations, Expenses: CelgeneOther Relationship: Servier Harpreet WasanHonoraria: Merck KGaA, Celgene, Sirtex Medical, Servier, Array BioPharma, Roche/Genentech, ERYTECH Pharma, Incyte, ZymeworksConsulting or Advisory Role: Roche Pharma AG, ERYTECH Pharma, Shire, Incyte, Sirtex medical, Pierre Fabre, Pfizer, BayerSpeakers' Bureau: Sirtex Medical, Celgene, Merck KGaA, Servier, IncyteResearch Funding: Sirtex Medical (Inst), Merck KGaA (Inst), Pfizer (Inst), Merck Sharp & Dohme (Inst) Paul RossStock and Other Ownership Interests: Perci HealthHonoraria: Sirtex Medical, Roche, AstraZeneca, MerckConsulting or Advisory Role: Sirtex Medical, Roche, AstraZeneca, Amgen, Boston ScientificSpeakers' Bureau: Amgen, Merck, Servier, Boston Scientific, Roche, EisaiResearch Funding: Sanofi (Inst), Bayer (Inst)Travel, Accommodations, Expenses: Roche, Ipsen Lucy WallResearch Funding: Merck Serono Jonathan WadsleyConsulting or Advisory Role: Lilly (Inst), Novartis (Inst), Roche (Inst), Ipsen (Inst), Bayer (Inst)Speakers' Bureau: Genzyme, EisaiResearch Funding: AstraZeneca, Genzyme Thomas R. EvansHonoraria: Eisai (Inst), Merck Sharp & Dohme (Inst), Bristol Myers Squibb (Inst), NuCana (Inst), Roche/Genentech (Inst), Ascelia (Inst), AstraZeneca (Inst), Bicycle Therapeutics (Inst), Medivir (Inst)Consulting or Advisory Role: Karus Therapeutics (Inst)Speakers' Bureau: Eisai (Inst), Bristol Myers Squibb (Inst), Merck Sharp & Dohme (Inst), NuCana (Inst), United Medical (Inst), AstraZeneca (Inst), Roche/Genentech (Inst), Medivir (Inst)Research Funding: Bristol Myers Squibb (Inst), AstraZeneca (Inst), BeiGene (Inst), Basilea (Inst), Celgene (Inst), Eisai (Inst), Merck Sharp & Dohme (Inst), MiNA Therapeutics (Inst), Bicycle Therapeutics (Inst), Lilly (Inst), Merck Serono (Inst), Janssen (Inst), Johnson & Johnson (Inst), Verastem (Inst), Roche/Genentech (Inst), Novartis (Inst), Iovance Biotherapeutics (Inst), Medivir (Inst), Sanofi/Aventis (Inst), Clovis Oncology (Inst), Plexxikon (Inst), NuCana (Inst), Sierra Pharma (Inst), GlaxoSmithKline (Inst), Halozyme (Inst), CytomX Therapeutics (Inst), Vertex (Inst), Athenex (Inst), Adaptimmune (Inst), Immunocore (Inst), Modulate Pharma (Inst), Berg Pharma (Inst), Starpharma (Inst), BiolineRx (Inst), iOnctura (Inst), UCB (Inst), Sapience Therapeutics (Inst), Astellas Pharma (Inst), Boehringer Ingelheim (Inst), Seattle Genetics (Inst)Expert Testimony: Medivir (Inst)Travel, Accommodations, Expenses: Bristol Myers Squibb, Merck Sharp & Dohme, Pierre Fabre, Eisai, NuCanaOther Relationship: Genmab (Inst) Deborah StockenResearch Funding: Abbott Diabetes (Inst) Yuk Ting MaHonoraria: Boston ScientificConsulting or Advisory Role: Eisai, Roche, AstraZeneca/MedImmune, Ipsen, Faron PharmaceuticalsResearch Funding: Eisai (Inst), AstraZeneca/Merck (Inst), Faron Pharmaceuticals (Inst), MINA Therapeutics (Inst) John NeoptolemosResearch Funding: Dietmar Hopp Stiftung (Inst), Stiftung Deutsche Krebshilfe (Inst), Heidelberger Stiftung Chirurgie (Inst) Timothy IvesonHonoraria: Servier, AmgenConsulting or Advisory Role: Servier, Bristol Myers Squibb, Pierre Fabre, MSD Oncology David CunninghamStock and Other Ownership Interests: OVIBIOConsulting or Advisory Role: OVIBIOResearch Funding: Celgene (Inst), MedImmune (Inst), Bayer (Inst), 4SC (Inst), Clovis Oncology (Inst), Lilly (Inst), Roche (Inst), Leap Oncology (Inst) Juan W. ValleHonoraria: IpsenConsulting or Advisory Role: Ipsen, Novartis, AstraZeneca, Merck, Agios, Pfizer, PCI Biotech, Incyte, Keocyt, QED Therapeutics, Pieris Pharmaceuticals, Genoscience Pharma, Mundipharma EDO GmbH, Wren Laboratories, NuCana, SERVIER, Debiopharm Group, Imaging Equipment Limited, Hutchison MediPharma, Zymeworks, Aptitude Health, Sirtex Medical, Baxter, Medivir, Cantargia ABSpeakers' Bureau: Novartis, Ipsen, NuCana, Imaging Equipment Limited, Mylan, Incyte, ServierTravel, Accommodations, Expenses: NuCana, Lilly, Roche, AstraZeneca/MedImmuneNo other potential conflicts of interest were reported.- Published
- 2022
- Full Text
- View/download PDF
22. Pembrolizumab Monotherapy for Previously Untreated Advanced Hepatocellular Carcinoma: Data from the Open-Label, Phase II KEYNOTE-224 Trial.
- Author
-
Verset G, Borbath I, Karwal M, Verslype C, Van Vlierberghe H, Kardosh A, Zagonel V, Stal P, Sarker D, Palmer DH, Vogel A, Edeline J, Cattan S, Kudo M, Cheng AL, Ogasawara S, Daniele B, Chan SL, Knox JJ, Qin S, Siegel AB, Chisamore M, Hatogai K, Wang A, Finn RS, and Zhu AX
- Subjects
- Humans, Antibodies, Monoclonal, Humanized adverse effects, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy
- Abstract
Purpose: KEYNOTE-224 cohort 1 demonstrated that pembrolizumab was efficacious and tolerable in patients with advanced hepatocellular carcinoma (HCC) previously treated with sorafenib. We report results from KEYNOTE-224 (NCT02702414) cohort 2, which enrolled patients with advanced HCC and no prior systemic therapy., Patients and Methods: KEYNOTE-224 was an open-label, multicountry phase II trial. Eligible patients in cohort 2 had advanced HCC not amenable or refractory to locoregional therapy and not previously treated with systemic therapy. Patients received pembrolizumab 200 mg intravenously every 3 weeks for ≤2 years. Primary endpoint was objective response rate (ORR) by central imaging review per RECIST v1.1. Secondary endpoints included duration of response (DOR), disease control rate (DCR), time to progression (TTP), progression-free survival (PFS), overall survival (OS), and safety/tolerability., Results: Between September 4, 2018, and February 20, 2019, 51 patients were allocated in cohort 2. The median time from the first dose to data cutoff (January 19, 2021) was 27 months (range, 23-29). ORR was 16% [95% confidence interval (CI), 7-29] and was similar across key subgroups. Median DOR was 16 months (range, 3-24+), and DCR was 57%. The median PFS was 4 months (95% CI, 2-8), and median TTP was 4 months (95% CI, 3-9). Median OS was 17 months (95% CI, 8-23). Grade ≥3 treatment-related adverse events occurred in 16% of patients., Conclusions: In patients with advanced HCC with no prior systemic therapy, pembrolizumab provided durable antitumor activity, promising OS, and had a safety profile consistent with previous observations. These findings support further evaluation of pembrolizumab-based regimens for HCC., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
23. Treatment of unresectable locally advanced pancreatic cancer with percutaneous irreversible electroporation (IRE) following initial systemic chemotherapy (LAP-PIE) trial: study protocol for a feasibility randomised controlled trial.
- Author
-
Rai ZL, Ranieri V, Palmer DH, Littler P, Ghaneh P, Gurusamy K, Manas D, Pizzo E, Psarelli EE, Gilmore R, Peddu P, Bartlett DC, de Liguori Carino N, and Davidson BR
- Subjects
- Feasibility Studies, Humans, Multicenter Studies as Topic, Pancreas, Randomized Controlled Trials as Topic, Treatment Outcome, Electroporation methods, Pancreatic Neoplasms drug therapy
- Abstract
Background: Approximately 30% of patients with pancreas cancer have unresectable locally advanced disease, which is currently treated with systemic chemotherapy. A new treatment option of irreversible electroporation (IRE) has been investigated for these patients since 2005. Cohort studies suggest that IRE confers a survival advantage, but with associated, procedure-related complications. Selection bias may account for improved survival and there have been no prospective randomised trials evaluating the harms and benefits of therapy. The aim of this trial is to evaluate the feasibility of a randomised comparison of IRE therapy with chemotherapy versus chemotherapy alone in patients with locally advanced pancreatic cancer (LAPC)., Methods and Analysis: Eligible patients with LAPC who have undergone first-line 5-FluoroUracil, Leucovorin, Irinotecan and Oxaliplatin chemotherapy will be randomised to receive either a single session of IRE followed by (if indicated) further chemotherapy or to chemotherapy alone (standard of care). Fifty patients from up to seven specialist pancreas centres in the UK will be recruited over a period of 15 months. Trial follow-up will be 12 months. The primary outcome measure is ability to recruit. Secondary objectives include practicality and technical success of treatment, acceptability of treatment to patients and clinicians and safety of treatment. A qualitative study has been incorporated to evaluate the patient and clinician perspective of the locally advanced pancreatic cancer with percutaneous irreversible electroporation trial. It is likely that the data obtained will guide the structure, the primary outcome measure, the power and the duration of a subsequent multicentre randomised controlled trial aimed at establishing the clinical efficiency of pancreas IRE therapy. Indicative procedure-related costings will be collected in this feasibility trial, which will inform the cost evaluation in the subsequent study on efficiency., Ethics and Dissemination: The protocol has received approval by London-Brent Research Ethics Committee reference number 21/LO/0077.Results will be analysed following completion of trial recruitment and follow-up. Results will be presented to international conferences with an interest in oncology, hepatopancreaticobiliary surgery and interventional radiology and be published in a peer-reviewed journal., Trial Registration Number: ISRCTN14986389., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
24. Updated efficacy and safety of KEYNOTE-224: a phase II study of pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib.
