Arkadiusz Czerwiński commited on
Commit
0d3e7f2
1 Parent(s): c5478f1

feat: initial changes

Browse files
samples/APOLLO-2-leaderboard.tsv ADDED
@@ -0,0 +1,118 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ year_of_birth days_to_birth gender race ethnicity bmi alcohol_history alcohol_intensity smokestat_apollo2 cigarettes_per_day2 years_smoked2 family_cancer_history dzextent_beyondabd dzextentpriortosurg primary_diagnosis site_of_resection_or_biopsy classification_of_tumor stage_figo_2014 morphology celltype tumor_grade sticpresent brcastat brca1ihc_apollo disease_distribution diseasehigh miliarypresent residual_disease anyresidualdisease days_to_last_known_disease_status last_known_disease_status fuflag days_to_recurrence pfstimeindays pfs_status survivaltimeindays vital_status causeofdeath_apollo2 year_of_death days_to_death days_to_treatment treatment_intent_type treatment_or_therapy apollo2_adv_hg_serous_complete
2
+ 1927 -30246 FEMALE Caucasian or White Hispanic or Latina 20.12307203 Yes 14 Previous Smoker 1 2 members Mother with liver cancer Father with prostate cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G2-moderately differentiated No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive Measurable > 1 cm Yes 1023 Residual or persistent disease no progression Not lost 607 607 Event 1023 Alive 19 Adjuvant Chemotherapy No 0
3
+ 1930 -29118 FEMALE Caucasian or White Hispanic or Latina 20.85277293 No Previous Smoker 1 3 members Maternal Uncle with esophageal cancer at 50 Other Relative nos 1 with breast cancer at 47 Other Relative nos with lung cancer at 60 Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DM R0 Ovary PRIMARY IIIA Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated Yes Unknown Unknown Moderate disease distribution greater than low but less than high No No Macroscopic < 1 cm Yes 278 Disease progression Died 217 217 Event 278 Deceased Disease progression 2010 278 26 Adjuvant Chemotherapy No 0
4
+ 1930 -28671 FEMALE Caucasian or White Non-Spanish / Non-Hispanic / Non-Latina 21.32720483 Yes Current Smoker 10 2 1 member Father with lung cancer at 74 High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the Fallopian tube DH R1 OVARY METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis No No None or microscopic No 695 Disease progression 129 Deceased 2013 903 55 Adjuvant Chemotherapy No 0
5
+ 1930 -29312 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.07856301 No Previous Smoker 20 4 2 members Father with colon cancer Mother with lung cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive Macroscopic < 1 cm Yes 200 Residual or persistent disease no progression Alive less than 1 year follow up then lost 308 Event 308 Alive 7 Adjuvant Chemotherapy No 0
6
+ 1931 -29686 FEMALE Caucasian or White Non-Hispanic or Non-Latina 23.32450917 Unknown Previous Smoker 17 4 3 members Sister with breast cancer Maternal Grandmother with breast cancer Other Family Member NOS with ovarian cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the Fallopian tube DH R0 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown Moderate disease distribution greater than low but less than high No No Macroscopic < 1 cm Yes 568 Disease progression Died 383 383 Event 568 Deceased Disease progression 2010 568 25 Adjuvant Chemotherapy No 0
7
+ 1934 -27475 FEMALE Caucasian or White Non-Hispanic or Non-Latina 34.38271971 Yes 0.6 Previous Smoker 10 7 2 members Mother with leukemia Daughter with breast cancer at 32 No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis No No None or microscopic No 1071 Disease progression Died 623 623 Event 1071 Deceased Disease progression 2013 1071 34 Adjuvant Chemotherapy No 0
8
+ 1934 -27670 FEMALE Caucasian or White Non-Hispanic or Non-Latina 20.47871 No Previous Smoker 0.5 7 2 members Mother with breast cancer Sister with breast cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the Fallopian tube DH R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild Macroscopic < 1 cm Yes 1323 Residual or persistent disease no progression Not lost 390 390 Event 1323 Alive 20 Adjuvant Chemotherapy No 0
9
+ 1935 -27658 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.49433107 Yes 2.5 Previous Smoker 20 8 3 members Father with lung cancer Maternal Aunt with breast cancer in 40s Paternal Aunt with colon cancer in late 50s Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DM R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 399 Disease progression Not lost 399 399 Event 399 Alive 58 Adjuvant Chemotherapy No 0
10
+ 1935 -27857 FEMALE Caucasian or White Non-Hispanic or Non-Latina 24.02958907 No Previous Smoker 8 2 members Father with prostate cancer Paternal Grandmother with breast cancer No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DM R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Positive expression level NOS High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Macroscopic < 1 cm Yes 1100 Residual or persistent disease no progression Not lost 432 432 Event 1294 Alive 16 Adjuvant Chemotherapy No 0
11
+ 1935 -28021 FEMALE Caucasian or White Non-Hispanic or Non-Latina 24.53531247 Yes 14 Previous Smoker 10 10 1 member Maternal Uncle with lung cancer at 40 No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the Fallopian tube DH R1 post NACT OVARY LEFT METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 273 Disease progression Died 64 64 Event 273 Deceased Disease progression 2010 273 46 Adjuvant Chemotherapy No 0
12
+ 1935 -27521 FEMALE Caucasian or White Non-Hispanic or Non-Latina 24.93917914 No Previous Smoker 20 13 1 member Paternal Grandfather with lung cancer Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DM R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown Moderate disease distribution greater than low but less than high No Yes Mild Macroscopic < 1 cm Yes 742 Residual or persistent disease no progression Not lost 517 517 Event 742 Alive 50 Adjuvant Chemotherapy No 0
13
+ 1936 -27256 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.18231105 Unknown Previous Smoker 60 14 No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 post NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 1579 Residual or persistent disease no progression Not lost 825 825 Event 1579 Alive 56 Adjuvant Chemotherapy No 0
14
+ 1937 -26527 FEMALE Caucasian or White Non-Hispanic or Non-Latina 29.09640326 Yes 0.5 Previous Smoker 20 15 2 members Brother with brain cancer died at 32 Brother with lung cancer No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 876 Disease progression Died 307 307 Event 876 Deceased Disease progression 2013 876 35 Adjuvant Chemotherapy No 0
15
+ 1937 -26675 FEMALE Caucasian or White Non-Hispanic or Non-Latina 31.16687996 Yes 3 Previous Smoker 10 15 Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R0 post NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Macroscopic < 1 cm Yes 876 Disease progression Died 307 307 Event 876 Deceased Disease progression 2013 876 23 Adjuvant Chemotherapy No 0
16
+ 1937 -27456 FEMALE Caucasian or White Non-Hispanic or Non-Latina 21.2584689 Yes 0.1 Previous Smoker 30 16 1 member Father with brain cancer died at 64 High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary HD R3 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Mutant Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 1086 Residual or persistent disease no progression Not lost 516 516 Event 1086 Alive 54 Adjuvant Chemotherapy No 0
17
+ 1937 -27130 FEMALE Caucasian or White Non-Hispanic or Non-Latina 24.50811353 Yes 3 Previous Smoker 10 20 2 members Maternal Cousin with breast cancer Maternal Cousin with uterine cancer Yes High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DM R0 post NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Macroscopic < 1 cm Yes 815 Disease progression Died 383 383 Event 815 Deceased Disease progression 2012 815 3 Neoadjuvant and Adjuvant Chemotherapy Yes 0
18
+ 1938 -26118 FEMALE Caucasian or White Non-Hispanic or Non-Latina 23.29590458 Yes 4 Previous Smoker 10 20 2 members Mother with basal cell carcinoma Maternal Grandmother with ovarian cancer and uterine cancer No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the Fallopian tube DH R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated Yes Wild Type Unknown Moderate disease distribution greater than low but less than high No No Macroscopic < 1 cm Yes 534 Disease progression Died 425 425 Event 534 Deceased Disease progression 2012 534 15 Neoadjuvant and Adjuvant Chemotherapy Yes 0
19
+ 1938 -26151 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.02064258 Yes Previous Smoker 15 20 4 members Father with stomach cancer at 56 and colon cancer Sister 1 with cancer type unknown at 61 Sister 2 with cancer type unknown Maternal Aunt with breast cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R1 OVARY LEFT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive Non-measurable > 1 cm Yes 1159 Disease progression Died 605 605 Event 1159 Deceased Disease progression 2013 1159 16 Adjuvant Chemotherapy No 0
20
+ 1938 -27029 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.53592684 Yes 0.2 Current Smoker 20 24 Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 1263 Disease progression Died 576 576 Event 1263 Deceased Disease progression 2013 1263 16 Neoadjuvant and Adjuvant Chemotherapy No 0
21
