1 / 76

GI ASCO 2012 Non-colon update

GI ASCO 2012 Non-colon update. Dr. Delilah Lisa Topic, MD, FRCPC Clinical Fellow – St. Michael’s Hospital Supervisor: Dr. Brezden-Masley. GI ASCO 2012 – Jan 19-21 st , 2012 San Francisco, CA. GI ASCO 2012. Non-colon Esophagogastric Pancreatic Hepatobiliary. ESOPHAGOGASTRIC.

kostya
Télécharger la présentation

GI ASCO 2012 Non-colon update

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. GI ASCO 2012Non-colon update Dr. Delilah Lisa Topic, MD, FRCPC Clinical Fellow – St. Michael’s Hospital Supervisor: Dr. Brezden-Masley

  2. GI ASCO 2012 – Jan 19-21st, 2012 San Francisco, CA

  3. GI ASCO 2012 • Non-colon • Esophagogastric • Pancreatic • Hepatobiliary

  4. ESOPHAGOGASTRIC

  5. Esophagogastric • Phase III trial of everolimus in previously treated patients with advanced gastric cancer (AGC): GRANITE-1 • Survival analysis according to disease subtype in AVAGAST: First-line capecitabine and cisplatin, plus bevacizumab or placebo, in patients with AGC

  6. Phase III trial of everolimus (EVE) in previously treated patients with advanced Gastric Cancer: GRANITE-1 Eric Van Cutsem, MD, PhD University Hospital Leuven/Belgium

  7. Background • Gastric cancer is aggressive and difficult to treat • 5 year survival for advanced/metastatic is <5% • Limited options upon failure of first-line chemotherapy

  8. PI3k/Akt/mtor pathway Key regulator of cell proliferation, growth, survival, metabolism, and angiogenesis Disregulated in 50-60% of gastric cancersEverolimus -Oral mTor inhibitor, efficacy in preclinical models of gastric cancer -Promising efficacy and tolerability in small phase II study (n=53) OS 10.1 mos, PFS 2.7mos

  9. Phase 3 GRANITE-1 Study Design • Confirmed advanced gastric cancer • Progression after 1 or 2 lines of previous systemic chemotherapy • Stratification by region: Asia vs rest of world • Stratification by number of lines of previous systemic chemotherapy (1 vs 2) Everolimus 10 mg PO daily+ BSC* (n = 439) RANDOMIZE 2:1 (N = 656) SCREEN Safety follow-up: EOT + 28 d Treatment until disease progression or intolerable toxicity Survival follow-up: every 3 mo Placebo PO daily + BSC (n = 217) BSC, best supportive care; EOT, end of treatment; PO, orally. ClinicalTrials.gov identifier: NCT00879333. Van Cutsem E et al. GI Cancer Symposium 2012 (Abstr LBA3).

  10. Eligibility criteria • Inclusion criteria • Age >18yrs • Confirmed gastric adenocarcinoma • Documented progression after 1-2 lines of chemo • ECOG ≤2 • Adequate bone marrow, renal, and hepatic function • Exclusion criteria • >2 lines of systemic treatment for advanced disease • Anticancer treatment within 3 wks or major surgery within 2 weeks of study randomization • Chronic treatment with steriods or immunosuppressive agents • Enteral feeding • CNS metastases • Any severe/uncontrolled medical condition

  11. Study Endpoints • Primary: • OS • Secondary: • PFS • ORR • AEs • Time to definitive deterioration of ECOG • Time to 5% deterioration in global health/QOL • Exploratory: • Correlation between biomarkers and clinical endpoints

  12. Participating Countries • North America • Canada • United States • Central and South America • Argentina • Colombia • Mexico • Peru • East Asia • China • Hong Kong • Japan • Korea • Taiwan • Other Asia and Pacific Region • Australia • New Zealand • Thailand • Europe and Middle East • Belgium • France • Germany • Israel • Italy • Netherlands • Russia • Spain • United Kingdom

  13. Baseline Patient Characteristics

  14. Baseline Disease Characteristics

  15. Overall Survival 100 Censoring Times Everolimus + BSC (n/N = 352/439) Placebo + BSC (n/N = 180/217) 80 Kaplan-Meier medians Everolimus + BSC: 5.39 months Placebo + BSC: 4.34 months 60 Hazard ratio: 0.90 (95% CI, 0.75-1.08) Probability of overall survival (%) Log-rank P value = 0.1244 40 20 0 0 2 4 6 8 10 12 14 16 18 20 22 24 Time (months) No. of patients still at risk Time (months) 0 2 4 6 8 10 12 14 16 18 20 22 24 Everolimus 439 355 253 195 139 87 52 30 13 6 3 1 0 217 172 1 17 82 60 35 28 16 12 8 4 1 0 Placebo CI, confidence interval. Van Cutsem E et al. GI Cancer Symposium 2012 (Abstr LBA3).