- Author
-
Kudo M, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer DH, Verslype C, Zagonel V, Fartoux L, Vogel A, Sarker D, Verset G, Chan SL, Knox J, Daniele B, Yau T, Gurary EB, Siegel AB, Wang A, Cheng AL, and Zhu AX
- Subjects
- Adult, Antibodies, Monoclonal, Humanized adverse effects, Humans, Sorafenib therapeutic use, Carcinoma, Hepatocellular pathology, Liver Neoplasms pathology
- Abstract
Objective: Pembrolizumab, a PD-1 inhibitor, demonstrated anti-tumour activity and tolerability in patients treated with sorafenib and with advanced hepatocellular carcinoma in KEYNOTE-224. Longer-term efficacy and safety after ∼2.5 years of additional follow-up are reported., Patients and Methods: Adults with confirmed hepatocellular carcinoma who experienced progression after or intolerance to sorafenib treatment received pembrolizumab 200 mg every 3 weeks for ≤35 cycles or until confirmed progression, unacceptable toxicity, withdrawal of consent or investigator decision. The primary end-point was objective response rate assessed by blinded independent central review per Response Evaluation Criteria in Solid Tumours v1.1. The secondary end-points included duration of response, disease control rate, time to progression, progression-free survival, overall survival and adverse events., Results: Efficacy and safety were assessed in 104 patients. The median time from first dose to data cutoff was 45.1 months (range, 41.3-49.3). Objective response rate was 18.3% (95% CI: 11.4-27.1), and median duration of response was 21.0 months (range, 3.1 to 39.5+). Disease control rate was 61.5%, and median time to progression was 4.8 months (95% CI: 3.9-7.0). Median progression-free survival was 4.9 months (95% CI: 3.5-6.7) and median overall survival was 13.2 months (95% CI: 9.7-15.3). Of 104 patients, 76 (73.1%) patients reported treatment-related adverse events; most were low grade in severity (grade 3-4, n = 26 [25.0%]; grade 5, n = 1 [1.0%]). Immune-mediated hepatitis occurred in 3 patients (all grade 3). No viral-induced hepatitis flares occurred., Conclusions: After ∼2.5 years of additional follow-up, pembrolizumab continued to provide durable anti-tumour activity and no new safety concerns were identified., Gov Identifier: NCT02702414., Competing Interests: Conflict of interest statement The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: M. Kudo has received a grant and fees from Eisai, and Bristol Myers Squibb; has received fees from Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, and Eli Lilly; has served a consultant for Bayer, Eisai, Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, Roche, Bristol Myers Squibb, and Ono; and has received a grant from Otsuka, Taiho, EA Pharma, Takeda, Gilead, AbbVie, and Sumitomo Dainippon Pharma; R.S. Finn has received fees and a grant to his institution from Merck, Bayer, Eli Lilly, Bristol Myers Squibb, Eisai, Pfizer, Roche/Genentech; and has received fees from AstraZeneca and CStone; J. Edeline has received personal fees from Bayer, Bristol Myers Squibb, AstraZeneca, Eisai, Ipsen, Boston Scientific, and Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA; and has received grants from Bristol Myers Squibb, BeiGene, and Boston Scientific; S. Cattan has nothing to disclose; S. Ogasawara has received a grant and fees from Bayer, Eisai, Eli Lilly; and has received fees from Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, AstraZeneca, and Chugai; D. Palmer has received a grant and fees from Bristol Myers Squibb, NuCana Inc, and Sirtex; has received a grant from AstraZeneca; and has received fees from Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, Roche, and Eisai; C. Verslype has received a grant from Baye and Ipsen and has served as a consultant for Bayer, Ipsen, and Roche; V. Zagonel has served in an advisory capacity or as a consultant for Bristol Myers Squibb and Merck; has served as a speaker for Bayer, Roche, Bristol Myers Squibb, Astellas Pharma, SERVIER, AstraZeneca, Eli Lilly; and has received travel, accommodations, and expenses from Bayer, Roche, and SERVIER; L. Fartoux has no conflicts of interest to report. A. Vogel has received fees for serving in an advisory capacity/consultant, has received speaking fees, has received fees for expert testimony, and has received honoraria from AstraZeneca, Bayer, Bristol Myers Squibb, BTG, Eli Lilly, Incyte Corporation, Ipsen, Janssen, Merck, Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, Novartis, Pierre Fabre, Roche, Sanofi, and Servier; D. Sarker has received fees from Eisai, Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, AstraZeneca, Bayer, and Surface Oncology; has received fees and non-financial support from Ipsen; has received non-financial support from MiNA Therapeutics; and has received a grant from Roche; G. Verset has received a grant from Terumo; has served as a consultant for Terumo and BTG; has served in an advisory capacity for Bayer, Eisai, and Roche; and has received travel expenses for attending congresses from Bayer, Bristol Myers Squibb, and Ipsen; S.L. Chan has participated as an advisor for AstraZeneca; J. Knox has received a grant for an investigator-initiated study from Merck; and has received fees for consulting from Merck, Ipsen, and Roche; B. Daniele has received fees from Ipsen, Eisai, Eli Lilly, AstraZeneca, Sanofi, Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, Bayer, Roche, and Amgen; and has received non-financial support from Ipsen, Bristol Myers Squibb; T. Yau has received fees for serving in an advisory capacity or as a consultant and has received honoraria from Bristol Myers Squibb, Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, Exelixis, Ipsen, Eisai, AstraZeneca, Bayer, Novartis, EMD Serono, AbbVie, Pfizer, Eli Lilly, Sirtex, SillaJen, Taiho, OrigiMed, New Beta Innovation, Sirtex, and H3 Biomedicine; E.B. Gurary and A. B. Siegel are employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA; A. Wang is an employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA and has stock in Merck & Co., Inc., Kenilworth, NJ, USA; A-L Cheng has received fees for consulting from AstraZeneca, Bristol Myers Squibb, Eisai, Merck Serono, Novartis, Ono Pharmaceutical, Exelixis, Nucleix, Ipsen Innovation, Bayer Healthcare, Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, Roche/Genentech, BeiGene, CSR Pharma Group, F Hoffmann-La Roche, and IQVIA; has received travel support from Roche/Genentech, IQVIA, and Bayer Yakuhin; and has received speaker fees from Eisai, Novartis, Ono Pharmaceutical, Bayer Yakuhin, and Amgen Taiwan; A.X. Zhu has received fees for consulting from Merck, Eli Lilly, Bayer, Sanofi, Eisai, Exelixis, and Roche., (Copyright © 2022 Masatoshi Kudo, Richard S. Finn, Julien Edeline, Stéphane Cattan, Sadahisa Ogasawara, Daniel H. Palmer, Chris Verslype, Vittorina Zagonel, Laetitia Fartoux, Arndt Vogel, Debashis Sarker, Gontran Verset, Stephen L. Chan, Jennifer Knox, Bruno Daniele, Thomas Yau, Ellen B. Gurary, Abby B. Siegel, Anran Wang, Ann-Lii Cheng, Andrew X. Zhu, KEYNOTE-224 Investigators. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
25. Regional variations in hepatocellular carcinoma incidence, routes to diagnosis, treatment and survival in England.
- Author
-
Burton A, Balachandrakumar VK, Driver RJ, Tataru D, Paley L, Marshall A, Alexander G, Rowe IA, Palmer DH, and Cross TJS
- Subjects
- Age of Onset, Aged, Algorithms, Carcinoma, Hepatocellular mortality, Disease Management, England epidemiology, Female, Humans, Incidence, Liver Neoplasms mortality, Male, Middle Aged, Sex Characteristics, Socioeconomic Factors, Survival Analysis, Carcinoma, Hepatocellular epidemiology, Liver Neoplasms epidemiology
- Abstract
Background: Hepatocellular carcinoma (HCC) incidence, management and survival across England were examined to determine if geographical inequalities exist., Method: 15,468 HCC cases diagnosed 2010-2016 were included. Age-standardised incidence rates, net survival and proportions receiving potentially curative treatment and presenting through each route to diagnosis adjusted for age at diagnosis, sex and area-based deprivation quintile, were calculated overall and by Cancer Alliance., Results: HCC incidence rates increased in men from 6.2 per 100,000 in 2010 to 8.8 in 2016, and in women from 1.5 to 2.2. The highest incidence rates, found in parts of the North of England and London, were nearly double the lowest. The adjusted proportion presenting as an emergency ranged 27-41% across Cancer Alliances. Odds increased with increasing deprivation quintile and age. Only one in five patients received potentially curative treatment (range 15-28%) and odds decreased with increasing deprivation and age. One-year survival in 2013-2016 ranged 38-53%., Conclusion: This population-based, nationwide analysis demonstrates clear differences in HCC incidence, management and survival across England. It highlights socioeconomic-associated variation and the need for improvement in early diagnosis and curative treatment of HCC. This research should assist policymakers, service providers and clinicians to identify regions where additional training, services and resources would be best directed., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
26. Correction to: Efficacy and Safety Results from a Phase 2, Randomized, Double Blind Study of Enzalutamide Versus Placebo in Advanced Hepatocellular Carcinoma.
- Author
-
Ryoo BY, Palmer DH, Park SR, Rimassa L, Sarker D, Daniele B, Steinberg J, López B, and Lim HY
- Published
- 2022
- Full Text
- View/download PDF
27. hENT1 Predicts Benefit from Gemcitabine in Pancreatic Cancer but Only with Low CDA mRNA.