+ 1939 -25166 FEMALE Caucasian or White Non-Hispanic or Non-Latina 20.58334743 Yes 0.1 10 25 5 members Mother with skin cancer Maternal Grandmother with skin cancer Maternal Aunt with pancreatic cancer at 86 Cousin with breast cancer at 58 Paternal Uncle with cancer type unknown Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the Fallopian tube DM R0 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 1428 Disease progression Not lost 471 471 Event 1428 Alive 21 Adjuvant Chemotherapy No 0
22
+ 1939 -26064 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.61644288 No Current Smoker 20 25 Father with prostate cancer and pancreatic cancer Mother with lung cancer No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Wild Type Unknown Moderate disease distribution greater than low but less than high No No Non-measurable > 1 cm Yes 570 Recurrence new lesion Not lost 570 570 Event 570 Alive 10 Neoadjuvant and Adjuvant Chemotherapy Yes 0
23
+ 1939 -25794 FEMALE Caucasian or White Non-Hispanic or Non-Latina 28.84186767 Yes 3.5 Previous Smoker 20 30 1 member Father with colorectal cancer No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R2 OVARY RIGHT PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Macroscopic < 1 cm Yes 784 No evidence of disease Not lost 784 Censored 784 Alive 10 Neoadjuvant and Adjuvant Chemotherapy No 0
24
+ 1940 -25649 FEMALE Caucasian or White Non-Hispanic or Non-Latina 29.79257498 Unknown Current Smoker 20 30 4 members Mother with breast cancer Maternal Aunt with breast cancer Paternal Aunt 1 with breast cancer Paternal Aunt 2 with colorectal cancer No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the peritoneum DH R2 OVARY METASTATIC IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive Measurable > 1 cm Yes 831 Disease progression Died 831 Censored 831 Deceased Disease progression 2013 831 4 Neoadjuvant and Adjuvant Chemotherapy Yes 0
25
+ 1940 -25094 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.5545495 Yes Previous Smoker 10 30 Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis No No Macroscopic < 1 cm Yes 1660 Disease progression Died 520 520 Event 1660 Deceased Disease progression 2013 1660 16 Neoadjuvant and Adjuvant Chemotherapy Yes 0
26
+ 1940 -26074 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.6265161 Yes 3.5 Previous Smoker 10 32 1 member Father with cancer type unknown No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R1 post NACT Ovary PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated No Unknown Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 244 Disease progression Died 238 238 Event 244 Deceased Disease progression 2012 244 24 Adjuvant Chemotherapy No 0
27
+ 1940 -25886 FEMALE Caucasian or White Non-Hispanic or Non-Latina 28.13113805 Yes 11 Previous Smoker 20 35 3 members Maternal Aunt with liver cancer and esophageal cancer Cousin with lung cancer Maternal Uncle cancer type unknown No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the Fallopian tube DH R0 post NACT FALLOPIAN TUBE PRIMARY Serous adenocarcinoma SEROUS ADENOCARCINOMA G3-poorly differentiated Yes Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Measurable > 1 cm Yes 474 No evidence of disease Alive 1 to 2 years follow up then lost 474 Censored 474 Alive 16 Neoadjuvant Chemotherapy Yes 0
28
+ 1941 -24531 FEMALE Caucasian or White Non-Hispanic or Non-Latina 27.7321388 Yes 2 Previous Smoker 20 40 3 members Father with lung cancer Sister with lung cancer and skin cancer Mother with pancreatic cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DM R3 OVARY PRIMARY IIIA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 142 No evidence of disease Alive less than 1 year follow up then lost 142 Censored 142 Alive 18 Neoadjuvant Chemotherapy Yes 0
29
+ 1941 -24541 FEMALE Caucasian or White Non-Hispanic or Non-Latina 32.91859384 No Previous Smoker 40 5 members Father with lung cancer Mother with bone cancer and ovarian cancer Maternal Grandmother with cancer type unknown Grandchild with cancer type unknown Nephew with cancer type unknown Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild Macroscopic < 1 cm Yes 283 Disease progression Died 272 272 Event 283 Deceased Disease progression 2011 283 25 Adjuvant Chemotherapy No 0
30
+ 1941 -24938 FEMALE Caucasian or White Non-Hispanic or Non-Latina 19.86351239 No Current Smoker 20 40 1 member Father with stomach cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IIIB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive Macroscopic < 1 cm Yes 319 Disease progression Died 319 319 Event 319 Deceased Disease progression 2011 319 56 Adjuvant Chemotherapy No 0
31
+ 1941 -24735 FEMALE Asian Non-Hispanic or Non-Latina 24.19655895 No Current Smoker 20 49 2 members Mother with breast cancer at 65 Father with stomach cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 OVARY LEFT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 446 Disease progression Died 310 310 Event 446 Deceased Disease progression 2013 446 21 Adjuvant Chemotherapy No 0
32
+ 1941 -24880 FEMALE Caucasian or White Non-Hispanic or Non-Latina 27.57718644 Yes Current Smoker 14 50 1 member Father with lung cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R3 post NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild Macroscopic < 1 cm Yes 824 Residual or persistent disease no progression Not lost 268 268 Event 824 Alive 32 Adjuvant Chemotherapy No 0
33
+ 1941 -25925 FEMALE Caucasian or White Non-Hispanic or Non-Latina 33.84257813 Unknown Current Smoker 20 55 3 members Father with prostate cancer Female Cousin with breast cancer at 35 Maternal Uncle with leukemia Yes Unknown Advanced high grade serous cancer of the peritoneum DH R1 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS None or microscopic No 1046 Recurrence new lesion Not lost 948 948 Event 1046 Alive 19 Neoadjuvant and Adjuvant Chemotherapy Yes 0
34
+ 1941 -25212 FEMALE Caucasian or White Non-Hispanic or Non-Latina 21.81183326 Yes 0 Never Smoked 2 members Mother with breast cancer Sister with leukemia No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DM R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen None or microscopic 1801 No evidence of disease Not lost 1214 1214 Event 1812 Alive 20 Adjuvant Chemotherapy No 0
35
+ 1942 -25583 FEMALE Caucasian or White Non-Hispanic or Non-Latina 24.46394815 Yes 0 Never Smoked 2 members Mother with breast cancer Sister with leukemia Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the Fallopian tube DH R1 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild None or microscopic No 1138 No evidence of disease Not lost 1138 Censored 1138 Alive 27 Adjuvant Chemotherapy No 0
36
+ 1942 -24801 FEMALE Caucasian or White Non-Hispanic or Non-Latina 32.98605086 Yes 0 Never Smoked No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the Fallopian tube DH R0 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 666 Disease progression Died 333 333 Event 666 Deceased Disease progression 2009 666 39 Adjuvant Chemotherapy No 0
37
+ 1942 -25668 FEMALE Caucasian or White Non-Hispanic or Non-Latina 16.65625 Yes 0 Never Smoked 3 members Aunt with breast cancer Cousin with ovarian cancer Grandfather with leukemia Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DL R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown Moderate disease distribution greater than low but less than high No No Measurable > 1 cm Yes 1039 Recurrence new lesion Not lost 1039 1039 Event 1272 Alive 20 Adjuvant Chemotherapy No 0
38
+ 1943 -25380 FEMALE Caucasian or White Non-Hispanic or Non-Latina 38.86423323 Yes 0 Never Smoked Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 1479 Disease progression Died 416 416 Event 1479 Deceased Disease progression 2014 1479 36 Adjuvant Chemotherapy No 0
39
+ 1943 -25357 FEMALE Caucasian or White Non-Hispanic or Non-Latina 20.224959 Yes 0 Never Smoked 4 members Mother with lung cancer at 72 Sister with lung cancer Daughter with breast cancer Paternal Aunt with gynecologic cancer nos No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the Fallopian tube DM R0 post NACT TERMINAL ILEUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Mutant Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 77 Disease progression Died 30 30 Event 77 Deceased Disease progression 2010 77 5 Neoadjuvant and Adjuvant Chemotherapy Yes 0
40
+ 1943 -25135 FEMALE Caucasian or White Non-Hispanic or Non-Latina 23.77378393 Yes 0.08 Never Smoked 1 member Mother with melanoma No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the Fallopian tube DL R0 OVARY RIGHT METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Mutant Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 882 Disease progression Died 372 372 Event 882 Deceased Disease progression 2012 882 12 Neoadjuvant and Adjuvant Chemotherapy Yes 0
41
+ 1944 -23961 FEMALE Caucasian or White Non-Hispanic or Non-Latina 24.65325444 Yes 0.08 Never Smoked 1 member Maternal Grandfather with prostate cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DM R2 OVARY RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Macroscopic < 1 cm Yes 727 Disease progression Not lost 377 377 Event 727 Alive 70 Adjuvant Chemotherapy No 0
42
+ 1944 -23822 FEMALE Caucasian or White Non-Hispanic or Non-Latina 17.8076743 Yes 0.1 Never Smoked 1 member Mother with leukemia Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 893 Residual or persistent disease no progression Not lost 403 403 Event 893 Alive 22 Adjuvant Chemotherapy No 0
43