  16. Overall Survival by Stratification Factors Hazard Ratio (95% CI) All (N = 656) 0.90 (0.75-1.08) Prior chemotherapy 1 (n = 313) 0.94 (0.73-1.23) 2 (n = 343) 0.90 (0.70-1.15) Region Asia (n = 363) 0.96 (0.75-1.23) ROW (n = 293) 0.85 (0.65-1.10) 1 prior chemo & Asia (n = 146) 0.94 (0.63-1.39) Cross-class. of strata 2 prior chemo & Asia (n = 217) 0.98 (0.71-1.35) 1 prior chemo & ROW (n = 167) 0.91 (0.64-1.31) 2 prior chemo & ROW (n = 126) 0.74 (0.50-1.09) 0.6 0.8 1.0 1.2 1.4 Everolimus 10 mg/d Placebo In favor of ROW, rest of world. Van Cutsem E et al. GI Cancer Symposium 2012 (Abstr LBA3).

  17. Progression-Free Survival 100 Censoring Times Everolimus + BSC (n/N = 386/439) Placebo + BSC (n/N = 206/217) 80 Kaplan-Meier medians Everolimus + BSC: 1.68 months Placebo + BSC: 1.41 months 60 Hazard ratio: 0.66 (95% CI, 0.56-0.78) Probability of progression-free survival (%) Log-rank P value < 0.0001 40 20 0 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Time (months) No. of patients still at risk Time (months) 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Everolimus 439 367 179 117 92 60 44 37 27 20 13 10 6 5 3 3 2 1 1 0 0 0 217 168 55 28 23 17 8 7 6 3 2 2 2 2 2 2 2 2 2 2 1 0 Placebo Van Cutsem E et al. GI Cancer Symposium 2012 (Abstr LBA3).

  18. Tumour Response

  19. Best Percentage Change From Baseline in Tumor Size 160% 160% 140% 140% 120% 120% 100% 100% 80% 80% Everolimus 10 mg/day (n = 304) Placebo (n = 154) 60% 60% Best % change from baseline (measurable lesions) 40% 40% 20% 20% 0% 0% –20% –20% –40% –40% –60% –60% –80% –80% –100% –100% Van Cutsem E et al. GI Cancer Symposium 2012 (Abstr LBA3).

  20. Adverse Events

  21. Most Common AEs

  22. GRANITE-1Conclusions • Everolimusmonotherapy did not significantly improve OS in patients with AGC as second/third line therapy • Everolimus did reduce the risk of progression, compared with BSC • Median PFS 1.411.68, HR 0.66, p<0.001 • Disease control 22%43%

  23. GRANITE-1 Conclusions • Safety profile was similar to that observed with everolimus in other malignancies • Disease control signal worth further study? • Biomarkers to identify those who benefit

  24. Survival analysis according to disease subtype in AVAGAST: First-line capecitabine and cisplatin plus bevacizumab or placebo in pts with advanced gastric cancer Manish Shah, MD

  25. Gastric cancer: not one disease! • Clinical/epidemiological data suggest there are three subtypes: • Type 1: proximal, non-diffuse • Type 2: diffuse • Type 3: distal, non-diffuse • Each subtype has different gene expression profile

  26. Gastric cancer: 3 subtypes

  27. AVAGAST • Global, randomized, phase III study • Bevacizumab + chemo vs. placebo + chemo (first-line treatment for AGC) • Primary endpoint: OS • Was not met (12 mosvs 10 mos, p=0.1002) • Regional efficacy differences were noted • Patients from Europe/Americas did better

  28. Regional differences • Several analyses have been performed to better understand regional differences • Analysis has revealed: • It is the ‘high risk’ pts from Europe/Americas that derive more benefit from bevacizumab

  29. Objectives • To examine the OS data according to gastric cancer subtype and region • To identify if disease subtype was: • Prognostic • Predictive of bevacizumab benefit (Europe/Americas) • To examine the distribution of angiogenic biomarkers across subtypes • Do gastric cancer subtypes have different biomarker expression profiles?