- Author
-
Aughton K, Elander NO, Evans A, Jackson R, Campbell F, Costello E, Halloran CM, Mackey JR, Scarfe AG, Valle JW, Carter R, Cunningham D, Tebbutt NC, Goldstein D, Shannon J, Glimelius B, Hackert T, Charnley RM, Anthoney A, Lerch MM, Mayerle J, Palmer DH, Büchler MW, Ghaneh P, Neoptolemos JP, and Greenhalf W
- Abstract
Gemcitabine or 5-fluorouracil (5-FU) based treatments can be selected for pancreatic cancer. Equilibrative nucleoside transporter 1 (hENT1) predicts adjuvant gemcitabine treatment benefit over 5-FU. Cytidine deaminase (CDA), inside or outside of the cancer cell, will deaminate gemcitabine, altering transporter affinity. ESPAC-3(v2) was a pancreatic cancer trial comparing adjuvant gemcitabine and 5-FU. Tissue microarray sections underwent in situ hybridization and immunohistochemistry. Analysis of both CDA and hENT1 was possible with 277 patients. The transcript did not correlate with protein levels for either marker. High hENT1 protein was prognostic with gemcitabine; median overall survival was 26.0 v 16.8 months ( p = 0.006). Low CDA transcript was prognostic regardless of arm; 24.8 v 21.2 months with gemcitabine ( p = 0.02) and 26.4 v 14.6 months with 5-FU ( p = 0.02). Patients with low hENT1 protein did better with 5-FU, but only if the CDA transcript was low (median survival of 5-FU v gemcitabine; 29.3 v 18.3 months, compared with 14.2 v 14.6 with high CDA). CDA mRNA is an independent prognostic biomarker. When added to hENT1 protein status, it may also provide treatment-specific predictive information and, within the frame of a personalized treatment strategy, guide to either gemcitabine or 5FU for the individual patient.
- Published
- 2021
- Full Text
- View/download PDF
28. Biopsy for advanced hepatocellular carcinoma: results of a multicentre UK audit.
- Author
-
Childs A, Zakeri N, Ma YT, O'Rourke J, Ross P, Hashem E, Hubner RA, Hockenhull K, Iwuji C, Khan S, Palmer DH, Connor J, Swinson D, Darby S, Braconi C, Roques T, Yu D, Luong TV, and Meyer T
- Subjects
- Adult, Aged, Aged, 80 and over, Biopsy statistics & numerical data, Carcinoma, Hepatocellular drug therapy, Cholangiocarcinoma, Humans, Liver Neoplasms drug therapy, Magnetic Resonance Imaging statistics & numerical data, Male, Middle Aged, Predictive Value of Tests, Prospective Studies, Tomography, X-Ray Computed statistics & numerical data, United Kingdom, Young Adult, Carcinoma, Hepatocellular diagnostic imaging, Carcinoma, Hepatocellular pathology, Liver Neoplasms diagnostic imaging, Liver Neoplasms pathology
- Abstract
Background: Advanced hepatocellular carcinoma (HCC) is commonly diagnosed using non-invasive radiological criteria (NIRC) defined by the European Association for the Study of the Liver or the American Association for the Study of Liver Diseases. In 2017, The National Institute for Clinical Excellence mandated histological confirmation of disease to authorise the use of sorafenib in the UK., Methods: This was a prospective multicentre audit in which patients suitable for sorafenib were identified at multidisciplinary meetings. The primary analysis cohort (PAC) was defined by the presence of Child-Pugh class A liver disease and performance status 0-2. Clinical, radiological and histological data were reported locally and collected on a standardised case report form., Results: Eleven centres reported 418 cases, of which 361 comprised the PAC. Overall, 76% had chronic liver disease and 66% were cirrhotic. The diagnostic imaging was computed tomography in 71%, magnetic resonance imaging in 27% and 2% had both. Pre-existing histology was available in 45 patients and 270 underwent a new biopsy, which confirmed HCC in 93.4%. Alternative histological diagnoses included cholangiocarcinoma (CC) and combined HCC-CC. In cirrhotic patients, NIRC criteria had a sensitivity of 65.4% and a positive predictive value of 91.4% to detect HCC. Two patients (0.7%) experienced mild post-biopsy bleeding., Conclusion: The diagnostic biopsy is safe and feasible for most patients eligible for systemic therapy., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
29. Efficacy and Safety Results from a Phase 2, Randomized, Double-Blind Study of Enzalutamide Versus Placebo in Advanced Hepatocellular Carcinoma.
- Author
-
Ryoo BY, Palmer DH, Park SR, Rimassa L, Debashis Sarker, Daniele B, Steinberg J, López B, and Lim HY
- Subjects
- Benzamides, Double-Blind Method, Humans, Nitriles, Phenylthiohydantoin, Sorafenib, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy
- Abstract
Background and Objective: Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related mortality worldwide. Despite recent advances, more effective therapeutic options for patients with advanced HCC are still required. The aim of this Phase 2, multicenter, multinational, randomized, double-blind, placebo-controlled study (NCT02528643) was to investigate the potential benefit of enzalutamide in the treatment of patients with advanced HCC., Methods: Patients aged ≥ 18 years diagnosed with advanced HCC (Barcelona Clinic Liver Cancer stage B or C and Child-Pugh class A at screening who had progressed on, or were intolerant to, sorafenib or other anti-vascular endothelial growth factor therapies) were randomized 2:1 to receive either enzalutamide 160 mg daily or placebo. The primary endpoint was overall survival (OS); secondary endpoints included progression-free survival (PFS) and safety., Results: In total, 165 patients were randomized to enzalutamide (n = 110) or placebo (n = 55). The hazard ratio (HR) (95% confidence interval [CI]) for OS was 1.15 (0.774-1.696) and median OS was 7.8 months and 7.7 months for enzalutamide and placebo, respectively. The HR (95% CI) for PFS was 1.04 (0.732-1.474) and median PFS was 2.2 months and 1.9 months for enzalutamide and placebo, respectively. The overall frequency of treatment-emergent adverse events (TEAEs) was broadly similar between the groups: 105 (98.1%) enzalutamide patients experienced ≥1 TEAEs compared with 49 (89.1%) placebo patients., Conclusions: The results of this study indicate that enzalutamide does not provide a benefit in patients with advanced HCC. No unexpected safety findings were observed in the trial. CLINICALTRIALS., Gov Identifier: NCT02528643., (© 2021. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
- Published
- 2021
- Full Text
- View/download PDF
30. Baseline Liver Function and Subsequent Outcomes in the Phase 3 REFLECT Study of Patients with Unresectable Hepatocellular Carcinoma.
- Author
-
Vogel A, Frenette C, Sung M, Daniele B, Baron A, Chan SL, Blanc JF, Tamai T, Ren M, Lim HJ, Palmer DH, Takami Y, and Kudo M
- Abstract
Introduction: Baseline liver function among patients starting treatment for unresectable hepatocellular carcinoma (uHCC) impacts survival and could impact efficacy outcomes and safety profiles of treatments. This post hoc analysis of the phase 3 REFLECT study examined the efficacy and safety outcomes for lenvatinib and for sorafenib in patients with uHCC, assessed by Child-Pugh score (CPS) and albumin-bilirubin (ALBI) grade., Methods: Efficacy and safety were assessed in patient cohorts from REFLECT according to study entry baseline ALBI grade and CPS., Results: Lenvatinib treatment generally provided survival benefits in all groups. Median overall survival (OS) among patients with an ALBI grade of 1 was consistently higher than among patients with an ALBI grade of 2 for both the lenvatinib and sorafenib arms (lenvatinib: 17.4 vs. 8.6 months; sorafenib: 14.6 vs. 7.7 months, respectively). Median OS among patients with a CPS of 5 was consistently higher than among patients with a CPS of 6 (lenvatinib: 15.3 vs. 9.4 months; sorafenib: 14.2 vs. 7.9 months, respectively). Progression-free survival and objective response rates for these ALBI grades and CPS demonstrated similar patterns. Among patients who received lenvatinib and experienced a treatment-related treatment-emergent adverse event leading to withdrawal, 6.6% had an ALBI grade of 1, while 13.3% had an ALBI grade of 2, and 7.9% had a CPS of 5, while 12.1% had a CPS of 6., Conclusions: Better liver function at baseline, as measured by ALBI grade or CPS, may be prognostic for better survival outcomes in patients with uHCC undergoing treatment with lenvatinib or sorafenib., Competing Interests: A.V. has provided a speaker, consultancy, and advisory role for Roche, Bayer, Sanofi, BMS, Lilly, Novartis, Eisai, AstraZeneca, Merck, Incyte, Medac, Ipsen, Servier, PierreFabre, MSD, BTG, and Janssen. C.F. serves on speakers' bureaus and advisory boards for Bayer and Eisai. M.S. has received honoraria from advisory board participation from Bayer, Eisai, Exelixis, and Genentech. B.D. has received personal fees and nonfinancial support for consultation from Ipsen, Sanofi, and Bayer; and personal fees from Roche, Eisai, Eli Lilly, AstraZeneca, MSD, and Incyte. A.B. has served on speakers' bureau for Bristol Myers Squibb, Merck, Lilly, Amgen, Eisai, Johnson & Johnson, and AbbVie. S.L.C. has acted as an advisor to AstraZeneca, Eisai, Ipsen, and MSD. J.F.B. has received honoraria from Bayer, Ipsen, Eisai, Roche, BMS, AstraZeneca, and Eli Lilly. T.T. is an employee of and has stock/other ownership with Eisai. M.R. is an employee of Eisai. H.J.L. has nothing to disclose. D.H.P. has received honoraria from Eisai and research funding and honoraria from Bayer, BMS, Roche, AstraZeneca, Sirtex, and BTG. Y.T. has nothing to disclose. M.K. has received honoraria from Merck Sharp & Dohme, Eisai, Bayer, Lilly Japan, EA Pharma, and Bristol Myers Squibb Japan; served in a consulting/advisory role for Merck Sharp & Dohme, Eisai, Ono Pharmaceuticals, Bristol Myers Squibb, and Roche; and received research funding (inst) from Otsuka Pharmaceutical, Taiho Pharmaceutical, AbbVie, Takeda Pharmaceuticals, Eisai, Gilead Sciences, EA Pharma, and Dainippon Sumitomo Pharma. M.K. is the Editor-in-Chief of Liver Cancer., (Copyright © 2021 by S. Karger AG, Basel.)
- Published
- 2021
- Full Text
- View/download PDF
31. Haematological malignancy and nosocomial transmission are associated with an increased risk of death from COVID-19: results of a multi-center UK cohort.