+ 1945 -23042 FEMALE Caucasian or White Non-Hispanic or Non-Latina 17.8953125 Yes 0.1 Never Smoked Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R2 OVARY LEFT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 958 No evidence of disease Not lost 374 374 Event 958 Alive 35 Adjuvant Chemotherapy No 0
44
+ 1945 -24368 FEMALE Caucasian or White Non-Hispanic or Non-Latina 18.59905312 Yes 0.1 Never Smoked 1 member Mother with colon cancer at 63 No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 1069 No evidence of disease Not lost 1069 Censored 1069 Alive 5 Neoadjuvant and Adjuvant Chemotherapy Yes 0
45
+ 1945 -24247 FEMALE Caucasian or White Non-Hispanic or Non-Latina 19.08880674 Yes 0.1 Never Smoked 1 member Paternal grandfather with colon cancer No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 1169 Disease progression Died 90 90 Event 1169 Deceased Disease progression 2013 1169 6 Neoadjuvant Chemotherapy No 0
46
+ 1945 -24719 FEMALE Caucasian or White Non-Hispanic or Non-Latina 19.31984017 Yes 0.2 Previous Smoker 4 members Mother with colon cancer in 70s Brother with colon cancer Aunt 1 with breast cancer at 57 Aunt 2 with bladder cancer Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DL R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Measurable > 1 cm Yes 404 Disease progression Died 287 287 Event 404 Deceased Disease progression 2010 404 20 Adjuvant Chemotherapy No 0
47
+ 1945 -23799 FEMALE Caucasian or White Non-Hispanic or Non-Latina 20.67823854 Yes 0.25 Never Smoked Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the peritoneum DH R3 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild Measurable > 1 cm Yes 405 Disease progression Died 356 356 Event 405 Deceased Disease progression 2013 405 41 Adjuvant Chemotherapy No 0
48
+ 1945 -23515 FEMALE Asian Non-Hispanic or Non-Latina 20.79755639 Yes 0.5 Never Smoked 3 members Maternal Grandmother with breast cancer Paternal Grandfather with cancer type unknown Paternal Aunt with cancer type unknown High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DM R1 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis No No None or microscopic No 784 No evidence of disease Not lost 784 Censored 784 Alive 18 Adjuvant Chemotherapy No 0
49
+ 1945 -24131 FEMALE Caucasian or White Non-Hispanic or Non-Latina 20.9469767 Yes 0.6 Never Smoked No Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DL R1 post NACT OVARY RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 904 Residual or persistent disease no progression Not lost 711 711 Event 904 Alive Neoadjuvant and Adjuvant Chemotherapy Yes 0
50
+ 1946 -23624 FEMALE Caucasian or White Non-Hispanic or Non-Latina 20.97265235 Yes 0.6 Never Smoked Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DM R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 1045 Residual or persistent disease no progression Not lost 502 502 Event 1273 Alive 37 Adjuvant Chemotherapy No 0
51
+ 1946 -23540 FEMALE Caucasian or White Non-Hispanic or Non-Latina 21 Yes 0.6 Never Smoked No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the peritoneum DM R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 751 Disease progression Died 386 386 Event 751 Deceased Disease progression 2012 751 13 Neoadjuvant and Adjuvant Chemotherapy Yes 0
52
+ 1946 -22853 FEMALE Caucasian or White Non-Hispanic or Non-Latina 21.20433359 Yes 1 Never Smoked 4 members Mother with breast Cancer at 60 recurred twice Brother with rectal cancer at 68 Paternal Aunt 1 with breast cancer Paternal Aunt 2 with breast cancer Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DM R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Low expression Moderate disease distribution greater than low but less than high No Yes Mild None or microscopic No 241 Disease progression Died 209 209 Event 241 Deceased Disease progression 2010 241 6 Neoadjuvant and Adjuvant Chemotherapy Yes 0
53
+ 1946 -23048 FEMALE Caucasian or White Non-Hispanic or Non-Latina 21.29729683 Yes 1 Never Smoked Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R1 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 807 Residual or persistent disease no progression Not lost 311 311 Event 807 Alive 24 Neoadjuvant and Adjuvant Chemotherapy Yes 0
54
+ 1946 -23247 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.44379628 Yes 1 Never Smoked No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the Fallopian tube DM R1 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS None or microscopic No 987 Disease progression Died 452 452 Event 987 Deceased Both disease progression and cancer treatment 2013 987 45 Adjuvant Chemotherapy No 0
55
+ 1946 -23534 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.96030049 Yes 2 Never Smoked 4 members Mother with pancreatic cancer Father with skin cancer Brother with cancer type unknown Maternal Grandmother with throat cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R1 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Unknown Unknown Moderate disease distribution greater than low but less than high No None or microscopic 1623 Disease progression Not lost 761 761 Event 1623 Alive 64 Adjuvant Chemotherapy No 0
56
+ 1946 -23168 FEMALE Caucasian or White Non-Hispanic or Non-Latina 22.9977045 Yes 3 Never Smoked 2 relatives Mother with uterine cancer in 1990 died unknown cause Father with lung cancer in 1989 died unknown cause No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the Fallopian tube DH R3 pre NACT UTERUS METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Non-measurable > 1 cm Yes 412 Disease progression Died 333 333 Event 412 Deceased Disease progression 2012 412 13 Adjuvant Chemotherapy No 0
57
+ 1947 -23240 FEMALE Caucasian or White Non-Hispanic or Non-Latina 23.65 Yes 5 Never Smoked 1 member Mother with pancreatic cancer No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the peritoneum DH R0 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Unknown Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis No No None or microscopic No 1082 Disease progression Died 85 85 Event 1082 Deceased Disease progression 2011 1082 15 Neoadjuvant and Adjuvant Chemotherapy No 0
58
+ 1947 -23519 FEMALE Black or African American Non-Hispanic or Non-Latina 23.92291643 Yes 14 Never Smoked 1 member Grandfather with colon cancer Yes Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R3 pre NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 117 Disease progression Died 31 31 Event 117 Deceased Both disease progression and cancer treatment 2010 117 21 Neoadjuvant and Adjuvant Chemotherapy Yes 0
59
+ 1947 -23545 FEMALE Asian Non-Hispanic or Non-Latina 24.40139559 No Never Smoked 1 member Maternal Grandmother with breast cancer at 55 No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the peritoneum DM R0 PERITONEUM PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild None or microscopic No 663 Residual or persistent disease no progression Not lost biopsy taken for suspious progression 663 663 Censored 663 Alive 11 Neoadjuvant and Adjuvant Chemotherapy Yes 0
60
+ 1948 -22734 FEMALE Caucasian or White Non-Hispanic or Non-Latina 24.60629921 No Never Smoked 1 member Mother with breast cancer No Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R1 post NACT OVARY METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Unknown Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 915 No evidence of disease Not lost 375 375 Event 915 Alive 8 Neoadjuvant and Adjuvant Chemotherapy Yes 0
61
+ 1948 -22688 FEMALE Caucasian or White Non-Spanish / Non-Hispanic / Non-Latina 25.05798003 Yes Previous Smoker 2 members Sister with colorectal cancer at 82 Sister with esophageal cancer at 72 Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the Fallopian tube DL R0 post NACT Fallopian Tube PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Measurable > 1 cm Yes 1452 Residual or persistent disease no progression Not lost 550 550 Event 1452 Alive 14 Adjuvant Chemotherapy No 0
62
+ 1948 -22349 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.3383081 No Previous Smoker 10 3 members Paternal Grandmother with colon cancer Paternal Grandfather with colon cancer Paternal Aunt with colon cancer No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DM R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Mutant Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 1516 Disease progression Died 782 782 Event 1516 Deceased Disease progression 2012 1516 29 Adjuvant Chemotherapy No 0
63
+ 1948 -22111 FEMALE Black or African American Non-Hispanic or Non-Latina 25.47007104 No Never Smoked 2 members Father with cancer type unknown Son with lung cancer Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R3 pre NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Unknown Positive expression level NOS Moderate disease distribution greater than low but less than high No No None or microscopic No 1571 No evidence of disease Not lost 1571 Censored 1571 Alive 34 Adjuvant Chemotherapy No 0
64
+ 1948 -22417 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.48954397 Yes Never Smoked 6 members Father with lung cancer and liver cancer Sister 1 with gynecologic cancer nos at 40 died at 44 Sister 2 with lung cancer at 65 died at 67 Brother with BRCA positive prostate cancer Paternal Uncle with colorectal cancer Paternal Aunt with colon cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R2 OVARY RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 954 No evidence of disease Not lost 954 Censored 954 Alive 34 Adjuvant Chemotherapy No 0