  30. AVAGAST: trial design Placebo + capecitabine + cisplatin (n=387) Locally advanced or metastatic gastric cancer(n=774) R Bevacizumab + capecitabine + cisplatin (n=387) Stratification factors: 1. Geographic region 2. Fluoropyrimidine backbone 3. Disease status Endpoints Primary: Overall survival Secondary: Progression-free survival Exploratory: Changes in candidate biomarkers: pVEGFA, NRP-1, VEGFA, VEGFR1, VEGFR2 5-FU allowed if capecitabine contraindicated Maximum of 6 cycles of cisplatin Capecitabine and bevacizumab / placebo until PD

  31. Disease subtype: distribution by region All patients (n=733) Europe/Americas (n=378) Asia-Pacific (n=355)

  32. Biomarker distribution • There appears to be differences in biomarker distribution amongst gastric cancer subtypes

  33. Disease subtype: prognostic effect (overall survival in non-Asian patients) Survival rate (%) Proximal Diffuse Distal 100 90 80 70 60 50 40 30 20 10 0 0 3 6 9 12 15 18 21 24 Studymonth No. at risk Type 1 Type 2 Type 3 30 91 72 28 72 62 22 47 47 18 28 35 14 22 21 7 13 12 3 5 4 1 1 0 0 0 0

  34. Overall survival by disease subtype

  35. Does disease subtype predict response to Bevacizumab? (Europe/Americas only)

  36. Overall survival in pts with diffuse/distal disease (Europe/Americas only) Population: Europe/Americas with type 2/3 disease 100 90 Survival rate (%) Pla + chemo Bev + chemo 80 70 60 50 Hazard ratio 0.67 (95% CI 0.52–0.88) 40 30 20 10 00 0 3 6 9 12 15 18 21 24 Study month No. at risk Pla + chemo Bev + chemo 43 63 94 119 25 28 163 159 0 0 9 10 134 144 1 3 63 94

  37. Predictive biomarkers with bevacizumab

  38. Biomarker distribution according to subtypes

  39. Biomarker distribution (summary) • Proximal (type 1) gastric cancer appears to have the ‘worst’ profile for bevacizumab • High NRP-1, low pVEGF-A • Diffuse (type 2) and distal (type 3) appear to have at least one biomarker that may support benefit to an antioangiogenic strategy • Diffuse (type 2)  low NRP-1 • Distal (type 3)  high pVEGF-A

  40. AVAGAST Conclusions • Gastric cancer is more than one disease! • Gastric cancer subtypes have different prognoses • In all regions, diffuse (type 2) did worse • The addition of bev to chemo appears to improve outcomes in pts from Europe/Americas with diffuse and distal disease • Biomarkers NRP-1 and plasma pVEGF-A provide a rationale for subtype-specific outcomes with bevacizumab • Additional evaluation warranted

  41. Pancreatic

  42. Malignant progression in intraductal papillary mucinousneoplasms (IPMN) of the pancreas: Results of 157 patients selected for radiographic surveillance Jennifer Lafemina, MD Memorial Sloan-Kettering

  43. “Its just a cyst – don’t worry!”

  44. Intraductal papillary mucinousneoplasms (IPMN) – not “just a cyst” • Represents a field defect of ductal instability • Main and branch duct IPMN carry a risk of malignancy intarget cyst itself • 57-95% for MD-IPMN • 6-46% for BD-IPMN • Risk of developing malignancy in region other than target cyst is poorly defined

  45. Objectives • Identify patients with IPMN who underwent resection • Define: • Pathologic characteristics of target cyst • Risk of developing a PDAC in a region separate from the target cyst

  46. Methods • Retrospective review • Pts evaluated at MSKCC from Feb ‘89-Aug ‘10: • Radiologic confirmation of target cyst • Pathologic confirmation of IPMN and/or cyst fluid CEA level ≥200ng/mL • Time from IPMN diagnosis to resection ≥6 mos

  47. Patient demographics

  48. Objectives • Identify patients with IPMN who underwent resection • Define pathologic characteristics of target cyst • Define the risk of developing a PDAC in a region separate from the target cyst

  49. Target cyst characteristics

More Related