- Author
-
Bhogal T, Khan UT, Lee R, Stockdale A, Hesford C, Potti-Dhananjaya V, Jathanna A, Rahman S, Tivey A, Shotton R, Sundar R, Valerio C, Norouzi A, Walker P, Suckling R, Armstrong A, Brearton G, Pettitt A, Kalakonda N, Palmer DH, Jackson R, Turtle L, and Palmieri C
- Subjects
- COVID-19 Testing, Female, Humans, Pandemics, Retrospective Studies, Risk Factors, SARS-CoV-2, United Kingdom epidemiology, COVID-19, Cross Infection epidemiology, Hematologic Neoplasms epidemiology
- Abstract
The COVID-19 pandemic has been a disruptive event for cancer patients, especially those with haematological malignancies (HM). They may experience a more severe clinical course due to impaired immune responses. This multi-center retrospective UK audit identified cancer patients who had SARS-CoV-2 infection between 1 March and 10 June 2020 and collected data pertaining to cancer history, COVID-19 presentation and outcomes. In total, 179 patients were identified with a median age of 72 (IQR 61, 81) and follow-up of 44 days (IQR 42, 45). Forty-one percent were female and the overall mortality was 37%. Twenty-nine percent had HM and of these, those treated with chemotherapy in the preceding 28 days to COVID-19 diagnosis had worse outcome compared with solid malignancy (SM): 62% versus 19% died [HR 8.33 (95% CI, 2.56-25), p < 0.001]. Definite or probable nosocomial SARS-CoV-2 transmission accounted for 16% of cases and was associated with increased risk of death (HR 2.47, 95% CI 1.43-4.29, p = 0.001). Patients with haematological malignancies and those who acquire nosocomial transmission are at increased risk of death. Therefore, there is an urgent need to reassess shielding advice, reinforce stringent infection control, and ensure regular patient and staff testing to prevent nosocomial transmission.
- Published
- 2021
- Full Text
- View/download PDF
32. Second-line FOLFOX chemotherapy for advanced biliary tract cancer - Authors' reply.
- Author
-
Lamarca A, Palmer DH, Wasan HS, Ross PJ, Ma YT, Arora A, Falk S, Gillmore R, Wadsley J, Patel K, Anthoney A, Maraveyas A, Iveson T, Waters JS, Hobbs C, Barber S, Ryder WD, Ramage J, Davies LM, Bridgewater JA, and Valle JW
- Subjects
- Humans, Leucovorin adverse effects, Bile Duct Neoplasms, Biliary Tract Neoplasms drug therapy
- Abstract
Competing Interests: The authors' declaration of interests remain the same as those declared in the original Article.
- Published
- 2021
- Full Text
- View/download PDF
33. The effects of neoadjuvant chemoradiotherapy and an in-hospital exercise training programme on physical fitness and quality of life in locally advanced rectal cancer patients: a randomised controlled trial (The EMPOWER Trial).
- Author
-
Loughney L, West MA, Moyses H, Bates A, Kemp GJ, Hawkins L, Varkonyi-Sepp J, Burke S, Barben CP, Calverley PM, Cox T, Palmer DH, Mythen MG, Grocott MPW, and Jack S
- Abstract
Background: The EMPOWER trial aimed to assess the effects of a 9-week exercise prehabilitation programme on physical fitness compared with a usual care control group. Secondary aims were to investigate the effect of (1) the exercise prehabilitation programme on psychological health; and (2) neoadjuvant chemoradiotherapy (NCRT) on physical fitness and psychological health., Methods: Between October 2013 and December 2016, adults with locally advanced rectal cancer undergoing standardised NCRT and surgery were recruited to a multi-centre trial. Patients underwent cardiopulmonary exercise testing (CPET) and completed HRQoL questionnaires (EORTC-QLQ-C30 and EQ-5D-5L) pre-NCRT and post-NCRT (week 0/baseline). At week 0, patients were randomised to exercise prehabilitation or usual care (no intervention). CPET and HRQoL questionnaires were assessed at week 0, 3, 6 and 9, whilst semi-structured interviews were assessed at week 0 and week 9. Changes in oxygen uptake at anaerobic threshold (VO
2 at AT (ml kg-1 min-1 )) between groups were compared using linear mixed modelling., Results: Thirty-eight patients were recruited, mean age 64 (10.4) years. Of the 38 patients, 33 were randomised: 16 to usual care and 17 to exercise prehabilitation (26 males and 7 females). Exercise prehabilitation significantly improved VO2 at AT at week 9 compared to the usual care. The change from baseline to week 9, when adjusted for baseline, between the randomised groups was + 2.9 ml kg-1 min-1 ; (95% CI 0.8 to 5.1), p = 0.011., Conclusion: A 9-week exercise prehabilitation programme significantly improved fitness following NCRT. These findings have informed the WesFit trial (NCT03509428) which is investigating the effects of community-based multimodal prehabilitation before cancer surgery., Trial Registration: ClinicalTrials.gov NCT01914068 . Registered 1 August 2013.- Published
- 2021
- Full Text
- View/download PDF
34. Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): a phase 3, open-label, randomised, controlled trial.
- Author
-
Lamarca A, Palmer DH, Wasan HS, Ross PJ, Ma YT, Arora A, Falk S, Gillmore R, Wadsley J, Patel K, Anthoney A, Maraveyas A, Iveson T, Waters JS, Hobbs C, Barber S, Ryder WD, Ramage J, Davies LM, Bridgewater JA, and Valle JW
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Biliary Tract Neoplasms mortality, Female, Fluorouracil adverse effects, Fluorouracil therapeutic use, Humans, Leucovorin adverse effects, Leucovorin therapeutic use, Male, Middle Aged, Organoplatinum Compounds adverse effects, Organoplatinum Compounds therapeutic use, Prospective Studies, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biliary Tract Neoplasms drug therapy
- Abstract
Background: Advanced biliary tract cancer has a poor prognosis. Cisplatin and gemcitabine is the standard first-line chemotherapy regimen, but no robust evidence is available for second-line chemotherapy. The aim of this study was to determine the benefit derived from second-line FOLFOX (folinic acid, fluorouracil, and oxaliplatin) chemotherapy in advanced biliary tract cancer., Methods: The ABC-06 clinical trial was a phase 3, open-label, randomised trial done in 20 sites with expertise in managing biliary tract cancer across the UK. Adult patients (aged ≥18 years) who had histologically or cytologically verified locally advanced or metastatic biliary tract cancer (including cholangiocarcinoma and gallbladder or ampullary carcinoma) with documented radiological disease progression to first-line cisplatin and gemcitabine chemotherapy and an Eastern Cooperative Oncology Group performance status of 0-1 were randomly assigned (1:1) centrally to active symptom control (ASC) and FOLFOX or ASC alone. FOLFOX chemotherapy was administered intravenously every 2 weeks for a maximum of 12 cycles (oxaliplatin 85 mg/m
2 , L-folinic acid 175 mg [or folinic acid 350 mg], fluorouracil 400 mg/m2 [bolus], and fluorouracil 2400 mg/m2 as a 46-h continuous intravenous infusion). Randomisation was done following a minimisation algorithm using platinum sensitivity, serum albumin concentration, and stage as stratification factors. The primary endpoint was overall survival, assessed in the intention-to-treat population. Safety was also assessed in the intention-to-treat population. The study is complete and the final results are reported. This trial is registered with ClinicalTrials.gov, NCT01926236, and EudraCT, 2013-001812-30., Findings: Between March 27, 2014, and Jan 4, 2018, 162 patients were enrolled and randomly assigned to ASC plus FOLFOX (n=81) or ASC alone (n=81). Median follow-up was 21·7 months (IQR 17·2-30·8). Overall survival was significantly longer in the ASC plus FOLFOX group than in the ASC alone group, with a median overall survival of 6·2 months (95% CI 5·4-7·6) in the ASC plus FOLFOX group versus 5·3 months (4·1-5·8) in the ASC alone group (adjusted hazard ratio 0·69 [95% CI 0·50-0·97]; p=0·031). The overall survival rate in the ASC alone group was 35·5% (95% CI 25·2-46·0) at 6 months and 11·4% (5·6-19·5) at 12 months, compared with 50·6% (39·3-60·9) at 6 months and 25·9% (17·0-35·8) at 12 months in the ASC plus FOLFOX group. Grade 3-5 adverse events were reported in 42 (52%) of 81 patients in the ASC alone group and 56 (69%) of 81 patients in the ASC plus FOLFOX group, including three chemotherapy-related deaths (one each due to infection, acute kidney injury, and febrile neutropenia). The most frequently reported grade 3-5 FOLFOX-related adverse events were neutropenia (ten [12%] patients), fatigue or lethargy (nine [11%] patients), and infection (eight [10%] patients)., Interpretation: The addition of FOLFOX to ASC improved median overall survival in patients with advanced biliary tract cancer after progression on cisplatin and gemcitabine, with a clinically meaningful increase in 6-month and 12-month overall survival rates. To our knowledge, this trial is the first prospective, randomised study providing reliable, high-quality evidence to allow an informed discussion with patients of the potential benefits and risks from second-line FOLFOX chemotherapy in advanced biliary tract cancer. Based on these findings, FOLFOX should become standard-of-care chemotherapy in second-line treatment for advanced biliary tract cancer and the reference regimen for further clinical trials., Funding: Cancer Research UK, StandUpToCancer, AMMF (The UK Cholangiocarcinoma Charity), and The Christie Charity, with additional funding from The Cholangiocarcinoma Foundation and the Conquer Cancer Foundation Young Investigator Award for translational research., Competing Interests: Declaration of interests AL has received travel and educational support from Ipsen, Pfizer, Bayer, AAA, SirtEx, Novartis, Mylan, and Delcath; speaker honoraria from Merck, Pfizer, Ipsen, and Incyte; advisory honoraria from EISAI, Nutricia Ipsen, QED, and Roche, outside of the submitted work. AL is also a member of the Knowledge Network and NETConnect Initiatives funded by Ipsen. DHP declares research grants from AstraZeneca, Bayer, Bristol-Myers Squibb, Nucana, and Sirtex; and consulting or advisory roles for AstraZeneca, Bayer, Bristol-Myers Squibb, Eisai, Merck Sharp & Dohme, Servier, and Roche, outside of the submitted work. HSW has participated in advisory boards for Incyte, Bayer, Roche/Genentech/Foundation Medicine, SIRTEX Medical, Celgene, and Zymeworks; has been a NICE expert for Bayer (uncompensated); and reports consultancies for ONCOSIL; speaker-related honoraria and travel support from BTG/Biocompatibles, Merck KGaA, and BMS; participation in trials steering committee for Pfizer and Zymeworks; creation of educational materials for AstraZeneca; and receipt of research funding from Sirtex, outside of the submitted work. PJR declares research grants from Sanofi and Bayer; consulting or advisory roles for Bayer, Bristol-Myers Squibb, Eisai, Sirtex, and Roche; speaker fees from Amgen, Roche, and Servier; and travel grants from Bayer, Roche, and Servier, outside of the submitted work. YTM declares speaker honoraria and advisory roles for Bayer, Eisai, and Roche, outside of the submitted work. JW declares research grants from AstraZeneca and Sanofi-Genzyme and consulting or advisory roles for Lilly, AstraZeneca, Sanofi Genyme, Eisai, AAA, Roche, Novartis, and Ipsen, outside of the submitted work. AM declares research grants from Pfizer, Bristol-Myers Squibb, and Bayer; speaker fees from Bristol-Myers Squibb, Bayer Ipsen, EUSA Pharma, Daiichi Sankyo, and Pfizer; and advisory roles for Bayer, Bristol-Myers Squibb, Leo, Pfizer, Merck, and Ipsen, outside of the submitted work. JSW has received educational support for travel and conference attendance from Mylan and Ipsen, and speaker honoraria from Mylan, outside of the submitted work. JR declares research grants, speaker fees, and advisory roles for Ipsen, Novartis, and AAA, outside of the submitted work. JAB declares consulting or advisory roles for Merck Serono, Servier, Roche, Bayer, AstraZeneca, Incyte, and Basilea; and travel support from MSD Oncology, Merck Serono, Servier, and Bristol-Myers Squibb, outside of the submitted work. JWV declares consulting or advisory roles for Agios, AstraZeneca, Delcath Systems, Keocyt, Genoscience Pharma, Incyte, Ipsen, Merck, Mundipharma EDO, Novartis, PCI Biotech, Pfizer, Pieris Pharmaceuticals, QED, and Wren Laboratories; speakers' bureau for Imaging Equipment, Ipsen, Novartis, and Nucana; and travel grants from Celgene and Nucana, outside of the submitted work. AAr, SF, RG, KP, AAn, TI, CH, SB, WDR, and LMD declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
35. A Phase Ib Study of NUC-1031 in Combination with Cisplatin for the First-Line Treatment of Patients with Advanced Biliary Tract Cancer (ABC-08).