65
+ 1949 -21991 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.49664697 No Never Smoked 3 members Mother with kidney cancer Father with prostate cancer Brother with bone cancer and lung cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DM R2 Ovary PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild Macroscopic < 1 cm Yes 1429 No evidence of disease Not lost 1429 Censored 1429 Alive 27 Adjuvant Chemotherapy No 0
66
+ 1949 -22841 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.54573027 Yes Previous Smoker 5 members Mother with lung cancer Brother with colorectal cancer Maternal Aunt with breast cancer Other Relative nos 1 with Breast Cancer Other Relative nos 2 with uterine cancer No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DL R0 OVARY RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS None or microscopic No 1493 Disease progression Died 527 527 Event 1493 Deceased Disease progression 2012 1493 47 Adjuvant Chemotherapy No 0
67
+ 1949 -22432 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.62408892 Yes Previous Smoker 2 members Sister 1 with ovarian cancer at 41 and breast cancer at 48 Sister 2 with ovarian cancer at 48 No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R3 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 828 No evidence of disease Not lost 926 Censored 926 Alive Neoadjuvant and Adjuvant Chemotherapy Yes 0
68
+ 1949 -21882 FEMALE Caucasian or White Non-Hispanic or Non-Latina 25.8912764 Yes Never Smoked 3 members Brother 1 with prostate cancer Sister with melanoma Brother 2 with non melanoma skin cancer Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the Fallopian tube DM R2 FALLOPIAN TUBE PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild Macroscopic < 1 cm Yes 1720 Disease progression Died 922 922 Event 1720 Deceased Disease progression 2013 1720 59 Adjuvant Chemotherapy No 0
69
+ 1950 -21322 FEMALE Caucasian or White Non-Hispanic or Non-Latina 26.3 Yes Never Smoked 1 member Aunt with uterine cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R2 pre NACT OVARY RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 1287 Disease progression Died 664 664 Event 1287 Deceased Disease progression 2013 1287 14 Neoadjuvant and Adjuvant Chemotherapy Yes 0
70
+ 1950 -22149 FEMALE Caucasian or white Non-Hispanic or Non-Latina 26.49345034 No Never Smoked 2 members Mother with lung cancer Maternal Aunt with breast cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 OVARY RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 1662 Residual or persistent disease no progression Not lost 1141 1141 Event 1662 Alive 13 Neoadjuvant and Adjuvant Chemotherapy Yes 0
71
+ 1950 -22506 FEMALE Caucasian or White Non-Hispanic or Non-Latina 26.63 No Current Smoker 20 1 member Maternal Aunt with kidney cancer at unspecified age Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the peritoneum DH R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Macroscopic < 1 cm Yes 1709 No evidence of disease Not lost 1709 Censored 1709 Alive 32 Adjuvant Chemotherapy No 0
72
+ 1951 -22435 FEMALE Caucasian or White Non-Hispanic or Non-Latina 26.92492959 No Previous Smoker 4 members Brother with bone cancer and prostate cancer at 11 deceased, Brother with cancer site not specified at 64 Sister with lymphoma died of cancer Son with leukemia Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the Fallopian tube DH R1 post NACT OVARY LEFT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 1532 Residual or persistent disease no progression 1322 Alive 58 Adjuvant Chemotherapy No 0
73
+ 1951 -21114 FEMALE Black or African American Non-Hispanic or Non-Latina 27.19353957 No Never Smoked 1 member Sister with colorectal cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Measurable > 1 cm Yes 1320 Residual or persistent disease no progression Not lost 365 365 Event 1320 Alive 13 Neoadjuvant and Adjuvant Chemotherapy Yes 0
74
+ 1952 -21500 FEMALE Caucasian or white Non-Hispanic or Non-Latina 27.64808636 No Never Smoked No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Unknown Moderate disease distribution greater than low but less than high No No Measurable > 1 cm Yes 1824 Residual or persistent disease no progression Not lost 581 581 Event 1824 Alive 2 Neoadjuvant and Adjuvant Chemotherapy No 0
75
+ 1952 -21116 FEMALE Caucasian or White Non-Hispanic or Non-Latina 27.87426146 Unknown Previous Smoker 1 member Sister with kidney cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DM R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 88 Disease progression Died 88 88 Event 88 Deceased Unknown 2011 88 49 Adjuvant Chemotherapy No 0
76
+ 1953 -19965 FEMALE Caucasian or White Non-Hispanic or Non-Latina 28.1141101 No Never Smoked 3 members Father with breast cancer Mother with breast cancer Sister with breast cancer and ovarian cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R1 post NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Low expression Moderate disease distribution greater than low but less than high No Yes Mild Macroscopic < 1 cm Yes 991 Disease progression Died 535 535 Event 991 Deceased Unknown 2013 991 5 Neoadjuvant and Adjuvant Chemotherapy Yes 0
77
+ 1953 -20786 FEMALE Caucasian or White Non-Hispanic or Non-Latina 28.84845009 No Never Smoked 1 member Father with colorectal cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R3 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS None or microscopic No 1399 No evidence of disease Not lost 1066 1066 Event 1399 Alive 43 Adjuvant Chemotherapy No 0
78
+ 1953 -20941 FEMALE Caucasian or White Non-Hispanic or Non-Latina 28.92685467 No Never Smoked Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R2 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No No Measurable > 1 cm Yes 1756 Residual or persistent disease no progression Not lost 544 544 Event 1756 Alive 26 Adjuvant Chemotherapy No 0
79
+ 1953 -21538 FEMALE Caucasian or White Non-Hispanic or Non-Latina 28.9528752 Unknown Never Smoked 1 member Father with prostate cancer No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R2 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild None or microscopic No 352 Disease progression Died 202 202 Event 352 Deceased Disease progression 2012 352 31 Adjuvant Chemotherapy No 0
80
+ 1953 -21538 FEMALE Black or African American Non-Hispanic or Non-Latina 29.12905234 No Never Smoked 1 member Mother with breast cancer Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive Measurable > 1 cm Yes 717 Residual or persistent disease no progression Not lost 389 389 Event 717 Alive 78 Adjuvant Chemotherapy No 0
81
+ 1953 -21318 FEMALE Caucasian or White Non-Spanish / Non-Hispanic / Non-Latina 29.49942579 Yes Never Smoked Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the peritoneum DM R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 990 Disease progression Died 381 381 Event 990 Deceased Disease progression 2013 990 21 Adjuvant Chemotherapy No 0
82
+ 1953 -20550 FEMALE Caucasian or White Non-Hispanic or Non-Latina 29.50592237 No Never Smoked 1 member Father with prostate cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R2 OVARY RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 1478 Residual or persistent disease no progression Not lost 770 770 Event 1478 Alive 37 Adjuvant Chemotherapy No 0
83
+ 1953 -21048 FEMALE Caucasian or White Hispanic or Latina 30.13932852 No Never Smoked 1 member Mother with breast cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown Moderate disease distribution greater than low but less than high No Yes Mild None or microscopic No 1290 Residual or persistent disease no progression Not lost 753 753 Event 1290 Alive 24 Adjuvant Chemotherapy No 0
84
+ 1953 -21048 FEMALE Caucasian or White Non-Hispanic or Non-Latina 30.22796383 No Never Smoked 1 member Father with lung cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R2 post NACT OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 1597 Residual or persistent disease no progression Not lost 422 422 Event 1597 Alive 63 Adjuvant Chemotherapy No 0
85
+ 1954 -20911 FEMALE Caucasian or White Non-Spanish / Non-Hispanic / Non-Latina 30.32366608 No Never Smoked Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 769 Disease progression Died 376 376 Event 769 Deceased Both disease progression and cancer treatment 2014 769 36 Adjuvant Chemotherapy No 0
86
+ 1954 -20375 FEMALE Caucasian or White Non-Hispanic or Non-Latina 30.58409897 No Never Smoked 1 member Mother with breast cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DM R0 OVARY LEFT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No No Macroscopic < 1 cm Yes 785 No evidence of disease Alive 1 to 2 years follow up then lost 785 Censored 785 Alive 27 Adjuvant Chemotherapy No 0
87
+ 1954 -19927 FEMALE Caucasian or White Non-Hispanic or Non-Latina 30.89261781 Unknown Never Smoked 1 member Father with cancer type unknown Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 1258 Disease progression Died 411 411 Event 1258 Deceased Disease progression 2013 1258 14 Neoadjuvant and Adjuvant Chemotherapy Yes 0
88
+ 1954 -20002 FEMALE Caucasian or White Non-Hispanic or Non-Latina 31.1809005 No Never Smoked 3 members Father with basal cell cancer of the skin at 71 Maternal Grandmother with vulvar cancer Paternal Grandmother with liver cancer No High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild None or microscopic No 1658 No evidence of disease Not lost 841 841 Event 1658 Alive 21 Neoadjuvant and Adjuvant Chemotherapy No 0
89
+ 1954 -21262 FEMALE Caucasian or White Non-Hispanic or Non-Latina 31.19638668 Unknown Never Smoked 1 member Paternal Grandmother with throat cancer Yes Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the peritoneum DH R1 UTERINE MYOMETRIUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 1911 No evidence of disease Not lost 1911 Censored 1911 Alive 40 Adjuvant Chemotherapy No 0
90
+ 1954 -20241 FEMALE Mixed Non-Hispanic or Non-Latina 31.22399686 No Never Smoked 3 members Mother with breast cancer Father with prostate cancer and non melanoma cancer Brother with prostate cancer No Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DL R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Measurable > 1 cm Yes 2325 Residual or persistent disease no progression Not lost 650 650 Event 2325 Alive 78 Adjuvant Chemotherapy No 0
91
+ 1954 -20079 FEMALE Caucasian or White Non-Hispanic or Non-Latina 31.30501386 Unknown Never Smoked 1 member Father with prostate cancer Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the Fallopian tube DH R2 pre NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild None or microscopic No 931 No evidence of disease Not lost 801 801 Event 1014 Alive 45 Adjuvant Chemotherapy No 0
92
+ 1955 -20338 FEMALE Caucasian or White Non-Spanish / Non-Hispanic / Non-Latina 31.609375 Yes Never Smoked 2 members Mother with squamous cell and basal cell cancer of the skin Cousin with colon cancer in 60s Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R2 OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Measurable > 1 cm Yes 1353 Disease progression Died 455 455 Event 1353 Deceased Disease progression 2012 1353 9 Neoadjuvant and Adjuvant Chemotherapy Yes 0
93
+ 1955 -20328 FEMALE Caucasian or White Non-Hispanic or Non-Latina 31.75071682 No Never Smoked 1 member Mother with breast cancer and colon or colorectal cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Unknown Unknown Moderate disease distribution greater than low but less than high No No Measurable > 1 cm Yes 660 Disease progression Died 171 171 Event 660 Deceased Both disease progression and cancer treatment 2011 660 14 Neoadjuvant and Adjuvant Chemotherapy Yes 0
94
+ 1955 -20500 FEMALE Caucasian or White Non-Hispanic or Non-Latina 31.91484375 Unknown Never Smoked 1 member Mother with pancreatic cancer at 80 Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the Fallopian tube DM R1 FALLOPIAN TUBE RIGHT PRIMARY IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 1165 No evidence of disease Not lost 1290 Censored 1290 Alive 43 Adjuvant Chemotherapy No 0
95
+ 1955 -20083 FEMALE Caucasian or White Non-Spanish / Non-Hispanic / Non-Latina 32.09560038 Yes Never Smoked Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the peritoneum DM R1 UTERUS METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 562 Residual or persistent disease no progression Alive 1 to 2 years follow up then lost 562 Censored 562 Alive 53 Adjuvant Chemotherapy No 0
96
+ 1955 -20262 FEMALE Caucasian or White Non-Hispanic or Non-Latina 32.25107042 No Never Smoked 1 member Mother with breast Cancer Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R2 post NACT OMENTUM METASTATIC IIIC Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No No None or microscopic No 1446 Disease progression Died 613 613 Event 1446 Deceased Disease progression 2012 1446 56 Adjuvant Chemotherapy No 0
97
+ 1956 -20415 FEMALE Caucasian or White Non-Hispanic or Non-Latina 32.80259733 Yes Previous Smoker Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R3 OVARY RIGHT PRIMARY IV Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 298 Disease progression Died 177 177 Event 298 Deceased Disease progression 2009 298 30 Adjuvant Chemotherapy No 0
98
+ 1956 -20217 FEMALE Caucasian or White Non-Hispanic or Non-Latina 32.91106072 No Never Smoked 2 members Father with skin cancer Sister with breast cancer Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the Fallopian tube DM R1 post NACT OMENTUM METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No Yes Mild Macroscopic < 1 cm Yes 1737 Residual or persistent disease no progression Not lost 955 955 Event 1737 Alive 30 Adjuvant Chemotherapy No 0
99
+ 1956 -19789 FEMALE Caucasian or White Non-Hispanic or Non-Latina 33.1998818 No Never Smoked Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R3 OMENTUM METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 923 Disease progression Died 905 905 Event 923 Deceased Disease progression 2012 923 No 0
100
+ 1957 -18914 FEMALE Caucasian or White Non-Hispanic or Non-Latina 33.2863569 Yes Never Smoked 2 members Father with prostate cancer Paternal Grandmother with cancer type unknown Yes Unknown Advanced high grade serous cancer of the Fallopian tube DH R1 OMENTUM METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis None or microscopic 1075 No evidence of disease Not lost 1075 Censored 1075 Alive 16 Adjuvant Chemotherapy No 0
101
+ 1957 -19279 FEMALE Caucasian or White Non-Hispanic or Non-Latina 34.26555808 No Never Smoked 1 member Mother with breast cancer Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the Fallopian tube DH R1 CUL DE SAC METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis No No None or microscopic No 34 No evidence of disease Alive less than 1 year follow up with progression missing date 216 216 Alive 28 Adjuvant Chemotherapy No 0
102
+ 1957 -19303 FEMALE Caucasian or White Non-Hispanic or Non-Latina 34.28814169 Unknown Never Smoked 4 members Mother with colorectal cancer Brother with cancer type unknown Maternal Uncle with cancer type unknown Other Relative nos with colorectal cancer Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS None or microscopic No 81 Disease progression Died 49 49 Event 81 Deceased Disease progression 2011 81 50 Adjuvant Chemotherapy No 0
103
+ 1958 -19358 FEMALE Caucasian or White Non-Hispanic or Non-Latina 34.48490921 No Never Smoked Yes Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R0 post NACT PELVIS METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild None or microscopic No 686 Recurrence new lesion Alive 2 to 3 years follow up then lost 686 Alive 18 Neoadjuvant and Adjuvant Chemotherapy Yes 0
104
+ 1958 -19724 FEMALE Caucasian or White Non-Hispanic or Non-Latina 34.95722144 No Never Smoked 2 members Mother with cervical cancer, Brother with brain cancer Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DM R1 post NACT OMENTUM METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Mild Macroscopic < 1 cm Yes 354 Disease progression Died 354 Censored 354 Deceased Unknown 2012 354 34 Adjuvant Chemotherapy No 0
105
+ 1958 -18997 FEMALE Caucasian or White Non-Hispanic or Non-Latina 35.27690173 No Never Smoked 4 members Mother with esophageal cancer Sister with esophageal cancer Sister with cervical cancer Son with prostate cancer Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the peritoneum DH R0 post NACT ENDOMETRIUM METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Moderate disease distribution greater than low but less than high No No None or microscopic No 912 Recurrence new lesion Alive 2 to 3 years follow up then lost 912 912 Event 912 Alive 20 Adjuvant Chemotherapy No 0
106
+ 1958 -19121 FEMALE Caucasian or white Non-Hispanic or Non-Latina 35.80307382 Unknown Previous Smoker 20 2 members Mother with ovarian cancer at 70 Maternal Aunt with breast cancer in 80s Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DM R1 UTERUS METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Mutant Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Non-measurable > 1 cm Yes 1659 Residual or persistent disease no progression Not lost 194 194 Event 1659 Alive 53 Adjuvant Chemotherapy No 0
107
+ 1959 -19253 FEMALE Caucasian or White Non-Hispanic or Non-Latina 36.14079497 No Never Smoked Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the Fallopian tube DH R1 post NACT OMENTUM METASTATIC IVA Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 119 No evidence of disease Alive less than 1 year follow up then lost 119 Censored 119 Alive 17 Adjuvant Chemotherapy No 0
108
+ 1960 -18068 FEMALE Caucasian or white Non-Hispanic or Non-Latina 36.98671084 No Never Smoked 1 member Father with cancer type unknown Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Macroscopic < 1 cm Yes 1073 Disease progression Died 583 583 Event 1073 Deceased Disease progression 2011 1073 31 Adjuvant Chemotherapy No 0
109
+ 1960 -18392 FEMALE Caucasian or White Non-Hispanic or Non-Latina 38.625 Unknown Never Smoked 1 relative Paternal Aunt with breast cancer No Low Disease Distribution, limited to the pelvis or retroperitoneal nodal metastasis Advanced high grade serous cancer of the ovary DH R2 OVARY PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive None or microscopic No 1578 No evidence of disease Not lost 569 569 Event 1578 Alive 16 Neoadjuvant and Adjuvant Chemotherapy Yes 0
110
+ 1960 -18616 FEMALE Caucasian or White Non-Spanish / Non-Hispanic / Non-Latina 38.73584423 No Never Smoked 1 member Sister with ovarian cancer and stomach cancer High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DM R2 UTERUS METASTATIC IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extent NOS Measurable > 1 cm Yes 1081 Disease progression Died 764 764 Event 1081 Deceased Disease progression 2011 1081 46 Adjuvant Chemotherapy No 0