- Author
-
McNamara MG, Bridgewater J, Palmer DH, Faluyi O, Wasan H, Patel A, Ryder WD, Barber S, Gnanaranjan C, Ghazaly E, Evans TRJ, and Valle JW
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Bile Ducts, Intrahepatic, Cisplatin therapeutic use, Cytidine Monophosphate analogs & derivatives, Disease-Free Survival, Humans, Male, Middle Aged, Treatment Outcome, Bile Duct Neoplasms drug therapy, Biliary Tract Neoplasms drug therapy
- Abstract
Background: Cisplatin/gemcitabine is standard first-line treatment for patients with advanced biliary tract cancer (ABC). NUC-1031 (phosphoramidate transformation of gemcitabine) is designed to enhance efficacy by maximizing intratumoral active metabolites., Methods: Patients with untreated ABC, Eastern Cooperative Oncology Group performance status 0-1 received NUC-1031 (625 or 725 mg/m
2 ) and cisplatin (25 mg/m2 ) on days 1 and 8, every 21 days. Primary objectives were safety and maximum tolerated dose; secondary objectives were objective response rate (ORR), pharmacokinetics, progression-free survival (PFS), and overall survival (OS)., Results: Twenty-one patients (median age 61 years, n = 13 male; 17 cholangiocarcinoma, 2 ampullary, and 2 gallbladder cancer) received NUC-1031 625 mg/m2 (n = 8 and expansion n = 7; median six cycles) or 725 mg/m2 (n = 6; median 7.5 cycles). Treatment was well tolerated; most common treatment-emergent grade 3-4 adverse events occurring in more than one patient with 625 mg/m2 NUC-1031 were increased gamma-glutamyl transferase (GGT), 40%; alanine aminotransferase, 20%; bilirubin, 13%; neutropenia, 27%; decreased white cell count, 20%; thrombocytopenia, 13%; nausea, 13%; diarrhea, 13%; fatigue, 13%; and thrombus, 20% and with 725 mg/m2 , increased GGT, 67%, and fatigue, 33%. NUC-1031 725 mg/m2 was selected as the recommended dose with cisplatin in ABC. ORR was 33% (one complete response, six partial responses), DCR was 76%, median PFS was 7.2 months (95% confidence interval [CI], 4.3-10.1), and median OS was 9.6 months (95% CI, 6.7-13.1). The median estimates of area under the plasma concentration-time curve from time 0 to last measurable time and maximum concentration were highest for NUC-1031 (218-324 μg•h/mL and 309-889 μg/mL, respectively) and lowest for di-fluoro-deoxycytidine (0.47-1.56 μg•h/mL and 0.284-0.522 μg/mL, respectively)., Conclusion: This is the first study reporting on the combination of NUC-1031 with cisplatin in ABC and demonstrated a favorable safety profile; 725 mg/m2 NUC-1031 in combination with cisplatin is undergoing phase III trial evaluation in ABC. (ClinicalTrials.gov ID: NCT02351765; EudraCT ID: 2015-000100-26)., Implications for Practice: The prognosis for patients with advanced biliary tract cancer (ABC) is approximately 1 year, and new treatment options are required. The cisplatin/gemcitabine combination is standard first-line treatment for patients with ABC. NUC-1031 is a phosphoramidate transformation of gemcitabine and is designed to enhance efficacy by maximizing intratumoral active metabolites. This phase Ib study (ABC-08) demonstrated a favorable safety profile of NUC-1031 in combination with cisplatin for the first-line treatment of patients with ABC, and 725 mg/m2 NUC-1031 was recommended in combination with cisplatin for phase III trial evaluation; the NuTide:121 global randomized study is currently enrolling., (© 2020 AlphaMed Press.)- Published
- 2021
- Full Text
- View/download PDF
36. Reply to Comment on "The UK consensus position on the treatment of pancreatic cancer during the COVID-19 pandemic".
- Author
-
Jones CM, Radhakrishna G, Aitken K, Bridgewater J, Corrie P, Eatock M, Goody R, Ghaneh P, Good J, Grose D, Holyoake D, Hunt A, Jamieson NB, Palmer DH, Soonawalla Z, Valle JW, Hawkins MA, and Mukherjee S
- Subjects
- Consensus, Humans, Pandemics, SARS-CoV-2, United Kingdom epidemiology, COVID-19, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms epidemiology
- Published
- 2021
- Full Text
- View/download PDF
37. Sex-Specific Selection Drives the Evolution of Alternative Splicing in Birds.
- Author
-
Rogers TF, Palmer DH, and Wright AE
- Subjects
- Animals, Birds metabolism, Female, Male, Phenotype, Protein Isoforms metabolism, Alternative Splicing, Biological Evolution, Birds genetics, Sex Characteristics, Sexual Selection
- Abstract
Males and females of the same species share the majority of their genomes, yet they are frequently exposed to conflicting selection pressures. Gene regulation is widely assumed to resolve these conflicting sex-specific selection pressures, and although there has been considerable focus on elucidating the role of gene expression level in sex-specific adaptation, other regulatory mechanisms have been overlooked. Alternative splicing enables different transcripts to be generated from the same gene, meaning that exons which have sex-specific beneficial effects can in theory be retained in the gene product, whereas exons with detrimental effects can be skipped. However, at present, little is known about how sex-specific selection acts on broad patterns of alternative splicing. Here, we investigate alternative splicing across males and females of multiple bird species. We identify hundreds of genes that have sex-specific patterns of splicing and establish that sex differences in splicing are correlated with phenotypic sex differences. Additionally, we find that alternatively spliced genes have evolved rapidly as a result of sex-specific selection and suggest that sex differences in splicing offer another route to sex-specific adaptation when gene expression level changes are limited by functional constraints. Overall, our results shed light on how a diverse transcriptional framework can give rise to the evolution of phenotypic sexual dimorphism., (© The Author(s) 2020. Published by Oxford University Press on behalf of the Society for Molecular Biology and Evolution.)