111
+ 1961 -17212 FEMALE Caucasian or White Non-Hispanic or Non-Latina 38.94824893 No Never Smoked 3 members Mother with uterine cancer in 50s died of cancer at 58 Maternal Aunt 1 with uterine cancer in 70s Maternal Aunt 2 with breast cancer in 70s High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS Yes Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 612 Disease progression Died 470 470 Event 612 Deceased Disease progression 2011 612 127 Adjuvant Chemotherapy No 0
112
+ 1961 -18124 FEMALE Caucasian or White Non-Hispanic or Non-Latina 41.30968293 No Previous Smoker 3 members Mother with skin cancer Maternal Aunt with breast cancer Paternal Aunt with colon cancer Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the Fallopian tube DH R0 DIAPHRAGM METASTATIC IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Low disease distribution limited to the pelvis or retroperitoneal nodal metastasis No No None or microscopic No 864 Disease progression Died 384 384 Event 864 Deceased Disease progression 2011 864 28 Adjuvant Chemotherapy No 0
113
+ 1962 -17290 FEMALE Black or African American Non-Hispanic or Non-Latina 42.5234375 No Current Smoker 2 High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the Fallopian tube DH R0 OVARY METASTATIC IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Macroscopic < 1 cm Yes 725 Disease progression Died 667 667 Event 725 Deceased Disease progression 2012 725 27 Adjuvant Chemotherapy No 0
114
+ 1962 -17960 FEMALE Caucasian or White Non-Hispanic or Non-Latina 42.64071833 No Never Smoked Disease Beyond the Abdomen and Pelvis Advanced high grade serous cancer of the ovary DH R1 OVARY PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown Moderate disease distribution greater than low but less than high No Yes Mild None or microscopic No 1880 Disease progression Not lost 789 789 Event 1989 Alive 31 Adjuvant Chemotherapy No 0
115
+ 1962 -16777 FEMALE Caucasian or White Non-Hispanic or Non-Latina 43.09869731 No Never Smoked Yes High Disease Distribution in the diaphragm, gallbladder, epigastrium, liver and/or spleen Advanced high grade serous cancer of the ovary DH R0 OVARY PRIMARY IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No None or microscopic No 1423 No evidence of disease Alive 3 to 4 years follow up then lost 1423 Censored 1423 Alive 43 Adjuvant Chemotherapy No 0
116
+ 1963 -17373 FEMALE Black or African American Non-Spanish / Non-Hispanic / Non-Latina 43.48031157 No Never Smoked 2 members Mother with breast cancer at 65 Father with prostate cancer No Moderate Disease Distribution greater than Low, but less than High Advanced high grade serous cancer of the ovary DH R3 pre NACT PLEURAL IMPLANT METASTATIC IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Negative expression Moderate disease distribution greater than low but less than high No No None or microscopic No 505 Disease progression Died 421 421 Event 505 Deceased Disease progression 2010 505 3 Neoadjuvant Chemotherapy Yes 0
117
+ 1969 -15054 FEMALE Caucasian or White Non-Hispanic or Non-Latina 49.59697498 No Never Smoked 5 members Paternal grandmother diagnosed with breast cancer, Second cousin diagnosed with ovarian cancer, Second cousin diagnosed with ovarian cancer, Father diagnosed with lung cancer, Brother diagnosed with lung cancer Advanced high grade serous cancer of the ovary DM R0 post NACT METASTATIC SITE NOS METASTATIC IVB Serous adenocarcinoma SEROUS ADENOCARCINOMA High grade NOS No Unknown Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes Yes Extensive None or microscopic No 749 Residual or persistent disease no progression Not lost 568 568 Event 749 Alive 31 Adjuvant Chemotherapy No 0
118
+ 1972 -13684 FEMALE Caucasian or White Non-Hispanic or Non-Latina 52.79704179 No Never Smoked Advanced low grade serous cancer of the peritoneum DL R0 OMENTUM METASTATIC Serous adenocarcinoma SEROUS ADENOCARCINOMA Low grade NOS No Wild Type Unknown High disease distribution in the diaphragm, gall bladder, epigastrium, liver and/ or spleen Yes No Measurable > 1 cm Yes 930 Disease progression Died 930 Censored 930 Deceased Unknown 2013 930 55 Adjuvant Chemotherapy No 0
samples/APOLLO-2-leaderboard_meta.csv ADDED
@@ -0,0 +1,45 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ dtype,count,unique,null,freq,min,max,PV_match,Enumerated,NonEnumerated
2
+ year_of_birth,int64,117,35,0,,1927.0,1972.0,0.0,False,True
3
+ days_to_birth,int64,117,115,0,,-30246.0,-13684.0,,False,True
4
+ gender,object,117,1,0,117,,,1.0,True,False
5
+ race,object,117,5,0,103,,,1.0,True,False
6
+ ethnicity,object,117,3,0,106,,,0.0,False,True
7
+ bmi,float64,117,117,0,,16.65625,52.79704179,,False,True
8
+ alcohol_history,object,117,3,0,55,,,1.0,True,False
9
+ alcohol_intensity,float64,40,17,77,,0.0,14.0,,False,True
10
+ smokestat_apollo2,object,116,4,1,73,,,0.6666666666666666,False,True
11
+ cigarettes_per_day2,float64,32,10,85,,0.5,60.0,,False,True
12
+ years_smoked2,float64,32,21,85,,1.0,55.0,,False,True
13
+ family_cancer_history,object,91,81,26,5,,,,False,True
14
+ dzextent_beyondabd,object,54,3,63,40,,,1.0,True,False
15
+ dzextentpriortosurg,object,115,6,2,38,,,0.2,False,True
16
+ primary_diagnosis,object,117,39,0,15,,,0.0,False,True
17
+ site_of_resection_or_biopsy,object,117,18,0,50,,,0.6111111111111112,False,True
18
+ classification_of_tumor,object,117,2,0,68,,,1.0,True,False
19
+ stage_figo_2014,object,115,7,2,85,,,1.0,True,False
20
+ morphology,object,117,1,0,117,,,1.0,True,False
21
+ celltype,object,117,1,0,117,,,1.0,True,False
22
+ tumor_grade,object,117,4,0,90,,,0.5,False,True
23
+ sticpresent,object,115,3,2,95,,,1.0,True,False
24
+ brcastat,object,113,4,4,79,,,1.0,True,False
25
+ brca1ihc_apollo,object,113,5,4,108,,,0.25,False,True
26
+ disease_distribution,object,117,3,0,80,,,0.0,False,True
27
+ diseasehigh,object,114,3,3,79,,,1.0,True,False
28
+ miliarypresent,object,115,5,2,73,,,0.25,False,True
29
+ residual_disease,object,117,4,0,54,,,0.0,False,True
30
+ anyresidualdisease,object,114,3,3,63,,,1.0,True,False
31
+ days_to_last_known_disease_status,int64,117,115,0,,34.0,2325.0,,False,True
32
+ last_known_disease_status,object,117,4,0,59,,,0.5,False,True
33
+ fuflag,object,115,9,2,53,,,0.125,False,True
34
+ days_to_recurrence,float64,94,92,23,,30.0,1322.0,,False,True
35
+ pfstimeindays,float64,114,111,3,,30.0,1911.0,,False,True
36
+ pfs_status,object,113,3,4,91,,,1.0,True,False
37
+ survivaltimeindays,float64,114,112,3,,77.0,2325.0,,False,True
38
+ vital_status,object,117,2,0,63,,,1.0,True,False
39
+ causeofdeath_apollo2,object,53,4,64,45,,,0.6666666666666666,False,True
40
+ year_of_death,float64,54,7,63,,2009.0,2014.0,,False,True
41
+ days_to_death,float64,54,54,63,,77.0,1720.0,,False,True
42
+ days_to_treatment,float64,114,56,3,,2.0,127.0,,False,True
43
+ treatment_intent_type,object,116,4,1,81,,,0.6666666666666666,False,True
44
+ treatment_or_therapy,object,117,2,0,88,,,1.0,True,False
45
+ apollo2_adv_hg_serous_complete,int64,117,1,0,,0.0,0.0,1.0,True,True
samples/APOLLO-2.txt ADDED
@@ -0,0 +1,493 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ Clin Pharmacol Ther. 2019 Jul; 106(1): 52–57. Published online 2019
2
+ Apr 29. doi: 10.1002/cpt.1425 PMCID: PMC6617989 PMID: 30838639
3
+
4
+ From
5
+ Discovery to Practice and Survivorship: Building a National Real‐World
6
+ Data Learning Healthcare Framework for Military and Veteran Cancer
7
+ Patients Jerry S. H. Lee,corresponding author 1 , 2 , 3 , 4 , 5 , 6
8
+ Kathleen M. Darcy, 4 , 7 , 8 Hai Hu, 9 Yovanni Casablanca, 7 , 8
9
+ Thomas P. Conrads, 10 Clifton L. Dalgard, 11 , 12 John B. Freymann, 13
10
+ Sean E. Hanlon, 5 Grant D. Huang, 6 Leonid Kvecher, 9 George
11
+ L. Maxwell, 10 Frank Meng, 14 , 15 Joel T. Moncur, 16 Clesson Turner,
12
+ 17 Justin M. Wells, 18 Matthew D. Wilkerson, 4 , 11 , 12 Kangmin Zhu,
13
+ 8 Rachel B. Ramoni, 6 and Craig D. Shriver corresponding author 8 , 19
14
+ Author information Article notes Copyright and License information
15
+ Disclaimer
16
+
17
+ The Applied Proteogenomics OrganizationaL Learning and Outcomes
18
+ (APOLLO) network is implementing a prospective curation and
19
+ translation of real‐world data (RWD) into real‐world evidence (RWE)
20
+ within the learning healthcare environment of the Department of
21
+ Defense and Department of Veterans Affairs. To support basic,
22
+ translational, clinical, and epidemiological sciences, APOLLO will
23
+ release data to public repositories for secondary analysis to assist
24
+ others in assessing whether similar molecular‐driven clinical practice
25
+ guidelines will improve health outcomes for their relevant cancer
26
+ populations.
27
+
28
+ In the United States, > 80% of patients with cancer are initially
29
+ diagnosed and treated in a community hospital setting rather than an
30
+ academic hospital setting. Despite the increased adoption of
31
+ electronic health records (EHRs), the lack of interoperable health
32
+ information systems makes it challenging to aggregate RWD generated
33
+ from a cancer patient’s journey before diagnosis, during treatment,
34
+ and throughout survivorship. RWD might include data collected as part
35
+ of routine health and cancer care delivery or for research
36
+ (translational, implementation science, and/or epidemiological)
37
+ efforts. Longitudinal collection of RWD is essential to generating RWE
38
+ and is often absent when elucidating long‐term consequences of care
39
+ strategies.