- Published
- 2021
- Full Text
- View/download PDF
38. Effect of ramucirumab on ALBI grade in patients with advanced HCC: Results from REACH and REACH-2.
- Author
-
Kudo M, Galle PR, Brandi G, Kang YK, Yen CJ, Finn RS, Llovet JM, Assenat E, Merle P, Chan SL, Palmer DH, Ikeda M, Yamashita T, Vogel A, Huang YH, Abada PB, Yoshikawa R, Shinozaki K, Wang C, Widau RC, and Zhu AX
- Abstract
Background & Aims: The albumin-bilirubin (ALBI) grade/score is derived from a validated nomogram to objectively assess prognosis and liver function in patients with hepatocellular carcinoma (HCC). In this post hoc analysis, we assessed prognosis in terms of survival by baseline ALBI grade and monitored liver function during treatment with ramucirumab or placebo using the ALBI score in patients with advanced HCC., Methods: Patients with advanced HCC, Child-Pugh class A with prior sorafenib treatment were randomised in REACH trials to receive ramucirumab 8 mg/kg or placebo every 2 weeks. Data were analysed by trial and as a meta-analysis of individual patient-level data (pooled population) from REACH (alpha-fetoprotein ≥400 ng/ml) and REACH-2. Patients from REACH with Child-Pugh class B were analysed as a separate cohort. The ALBI grades and scores were calculated at baseline and before each treatment cycle., Results: Baseline characteristics by ALBI grade were balanced between treatment arms among patients in the pooled population (ALBI-1, n = 231; ALBI-2, n = 296; ALBI-3, n = 7). Baseline ALBI grade was prognostic for overall survival (OS; ALBI grade 2 vs. 1; hazard ratio [HR]: 1.38 [1.13-1.69]), after adjusting for other significant prognostic factors. Mean ALBI scores remained stable in both treatment arms compared with baseline and were unaffected by baseline ALBI grade, macrovascular invasion, tumour response, geographical region, or prior locoregional therapy. Baseline ALBI grades 2 and 3 were associated with increased incidence of liver-specific adverse events and discontinuation rates in both treatments. Ramucirumab improved OS in patients with baseline ALBI grade 1 (HR 0.605 [0.445-0.824]) and ALBI grade 2 (HR 0.814 [0.630-1.051])., Conclusions: Compared with placebo, ramucirumab did not negatively impact liver function and improved survival irrespective of baseline ALBI grade., Lay Summary: Hepatocellular carcinoma is the third leading cause of cancer-related death worldwide. Prognosis is affected by many clinical factors including liver function both before and during anticancer treatment. Here we have used a validated approach to assess liver function using 2 laboratory parameters, serum albumin and bilirubin (ALBI), both before and during treatment with ramucirumab in 2 phase III placebo-controlled studies. We confirm the practicality of using this more simplistic approach in assessing liver function prior to and during anticancer therapy, and demonstrate ramucirumab did not impair liver function when compared with placebo., Competing Interests: M.K.: conflict with Daiichi Sankyo, Otsuka, Taiho, Astellas Pharma, Chugai, AbbVie, BMS, EA Pharma, Takeda, Gilead, Eisai, Ono, Bayer, MSD, BMS, and Eli Lilly and Company. P.G.: conflict with Bayer, BMS, MSD, Merck, SIRTEX, AstraZeneca, Sillajen, Eli Lilly and Company, Ipsen, Roche, and Eisai. G.B.: declares no conflicts of interest that pertain to this work. Y-K.K.: conflict with ONO, BMS, Eli Lilly and Company, Roche, Daehwa, and Taiho. C-J. Yen: declares no conflicts of interest that pertain to this work. R.S.F.: conflict with Astra Zeneca, Bayer, BMS, Eli Lilly and Company, Pfizer, Merck, Novartis, Roche/Genentech, and Eisai. J.M.L.: conflict with Bayer, Eisai Inc, BMS, IPSEN, Eli Lilly and Company, Celsion Corp, Elelixis, Merck, Glycotest, Can-Fite, Blueprint, Incyte, Navigant, Leerink Swann LLC, Midatech Ltd, Fortress Biotech Inc, and Spring Bank Pharmaceuticals. E.A.: received honoraria from IPSEN, Bayer, Terasphere, Sirtex, Sanofi, Novartis, and Servier; served as an advisor and consultant for IPSEN, Bayer, Terasphere, Sirtex, Sanofi, Novartis, and Servier; and reports receipt of research grant and funding from Eli Lilly and Company. P.M.: conflict with Bayer, Ipsen/Exelixis, BMS, Onxeo, MSD, AstraZeneca, Eisai, and Roche. S.L.C.: conflict with Eisai, Merck, and AstraZeneca. D.H.P.: conflict with Bayer, Eisai, BMS, and Sirtex. M.I.: conflict with Bayer, Bristol-Myers Squibb, Eisai, Sumitomo Dainippon, EA Pharma, Takeda Pharmaceutical, AstraZeneca, Chugai, Merck Serono, Gilead, ASLAN, Daiichi Sankyo, Novartis, Teijin Pharma, Kyowa Hakko Kirin, NanoCarrier, Shire, Yakult, Taiho, Baxalta, Nobelpharma, Otsuka, Ono, MSD, J-Pharma, Mylan, NIHON SERVIER, and Eli Lilly and Company. T.Y.: received personal fees from Eli Lilly and Eisai. A.V.: conflict with Eli Lilly and Company, EISAI, Roche, BMS, MSD, Bayer, BTG, AstraZeneca, Ipsen, Novartis; Y-H. Huang: Eisai, BMS, Merck, AstraZeneca, Bayer, Gilead, Abbvie, Roche, Eli Lilly, and IPSEN. P.B.A.: employed by Eli Lilly and Company, stocks held in Eli Lilly and Company. R.Y.: Eli Lilly Japan KK, stocks held in Eli Lilly and Company. K.S.: Eli Lilly Japan KK. C.W.: employed by Eli Lilly and Company, stocks held in Eli Lilly and Company. R.C.W.: employed by Eli Lilly and Company, stocks held in Eli Lilly and Company. A.X.Z.: conflict with Eisai, BMS, Merck, Novartis, Sanofi, AstraZeneca, Bayer, Roche, Eli Lilly and Company, and Exelixis. Please refer to the accompanying ICMJE disclosure forms for further details., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
39. Considerations for the treatment of pancreatic cancer during the COVID-19 pandemic: the UK consensus position.
- Author
-
Jones CM, Radhakrishna G, Aitken K, Bridgewater J, Corrie P, Eatock M, Goody R, Ghaneh P, Good J, Grose D, Holyoake D, Hunt A, Jamieson NB, Palmer DH, Soonawalla Z, Valle JW, Hawkins MA, and Mukherjee S
- Subjects
- COVID-19, Coronavirus Infections prevention & control, Coronavirus Infections virology, Humans, Incidence, Pandemics prevention & control, Pneumonia, Viral prevention & control, Pneumonia, Viral virology, Quarantine methods, Risk, SARS-CoV-2, United Kingdom epidemiology, Betacoronavirus, Consensus, Coronavirus Infections epidemiology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms radiotherapy, Pneumonia, Viral epidemiology, Practice Guidelines as Topic
- Abstract
The coronavirus disease 2019 (COVID-19) pandemic epicentre has moved to the USA and Europe, where it is placing unprecedented demands on healthcare resources and staff availability. These service constraints, coupled with concerns relating to an increased incidence and severity of COVID-19 among patients with cancer, should lead to re-consideration of the risk-benefit balance for standard treatment pathways. This is of particular importance to pancreatic cancer, given that standard diagnostic modalities such as endoscopy may be restricted, and that disease biology precludes significant delays in treatment. In light of this, we sought consensus from UK clinicians with an interest in pancreatic cancer for management approaches that would minimise patient risk and accommodate for healthcare service restrictions. The outcomes are described here and include recommendations for treatment prioritisation, strategies to bridge to later surgical resection in resectable disease and factors that modify the risk-benefit balance for treatment in the resectable through to the metastatic settings. Priority is given to strategies that limit hospital visits, including through the use of hypofractionated precision radiotherapy and chemoradiotherapy treatment approaches.
- Published
- 2020
- Full Text
- View/download PDF
40. MTL-CEBPA, a Small Activating RNA Therapeutic Upregulating C/EBP-α, in Patients with Advanced Liver Cancer: A First-in-Human, Multicenter, Open-Label, Phase I Trial.
- Author
-
Sarker D, Plummer R, Meyer T, Sodergren MH, Basu B, Chee CE, Huang KW, Palmer DH, Ma YT, Evans TRJ, Spalding DRC, Pai M, Sharma R, Pinato DJ, Spicer J, Hunter S, Kwatra V, Nicholls JP, Collin D, Nutbrown R, Glenny H, Fairbairn S, Reebye V, Voutila J, Dorman S, Andrikakou P, Lloyd P, Felstead S, Vasara J, Habib R, Wood C, Saetrom P, Huber HE, Blakey DC, Rossi JJ, and Habib N
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, CCAAT-Enhancer-Binding Proteins genetics, Carcinoma, Hepatocellular diagnosis, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular pathology, Dose-Response Relationship, Drug, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Infusions, Intravenous, Liposomes, Liver Neoplasms diagnosis, Liver Neoplasms genetics, Liver Neoplasms pathology, Male, Middle Aged, Nanoparticles administration & dosage, Neoplasm Staging, Oligoribonucleotides adverse effects, Oligoribonucleotides pharmacokinetics, Treatment Outcome, Tumor Microenvironment drug effects, Up-Regulation drug effects, Antineoplastic Agents administration & dosage, CCAAT-Enhancer-Binding Proteins agonists, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy, Oligoribonucleotides administration & dosage
- Abstract
Purpose: Transcription factor C/EBP-α (CCAAT/enhancer-binding protein alpha) acts as a master regulator of hepatic and myeloid functions and multiple oncogenic processes. MTL-CEBPA is a first-in-class small activating RNA oligonucleotide drug that upregulates C/EBP-α., Patients and Methods: We conducted a phase I, open-label, dose-escalation trial of MTL-CEBPA in adults with advanced hepatocellular carcinoma (HCC) with cirrhosis, or resulting from nonalcoholic steatohepatitis or with liver metastases. Patients received intravenous MTL-CEBPA once a week for 3 weeks followed by a rest period of 1 week per treatment cycle in the dose-escalation phase (3+3 design)., Results: Thirty-eight participants have been treated across six dose levels (28-160 mg/m
2 ) and three dosing schedules. Thirty-four patients were evaluable for safety endpoints at 28 days. MTL-CEBPA treatment-related adverse events were not associated with dose, and no maximum dose was reached across the three schedules evaluated. Grade 3 treatment-related adverse events occurred in nine (24%) patients. In 24 patients with HCC evaluable for efficacy, an objective tumor response was achieved in one patient [4%; partial response (PR) for over 2 years] and stable disease (SD) in 12 (50%). After discontinuation of MTL-CEBPA, seven patients were treated with tyrosine kinase inhibitors (TKIs); three patients had a complete response with one further PR and two with SD., Conclusions: MTL-CEBPA is the first saRNA in clinical trials and demonstrates an acceptable safety profile and potential synergistic efficacy with TKIs in HCC. These encouraging phase I data validate targeting of C/EBP-α and have prompted MTL-CEBPA + sorafenib combination studies in HCC., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
41. NUC-1031/cisplatin versus gemcitabine/cisplatin in untreated locally advanced/metastatic biliary tract cancer (NuTide:121).
- Author
-
McNamara MG, Goyal L, Doherty M, Springfeld C, Cosgrove D, Sjoquist KM, Park JO, Verdaguer H, Braconi C, Ross PJ, Gramont A, Zalcberg JR, Palmer DH, Valle JW, and Knox JJ
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Young Adult, Cisplatin administration & dosage, Cytidine Monophosphate administration & dosage, Cytidine Monophosphate analogs & derivatives, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Gemcitabine, Neoplasm Metastasis, Neoplasm Staging, Survival Rate, Treatment Outcome, Clinical Trials, Phase III as Topic, Multicenter Studies as Topic, Randomized Controlled Trials as Topic, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bile Ducts, Intrahepatic drug effects, Bile Ducts, Intrahepatic pathology, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms pathology, Cholangiocarcinoma drug therapy, Cholangiocarcinoma pathology
- Abstract
Gemcitabine/cisplatin is standard of care for first-line treatment of patients with advanced biliary tract cancer (aBTC); new treatments are needed. NUC-1031 is designed to overcome key cancer resistance mechanisms associated with gemcitabine. The tolerability/efficacy signal of NUC-1031/cisplatin in the Phase Ib ABC-08 study suggested that this combination may represent a more efficacious therapy than gemcitabine/cisplatin for patients with aBTC, leading to initiation of the global NuTide:121 study which will include 828 patients ≥18 years with untreated histologically/cytologically-confirmed aBTC (including cholangiocarcinoma, gallbladder or ampullary cancer); randomized (1:1) to NUC-1031 (725 mg/m
2 )/cisplatin (25 mg/m2 ) or gemcitabine (1000 mg/m2 )/cisplatin (25 mg/m2 ), on days 1/8, Q21-days. Primary objectives are overall survival and objective response rate. Secondary objectives: progression-free survival, safety, pharmacokinetics, patient-reported quality of life and correlative studies. (Investigational new drug (IND) number : 139058, European Clinical Trials database : EudraCT Number 2019-001025-28, ClinicalTrials.gov identifier : NCT04163900).- Published
- 2020
- Full Text
- View/download PDF
42. TG01/GM-CSF and adjuvant gemcitabine in patients with resected RAS-mutant adenocarcinoma of the pancreas (CT TG01-01): a single-arm, phase 1/2 trial.
- Author
-
Palmer DH, Valle JW, Ma YT, Faluyi O, Neoptolemos JP, Jensen Gjertsen T, Iversen B, Amund Eriksen J, Møller AS, Aksnes AK, Miller R, and Dueland S
- Subjects
- Adenocarcinoma pathology, Aged, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Female, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Humans, Male, Middle Aged, Pancreatic Neoplasms pathology, Pancreatic Neoplasms surgery, Gemcitabine, Adenocarcinoma drug therapy, Deoxycytidine analogs & derivatives, Granulocyte-Macrophage Colony-Stimulating Factor therapeutic use, Pancreatic Neoplasms drug therapy
- Abstract
Background: TG01 is the first cancer immunotherapy targeting KRAS oncogenic mutations. This study assessed the safety and efficacy of TG01/GM-CSF in patients with resected pancreatic adenocarcinoma., Methods: Patients with stage I or II pancreatic adenocarcinoma who had undergone surgical resection (R0 or R1) received adjuvant gemcitabine with TG01/GM-CSF using two schedules of vaccination. Immune response was defined as a positive delayed-type hypersensitivity (DTH) response and/or positive T-cell proliferation assay., Results: Thirty-two patients were enrolled between February 2013 and May 2016. Nineteen were treated with the high antigen burden, with four serious adverse reactions considered possibly related to TG01 treatment, including three allergic reactions. On this basis, a further 13 patients received a modified vaccination schedule with reduced antigen burden, with no serious adverse events related to TG01. Ninety-five percent patients in the main cohort and 92% in the modified cohort had a positive immune response. Median overall survival (OS) was 33.1 months, and median disease-free survival (DFS) was 13.9 months for the main cohort. For the modified cohort, the median OS was 34.3 months and median DFS was 19.5 months., Conclusions: TG01/GM-CSF with gemcitabine was well tolerated, with high levels of immune activation. OS and DFS compare favourably with published data for adjuvant gemcitabine., Clinical Trial Registration: This clinical trial was registered at ClinicalTrials.gov (NCT02261714).
- Published
- 2020
- Full Text
- View/download PDF
43. Role of locoregional therapies in the wake of systemic therapy.
- Author
-
Palmer DH, Malagari K, and Kulik LM
- Subjects
- Combined Modality Therapy methods, Humans, Patient Selection, Carcinoma, Hepatocellular therapy, Chemoembolization, Therapeutic methods, Immune Checkpoint Inhibitors administration & dosage, Immunotherapy methods, Liver Neoplasms therapy
- Abstract
Multiple systemic agents have recently been approved in the first- and second-line setting for hepatocellular carcinoma (HCC), increasing the therapeutic options for patients and treating physicians. The randomised controlled trials that led to these approvals were predominantly conducted in a population comprised of patients with advanced HCC. However, these trials also included a subset of patients who had progressed after locoregional therapies (LRTs), mostly transarterial chemoembolisation. With a greater number of systemic agents available, the role of LRTs has become a topic of debate, specifically regarding when to transition to systemic therapy in unresectable HCC and the potential opportunities for combining locoregional and systemic therapies. Trials of immuno-oncology agents (notably T cell checkpoint inhibitors) are ongoing in the advanced disease setting and these agents also present opportunities for combination therapies, both with other systemic agents and with LRTs in earlier stage disease. This article will review strategies to guide patient selection for LRT as well as the development of locoregional-systemic combinations based on scientific rationale and the challenges of clinical trial design in this setting., (Copyright © 2019. Published by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
44. CD40L membrane retention enhances the immunostimulatory effects of CD40 ligation.
- Author
-
Elmetwali T, Salman A, Wei W, Hussain SA, Young LS, and Palmer DH
- Subjects
- Antigens, CD metabolism, Antigens, Neoplasm metabolism, Apoptosis drug effects, CD40 Antigens metabolism, CD40 Ligand genetics, CD40 Ligand pharmacology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Line, Tumor, Cell Proliferation, Dendritic Cells cytology, Dendritic Cells immunology, Dendritic Cells metabolism, Gene Expression Regulation, Neoplastic drug effects, Genetic Vectors genetics, Genetic Vectors metabolism, Humans, Immunoglobulins metabolism, Interferon-gamma metabolism, Interleukin-10 metabolism, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear metabolism, Membrane Glycoproteins metabolism, T-Lymphocytes cytology, T-Lymphocytes metabolism, Urinary Bladder Neoplasms immunology, Urinary Bladder Neoplasms metabolism, Urinary Bladder Neoplasms pathology, CD83 Antigen, CD40 Ligand metabolism, Cell Membrane metabolism, T-Lymphocytes immunology
- Abstract
In carcinomas, the nature of CD40 ligand shapes the outcome of CD40 ligation. To date, the consequences of membrane-bound CD40L (mCD40L) on its immune-stimulatory function are unknown. Here, we examined the impact of mCD40L versus soluble CD40L (sCD40L) on T24 bladder carcinoma gene expression profiling. Of 410 differentially expressed genes, 286 were upregulated and 124 downregulated by mCD40L versus sCD40L. Gene ontology enrichment analysis revealed immune-stimulatory function as the most significant enriched biological process affected by upregulated transcripts, while those downregulated were critical for cell growth and division. Furthermore, immature dendritic cells (iDC) responded to mCD40L with enhanced maturation and activation over sCD40L evidenced by higher expression levels of CD83, CD86, HLA-DR and CD54, increased secretion of IL12 and IL10 and higher tumour-antigen (TA) uptake capacity. Furthermore, autologus CD3+ T cells responded to TA-loaded mCD40L-activated DC with increased proliferation and cytotoxic response (CD107a and IFN-γ-producing CD3+ CD8+ T cells) to the tumour-loaded autologous PBMCs compared to sCD40L. Thus, these data indicate that mCD40L enhances the immunostimulatory capacity over sCD40L. Furthermore, the ability of mCD40L to also directly induce cell death in CD40-expressing carcinomas, subsequently releasing tumour-specific antigens into the tumour microenvironment highlights the potential for mCD40L as a multi-faceted anti-cancer immunotherapeutic.
- Published
- 2020
- Full Text
- View/download PDF
45. A shared genetic basis of mimicry across swallowtail butterflies points to ancestral co-option of doublesex.
- Author
-
Palmer DH and Kronforst MR
- Subjects
- Animals, Biological Mimicry, Butterflies classification, Butterflies metabolism, DNA-Binding Proteins metabolism, Evolution, Molecular, Female, Insect Proteins metabolism, Male, Phenotype, Phylogeny, Species Specificity, Butterflies genetics, DNA-Binding Proteins genetics, Insect Proteins genetics
- Abstract
Uncovering whether convergent adaptations share a genetic basis is consequential for understanding the evolution of phenotypic diversity. This information can help us understand the extent to which shared ancestry or independent evolution shape adaptive phenotypes. In this study, we first ask whether the same genes underlie polymorphic mimicry in Papilio swallowtail butterflies. By comparing signatures of genetic variation between polymorphic and monomorphic species, we then investigate how ancestral variation, hybridization, and independent evolution contributed to wing pattern diversity in this group. We report that a single gene, doublesex (dsx), controls mimicry across multiple taxa, but with species-specific patterns of genetic differentiation and linkage disequilibrium. In contrast to widespread examples of phenotypic evolution driven by introgression, our analyses reveal distinct mimicry alleles. We conclude that mimicry evolution in this group was likely facilitated by ancestral polymorphism resulting from early co-option of dsx as a mimicry locus, and that evolutionary turnover of dsx alleles may underlie the wing pattern diversity of extant polymorphic and monomorphic lineages.
- Published
- 2020
- Full Text
- View/download PDF
46. Tumor burden and liver function in HCC patient selection for selective internal radiation therapy: SARAH post-hoc study.
- Author
-
Palmer DH, Hawkins NS, Vilgrain V, Pereira H, Chatellier G, and Ross PJ
- Subjects
- Aged, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular pathology, Carcinoma, Hepatocellular radiotherapy, Female, Follow-Up Studies, Humans, Liver Function Tests, Liver Neoplasms drug therapy, Liver Neoplasms pathology, Liver Neoplasms radiotherapy, Male, Microspheres, Prognosis, Survival Rate, Tumor Burden, Antineoplastic Agents therapeutic use, Brachytherapy mortality, Carcinoma, Hepatocellular mortality, Liver Neoplasms mortality, Patient Selection, Sorafenib therapeutic use, Yttrium Radioisotopes therapeutic use
- Abstract
Aim: To determine whether a liver tumor burden ≤25% and well-preserved liver function (albumin-bilirubin grade 1) are appropriate criteria for identifying patients with unresectable hepatocellular carcinoma who may benefit from selective internal radiation therapy (SIRT) using
90 yttrium resin microspheres versus sorafenib. Patients & methods: Post-hoc analysis of patients in the intention-to-treat population of the SARAH trial (SIRT vs sorafenib) with ≤25% tumor burden and albumin-bilirubin grade 1. Primary end point: overall survival. Results: Median overall survival was 21.9 months (95% CI: 15.2-32.5, n = 37) with SIRT and 17.0 months (11.6-20.8, n = 48) with sorafenib (hazard ratios: 0.73; 95% CI: 0.44-1.21; p = 0.22). Conclusion: A combination of good liver function and low tumor burden may be relevant for selection of hepatocellular carcinoma patients for SIRT.- Published