40
+
41
+ Recent studies have demonstrated the success of individualized cancer
42
+ care strategies enabled by molecular profiling and targeted
43
+ therapies. In the past 2 years, the US Food and Drug Administration
44
+ (FDA) has approved tumor site–agnostic, biomarker‐driven cancer
45
+ treatments and next‐generation sequencing in vitro diagnostic
46
+ devices.1 A parallel review process by the Center for Medicare &
47
+ Medicaid Services led to a national coverage determination
48
+ next‐generation sequencing‐based in vitro diagnostics. The rapid
49
+ development and approval of such technologies underscored this
50
+ widening gap in capturing real‐world use of molecular‐driven cancer
51
+ care to generate RWE to help inform regulatory and clinical
52
+ decisions.2
53
+
54
+ Conducting valid real‐world studies requires data quality assurance
55
+ through auditable data abstraction methods and incentives to drive
56
+ electronic capture of data during delivery of care.2 The Department of
57
+ Veterans Affairs (VA) has the nation's largest integrated healthcare
58
+ system with over 9 million veterans enrolled and is a high‐volume
59
+ provider of cancer care with nearly 50,000 incident cancer cases
60
+ reported in 2010.3 The VA Office of Research and Development has as
61
+ its three major priorities to: (i) enhance veteran access to multisite
62
+ clinical trials, (ii) make VA data a national resource, and (iii)
63
+ increase the real‐world impact of research findings. The VA Office of
64
+ Research and Development's national Cooperative Studies Program4 and
65
+ data resources enable researchers to access and identify initial
66
+ cohorts for further studies to advance RWD analysis have been
67
+ leveraged through partnerships with federal collaborators to further a
68
+ learning health care system within the VA. The Department of Defense
69
+ (DoD) Military Health System (MHS) is responsible for maintaining the
70
+ health and readiness of 1.7 million active‐duty and reserve service
71
+ members (SMs) and caring for 9.4 million beneficiaries in TRICARE
72
+ health benefit plans. The John P. Murtha Cancer Center at Uniformed
73
+ Services University and Walter Reed National Military Medical Center
74
+ offers a comprehensive cancer care operational view in 64 capability
75
+ areas to proactively mitigate and close gaps in cancer care and
76
+ research in the MHS. The John P. Murtha Cancer Center utilizes
77
+ agreements with other federal agencies and extramural collaborators to
78
+ provide return on investment by deploying the most robust and modern
79
+ molecular technologies under various programs. The administrative and
80
+ medical care data from both direct and indirect care are stored in the
81
+ military data repository, which includes detailed information on
82
+ demographics, diagnoses, diagnostic procedures, prescriptions,
83
+ ancillary and radiology services, treatments, cost of care, and vital
84
+ status. The DoD also has a cancer registry that collects detailed data
85
+ on cancer diagnosis and features, including some cancer
86
+ biomarkers. These RWD have been widely used for cancer research among
87
+ DoD beneficiaries.5, 6
88
+
89
+ Leveraging the two largest nationwide connected healthcare systems,
90
+ the APOLLO network was launched in 2016 with the intent of curating
91
+ longitudinal RWD and health outcome data to create and assess adoption
92
+ of new molecular‐driven clinical practice guidelines. By developing,
93
+ defining, and aligning RWD elements of MHS, patients with cancer from
94
+ prediagnosis through survivorship among the federal and civilian
95
+ partners, the APOLLO network is implementing an integrated
96
+ multifederal network for prospective curation and translation of RWD
97
+ into RWE in a learning healthcare environment that will assist other
98
+ payers in assessing whether similar clinical practice guidelines will
99
+ improve health outcomes for their relevant populations.
100
+
101
+ MOVING TOWARD RWD: LESSONS LEARNED AND ONGOING PILOTS TO BUILD
102
+ THE APOLLO ECOSYSTEM Previous large‐scale tumor characterization
103
+ projects, such as The Cancer Genome Atlas and the ongoing Clinical
104
+ Proteomics Tumor Analysis Consortium, focused on analyzing the
105
+ genomics and proteomics profile of tumors at a single time point.7 The
106
+ lack of focus on longitudinal RWD collection limits the clinical
107
+ utilization of these programs’ data.8 APOLLO is distinct from The
108
+ Cancer Genome Atlas and other previous tumor characterization projects
109
+ as it was focused on integrated proteogenomic analyses, the collection
110
+ of longitudinal RWD, and development of a sustainable collection
111
+ pipeline from its inception. The foundation of the approach is a
112
+ network of biospecimen collection sites throughout the DoD and VA plus
113
+ select civilian sites. APOLLO tissue collection is infused into
114
+ pathology departments to preserve patient care, optimize collections,
115
+ and control for preanalytic variables while involving the local
116
+ organizations as true partners. This culture of collaboration also
117
+ promotes the capture of longitudinal clinical, radiology imaging, and
118
+ patient data throughout patients’ disease cycles that can otherwise be
119
+ difficult to obtain. This culture expands to Clinical Laboratory
120
+ Improvement Amendment (CLIA) laboratories, biobanking, imaging
121
+ characterization, and proteogenomic analysis centers to form a robust
122
+ APOLLO ecosystem that will be leveraged to enable additional
123
+ longitudinal oncology studies of both established and new patients.
124
+
125
+ To maximize longitudinal clinical data collection, APOLLO uniquely
126
+ designed a combination of disease‐specific pilot retrospective studies
127
+ of hundreds of cases (APOLLOs 1–4) and prospective studies of ~ 8,000
128
+ cases (APOLLO 5). Successes and lessons learned during the
129
+ implementation of these pilot projects, as well as those from past
130
+ large‐scale molecular and clinical studies, are being leveraged to
131
+ successfully forge the APOLLO ecosystem. Central to generating RWE
132
+ from RWD in combination with molecular data is the challenge of
133
+ balancing effective biospecimen matching and integration of data from
134
+ multiple modalities from the same patient while maintaining accuracy
135
+ and privacy over time. One way the network tackled this issue was
136
+ bringing together early stakeholders to develop and adopt a
137
+ prospectively generated unique APOLLO participant and aliquot
138
+ identifiers (APOLLO ID; Figure 1). APOLLO ID will also be linked to a
139
+ 128‐byte global unique participant and aliquot identifiers with an
140
+ “AP‐” prefix when data are uploaded to public repositories for
141
+ secondary analysis. The APOLLO system is electronically supported by
142
+ an enterprise informatics infrastructure, which includes a Data
143
+ Tracking System (DTS‐APOLLO) for transactional activities, a Data
144
+ Warehouse for Translational Research for (DW4TR‐APOLLO),9 and a
145
+ network of connected public data repositories to support capturing,
146
+ management, and delivery of RWD to the study team and the public to
147
+ enable discovery of RWE. Initial pilot datasets have been successfully
148
+ uploaded to the National Cancer Institute's Genomic Data Commons and
149
+ The Cancer Imaging Archive (TCIA) from both VA and DoD studies. The
150
+ length of patient follow‐up time within APOLLO will be pre‐estimated
151
+ for each cancer type using prior literature rather than by duration of
152
+ a funding cycle, so advanced planning will enable continued capturing
153
+ of such data from both the regulatory and technical perspectives.
154
+
155
+ An external file that holds a picture, illustration, etc. Object name
156
+ is CPT-106-52-g001.jpg Figure 1 Applied Proteogenomics OrganizationaL
157
+ Learning and Outcomes (APOLLO) data ecosystem and workflow to enable
158
+ longitudinal real‐world data (RWD) collection and analysis. Clinical
159
+ activities are separated from research functions by a firewall so that
160
+ only de identified, limited datasets are available for research and
161
+ further, only safe‐harbor datasets are made publicly
162
+ available. Patient will be followed from the time of diagnosis through
163
+ remission and when disease recurs, for as long as possible. Tracking
164
+ of all such RWD is enabled by APOLLO IDs in a program‐wide Data
165
+ Tracking System for APOLLO (DTS‐APOLLO). Activities in molecular
166
+ center are tracked by local LIMS with metadata and higher‐level
167
+ molecular data tracked in DTS‐APOLLO. Transactional data in DTS‐APOLLO
168
+ will be quality assured and integrated in the Data Warehouse for
169
+ Translational Research for APOLLO (DW4TR‐APOLLO) for integrated
170
+ analysis to generate real‐world evidence (RWE), which will in turn
171
+ directly impact patient clinical services. Lower‐level raw molecular
172
+ and imaging data of very large size, on the other hand, will be
173
+ directly uploaded to public data repositories, including The Cancer
174
+ Imaging Archive (TCIA),11 Genomic Data Commons (GDC),12 and upcoming
175
+ Proteomic Data Commons (PDC) maintained by the National Cancer
176
+ Institute (NCI) following appropriate protocols and regulatory
177
+ procedures coordinated through DW4TR‐APOLLO. Such raw data, after
178
+ integration with the data in the DW4TR‐APOLLO enabled by APOLLO ID,
179
+ will become substrates for integrated research analysis for hypothesis
180
+ generation and testing, which will be the basis for the design of new
181
+ scientific experiments and clinical trials with results will
182
+ eventually impact future patient clinical care. Solid lines are for
183
+ clinical‐grade RWD and dotted lines for research‐grade RWD. DoD,
184
+ Department of Defense; EHR, electronic health record; VA, Veteran's
185
+ Affairs.
186
+
187
+ LOOKING AHEAD: INITIAL EFFORTS TO ELEVATE RWD TO RWE The APOLLO
188
+ program aspires to accelerate the application of next‐generation
189
+ proteogenomic profiling with deep baseline and longitudinal RWD from
190
+ DoD and VA EHRs and research records into RWE for FDA‐approved tests
191
+ and treatments for development and deployment of tools and strategies
192
+ used in the prevention, diagnosis, and treatment of cancer. These
193
+ activities support readiness and health by empowering patients and
194
+ providers to optimize their care and health through customized and
195
+ enterprise solutions. The program will deploy both retrospective and
196
+ prospective observational designs with provisions for clinical trial
197
+ participation. Select civilian cohorts with aggressive or rare cancers
198
+ will be incorporated with SMs and veterans to contribute diversity,
199
+ events, experiences, and outcomes to the disease‐oriented and
200
+ pan‐cancer cohorts to learn about, treat, and prevent cancers that
201
+ develop in warfighters.