- 2020
- Full Text
- View/download PDF
47. Development of an orthotopic syngeneic murine model of colorectal cancer for use in translational research.
- Author
-
Evans JP, Winiarski BK, Sutton PA, Ressel L, Duckworth CA, Pritchard DM, Palmer DH, Goldring CE, and Kitteringham NR
- Subjects
- Animals, Cell Line, Tumor, Liver Neoplasms, Experimental secondary, Male, Mice, Mice, Inbred BALB C, Pilot Projects, Translational Research, Biomedical, Colorectal Neoplasms pathology, Disease Models, Animal
- Abstract
Improving outcomes in colorectal cancer requires more accurate in vivo modelling of the disease in humans, allowing more reliable pre-clinical assessment of potential therapies. Novel imaging techniques are necessary to improve the longitudinal assessment of disease burden in these models, reducing the number of animals required for translational studies. This report describes the development of an immune-competent syngeneic orthotopic murine model of colorectal cancer, utilising caecal implantation of CT26 cells stably transfected with the luciferase gene into immune-competent BALB/c mice, allowing serial bioluminescent imaging of cancer progression. Luminescence in the stably transfected CT26 cell line, after pre-conditioning in the flank of a BALB/c mouse, accurately reflected cell viability and resulted in primary caecal tumours in five of eight (63%) mice in the initial pilot study following caecal injection. Luminescent signal continued to increase throughout the study period with one mouse (20%) developing a liver metastasis. Histopathological assessment confirmed tumours to be consistent with a poorly differentiated adenocarcinoma. We have now performed this technique in 68 immune-competent BALB/c mice. There have been no complications from the procedure or peri-operative deaths, with primary tumours developing in 44 (65%) mice and liver metastases in nine (20%) of these. This technique provides an accurate model of colorectal cancer with tumours developing in the correct microenvironment and metastasising to the liver with a similar frequency to that seen in patients presenting with colorectal cancer, with serial bioluminescent reducing the murine numbers required in studies by removing the need for cull for assessment of disease burden.
- Published
- 2019
- Full Text
- View/download PDF
48. First-in-Human Phase I Study of Fisogatinib (BLU-554) Validates Aberrant FGF19 Signaling as a Driver Event in Hepatocellular Carcinoma.
- Author
-
Kim RD, Sarker D, Meyer T, Yau T, Macarulla T, Park JW, Choo SP, Hollebecque A, Sung MW, Lim HY, Mazzaferro V, Trojan J, Zhu AX, Yoon JH, Sharma S, Lin ZZ, Chan SL, Faivre S, Feun LG, Yen CJ, Dufour JF, Palmer DH, Llovet JM, Manoogian M, Tugnait M, Stransky N, Hagel M, Kohl NE, Lengauer C, Sherwin CA, Schmidt-Kittler O, Hoeflich KP, Shi H, Wolf BB, and Kang YK
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor metabolism, Carcinoma, Hepatocellular metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Drug Administration Schedule, Female, Humans, Liver Neoplasms metabolism, Male, Middle Aged, Pyrans adverse effects, Quinazolines adverse effects, Receptor, Fibroblast Growth Factor, Type 4 antagonists & inhibitors, Signal Transduction drug effects, Treatment Outcome, Young Adult, Carcinoma, Hepatocellular drug therapy, Fibroblast Growth Factors metabolism, Liver Neoplasms drug therapy, Pyrans administration & dosage, Quinazolines administration & dosage
- Abstract
Outcomes for patients with advanced hepatocellular carcinoma (HCC) remain poor despite recent progress in drug development. Emerging data implicate FGF19 as a potential HCC driver, suggesting its receptor, FGFR4, as a novel therapeutic target. We evaluated fisogatinib (BLU-554), a highly potent and selective oral FGFR4 inhibitor, in a phase I dose-escalation/dose-expansion study in advanced HCC using FGF19 expression measured by IHC as a biomarker for pathway activation. For dose escalation, 25 patients received 140 to 900 mg fisogatinib once daily; the maximum tolerated dose (600 mg once daily) was expanded in 81 patients. Fisogatinib was well tolerated; most adverse events were manageable, grade 1/2 gastrointestinal events, primarily diarrhea, nausea, and vomiting. Across doses, the overall response rate was 17% in FGF19-positive patients [median duration of response: 5.3 months (95% CI, 3.7-not reached)] and 0% in FGF19-negative patients. These results validate FGFR4 as a targetable driver in FGF19-positive advanced HCC. SIGNIFICANCE: Fisogatinib elicited clinical responses in patients with tumor FGF19 overexpression in advanced HCC. These results validate the oncogenic driver role of the FGFR4 pathway in HCC and the use of FGF19 as a biomarker for patient selection. See related commentary by Subbiah and Pal, p. 1646 . This article is highlighted in the In This Issue feature, p. 1631 ., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
49. Patterns of Recurrence After Resection of Pancreatic Ductal Adenocarcinoma: A Secondary Analysis of the ESPAC-4 Randomized Adjuvant Chemotherapy Trial.
- Author
-
Jones RP, Psarelli EE, Jackson R, Ghaneh P, Halloran CM, Palmer DH, Campbell F, Valle JW, Faluyi O, O'Reilly DA, Cunningham D, Wadsley J, Darby S, Meyer T, Gillmore R, Anthoney A, Lind P, Glimelius B, Falk S, Izbicki JR, Middleton GW, Cummins S, Ross PJ, Wasan H, McDonald A, Crosby T, Ting Y, Patel K, Sherriff D, Soomal R, Borg D, Sothi S, Hammel P, Lerch MM, Mayerle J, Tjaden C, Strobel O, Hackert T, Büchler MW, and Neoptolemos JP
- Subjects
- Adult, Aged, Aged, 80 and over, Capecitabine administration & dosage, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal mortality, Chemotherapy, Adjuvant, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Disease-Free Survival, Female, Humans, Male, Middle Aged, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local mortality, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms mortality, Prospective Studies, Treatment Outcome, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Pancreatic Ductal surgery, Neoplasm Recurrence, Local etiology, Pancreatic Neoplasms surgery
- Abstract
Importance: The patterns of disease recurrence after resection of pancreatic ductal adenocarcinoma with adjuvant chemotherapy remain unclear., Objective: To define patterns of recurrence after adjuvant chemotherapy and the association with survival., Design, Setting, and Participants: Prospectively collected data from the phase 3 European Study Group for Pancreatic Cancer 4 adjuvant clinical trial, an international multicenter study. The study included 730 patients who had resection and adjuvant chemotherapy for pancreatic cancer. Data were analyzed between July 2017 and May 2019., Interventions: Randomization to adjuvant gemcitabine or gemcitabine plus capecitabine., Main Outcomes and Measures: Overall survival, recurrence, and sites of recurrence., Results: Of the 730 patients, median age was 65 years (range 37-81 years), 414 were men (57%), and 316 were women (43%). The median follow-up time from randomization was 43.2 months (95% CI, 39.7-45.5 months), with overall survival from time of surgery of 27.9 months (95% CI, 24.8-29.9 months) with gemcitabine and 30.2 months (95% CI, 25.8-33.5 months) with the combination (HR, 0.81; 95% CI, 0.68-0.98; P = .03). The 5-year survival estimates were 17.1% (95% CI, 11.6%-23.5%) and 28.0% (22.0%-34.3%), respectively. Recurrence occurred in 479 patients (65.6%); another 78 patients (10.7%) died without recurrence. Local recurrence occurred at a median of 11.63 months (95% CI, 10.05-12.19 months), significantly different from those with distant recurrence with a median of 9.49 months (95% CI, 8.44-10.71 months) (HR, 1.21; 95% CI, 1.01-1.45; P = .04). Following recurrence, the median survival was 9.36 months (95% CI, 8.08-10.48 months) for local recurrence and 8.94 months (95% CI, 7.82-11.17 months) with distant recurrence (HR, 0.89; 95% CI, 0.73-1.09; P = .27). The median overall survival of patients with distant-only recurrence (23.03 months; 95% CI, 19.55-25.85 months) or local with distant recurrence (23.82 months; 95% CI, 17.48-28.32 months) was not significantly different from those with only local recurrence (24.83 months; 95% CI, 22.96-27.63 months) (P = .85 and P = .35, respectively). Gemcitabine plus capecitabine had a 21% reduction of death following recurrence compared with monotherapy (HR, 0.79; 95% CI, 0.64-0.98; P = .03)., Conclusions and Relevance: There were no significant differences between the time to recurrence and subsequent and overall survival between local and distant recurrence. Pancreatic cancer behaves as a systemic disease requiring effective systemic therapy after resection., Trial Registration: ClinicalTrials.gov identifier: NCT00058201, EudraCT 2007-004299-38, and ISRCTN 96397434.
- Published
- 2019
- Full Text
- View/download PDF
50. How to identify sex chromosomes and their turnover.
- Author
-
Palmer DH, Rogers TF, Dean R, and Wright AE
- Subjects
- Animals, Eukaryota genetics, Evolution, Molecular, Female, Male, Sex Chromosomes genetics, Sex Determination Processes genetics
- Abstract
Although sex is a fundamental component of eukaryotic reproduction, the genetic systems that control sex determination are highly variable. In many organisms the presence of sex chromosomes is associated with female or male development. Although certain groups possess stable and conserved sex chromosomes, others exhibit rapid sex chromosome evolution, including transitions between male and female heterogamety, and turnover in the chromosome pair recruited to determine sex. These turnover events have important consequences for multiple facets of evolution, as sex chromosomes are predicted to play a central role in adaptation, sexual dimorphism, and speciation. However, our understanding of the processes driving the formation and turnover of sex chromosome systems is limited, in part because we lack a complete understanding of interspecific variation in the mechanisms by which sex is determined. New bioinformatic methods are making it possible to identify and characterize sex chromosomes in a diverse array of non-model species, rapidly filling in the numerous gaps in our knowledge of sex chromosome systems across the tree of life. In turn, this growing data set is facilitating and fueling efforts to address many of the unanswered questions in sex chromosome evolution. Here, we synthesize the available bioinformatic approaches to produce a guide for characterizing sex chromosome system and identity simultaneously across clades of organisms. Furthermore, we survey our current understanding of the processes driving sex chromosome turnover, and highlight important avenues for future research., (© 2019 The Authors. Molecular Ecology published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.