202
+
203
+ Types of clinical and research RWD that will be collected by the
204
+ APOLLO network are listed in Table 1. This program will require and
205
+ utilize operationalized processes and procedures tracked via a
206
+ user‐friendly APOLLO Dashboard. Integrated analyses will incorporate a
207
+ deep complement of RWD from medical and research records. Sequencing
208
+ and proteomic data generated by CLIA facilities and analytical core
209
+ facilities will not only be analyzed using current clinical databases
210
+ but will be available for iterative reanalysis over time applying new
211
+ clinical databases and trusted sources to advance reinterpretation of
212
+ the patients’ molecular profiling data to determine future access to
213
+ new FDA‐approved drugs and/or clinical trial opportunities. This
214
+ program will provide data in support studies of basic science,
215
+ translational medicine, epidemiology, comparative effectiveness,
216
+ cost‐effectiveness, and health disparities. Various data‐release
217
+ provisions were incorporated into the APOLLO framework, including
218
+ release to repositories for future research, clinical trials,
219
+ indications and guidelines, dissemination to scientists, healthcare
220
+ professionals, and the public, release to study doctors when research
221
+ results meet guidelines for medical consideration for follow‐up and
222
+ clinical assessments, and return to patients when the research results
223
+ qualifies for release without clinical certification, as recommended
224
+ recently by the National Academies of Sciences, Engineering, and
225
+ Medicine.10
226
+
227
+ Table 1 Types of RWD from medical and research records for APOLLO
228
+
229
+ Captured into smart electronic clinical reporting and XML forms with
230
+ data dictionaries, valid value requirements, logging features, and
231
+ business rules. Data elements are labeled with a unique coded APOLLO
232
+ ID participant identifier. Baseline data: Registration, eligibility,
233
+ consent, demographics, height, weight, risk factors, smoking status,
234
+ marital status, type of insurance, medical history, medications,
235
+ supplements, reproductive history, and family cancer history.
236
+ Surgical treatment: Surgical date, surgical procedures performed, AJCC
237
+ stage with edition details, and disease site–specific surgical
238
+ findings, including primary tumor size, disease distribution (location
239
+ and size pre/post surgery), residual disease status, military disease,
240
+ laterality, margins, redacted operative report(s), and comments.
241
+ Pathologic findings: Diagnosis date, definitive surgery date, ICD site
242
+ and behavior codes, detailed College of American Pathology electronic
243
+ cancer checklist13 with harmonized data dictionaries and conversion
244
+ between versions, redacted pathology reports, including cytologic
245
+ findings, clinical biomarker assessments, and other findings.
246
+ Case‐level data: Case organ type, lesion type, malignancy type,
247
+ primary site of diagnosis, ICD‐10 code, histology code, TNM edition
248
+ number, pathological group stage at diagnosis, CAP organ data creation
249
+ status, and biomarker creation status. Research pathology
250
+ characterization: Baseline and in‐depth research pathology
251
+ characterization will be provided and compared with the clinical
252
+ diagnosis for tumor samples by expert pathologists and tissue imaging
253
+ researchers. The types of annotation may include tissue composition
254
+ details, clinical biomarker staining, and computer‐generated
255
+ annotation in imaged slides with intact tumor tissues or tissues
256
+ before and after laser microdissection. Molecular data: Including
257
+ redacted report, primary findings, and secondary findings when
258
+ applicable from CLIA testing, clinical recommendations, clinical
259
+ actions taken and outcomes, and XML data from CLIA assays when
260
+ available implementing best practices and guidelines from the College
261
+ of American Pathology, American Society of Clinical Oncology, National
262
+ Comprehensive Cancer Network, and American College of Genetics and
263
+ Genomic for risk assessments, interpretation, certification, and
264
+ genetic counseling health conditions, including cancer. DoD uses the
265
+ Illumina TruSight Tumor 15 tumor profiling assay with plans to deploy
266
+ the TruSight Oncology 500 tumor profiling DNA + RNA assay. VA uses the
267
+ Personalis AC CancerPlus DNA + RNA assay to evaluate 181 clinically
268
+ actionable genes or the PGDx Cancer Select 125 assay. Research
269
+ analytical facilities generate next generation sequencing and multiple
270
+ proteomic data. Immunoassay, cell‐free DNA, metabolomic, glycoprotein,
271
+ and lipidomic data may be available in subsets. Clinical imaging: May
272
+ be acquired when accessible from medical records, imaging facilities,
273
+ and research records with regulatory approval and consent at a
274
+ baseline time point and as longitudinal series of collections to
275
+ monitor and document disease distribution patterns and features
276
+ utilizing enterprise solutions by the VA and customized solutions by
277
+ DoD programs in partnership with TCIA. Baseline details regarding
278
+ imaging, including method, contrast, facility location, and dates for
279
+ acquisition, curation, and submissions to and receipt of annotation.11
280
+ Disease‐oriented features will be annotated by expert radiologists
281
+ using custom workstation configuration and standardized data
282
+ dictionary, including assessments of mass: laterality, calcifications,
283
+ thick septations, internal architecture; disease: presence,
284
+ calcification, locations, shape; ascites or effusion: volume;
285
+ lymphadenopathy: pathologic lymph nodes. Computer‐generated features,
286
+ including but not limited to segmentation using machine learning and
287
+ artificial intelligence. Pharmacologic therapies: Pharmacologic
288
+ therapy status by regimen, treatment line, or indication with
289
+ individual agent details with drug name, ICD‐O cancer site for
290
+ treatment, doses, route/delivery method, cycles, date first dose/start
291
+ date, date last dose/end date, dose schedule, active medication, dose
292
+ reduction, treatment selection (approved assay or an integral,
293
+ integrated, or exploratory biomarker), best response, and serious
294
+ adverse events. FDA indication with companion diagnostic assays:
295
+ Non‐small cell lung cancer: Treat an EGFR exon 19 deletions or EGFR
296
+ exon 21 L858R alterations with afatinib, gefitinib, or erlotinib; an
297
+ EGFR exon 20 T790M alteration with osimertinib; ALK rearrangement with
298
+ alectinib, crizotinib, or ceritinib; BRAF V600E with dabrafenib and
299
+ trametinib. Melanoma: Treat BRAF V600E with dabrafenib or vemurafenib;
300
+ BRAF V600E or V600K with trametinib or cobimetinib with
301
+ vemurafenib. Breast cancer: Treat ERBB2/HER2 amplification with
302
+ trastuzumab, ado‐trastuzumab emtansine, or pertuzumab. Colorectal
303
+ cancer: Treat wild‐type KRAS (absence of mutations in codons 12 and
304
+ 13) with cetuximab; wild‐type KRAS (absence of mutations in exons 2,
305
+ 3, and 4) or wild‐type NRAS (absence of mutations in exons 2, 3, and
306
+ 4) with panitumumab. Ovarian cancer: Treat BRCA1/2 alterations with
307
+ rucaparib. Treatment of adult and pediatric patients with cancer with
308
+ an NTRK fusion, including solid tumors and hematologic malignancies
309
+ with larotrectinib. Radiotherapies: Radiotherapy status by location,
310
+ indication, radiation treatment line/regimen, laterality, field
311
+ treated, radiation site code (ICD‐O), start date, end date, number of
312
+ fractions, dose/fraction cGy, total dose cGy, best response, and best
313
+ response assessment method, and comments. Outcome assessments: If
314
+ living: Disease status (alive with disease, no evidence of disease),
315
+ date of last visit or date last activity if different than visit and
316
+ capture individual dates of recurrence or progression with assessment
317
+ method(s) and additional details when available. If deceased: Date of
318
+ death and cause of death (cancer‐related, noncancer related, and
319
+ unknown), if other cause then specify. Clinical trial participation
320
+ will also be documented. Epidemiologic data: May be provided directly
321
+ by patients or with research staff during interviews with patients
322
+ using a standardized data dictionary. Veterans may also contribute
323
+ data through the Million's Veterans Program. Patient demographics,
324
+ including race, ethnicity, sex, marital status, education, employment,
325
+ and military service. Medical history regarding health conditions,
326
+ prior cancer diagnoses and treatments, height, and weight. Physical
327
+ activity for 12 months prior to the current diagnosis. Alcohol history
328
+ in entire life and currently. Tobacco products use in entire life and
329
+ currently. Work environment, including occupations, exposures, and
330
+ deployments. Family cancer history for blood relatives, including half
331
+ blood relatives. Reproductive history for women. Patient‐reported
332
+ outcomes: Using validated instruments from trusted sources. Patient
333
+ Reported Outcomes Measurements for Personalizing Treatment (PROMPT
334
+ Assessments): Quality of life using the 28‐item FACT‐G for physical,
335
+ social/family, emotional, and functional well‐being. Global health
336
+ using the 10‐item PROMIS Global Health version 1.2 instrument. Pain
337
+ and fatigue using the 3‐item PROMIS Pain 3a and the 4‐item PROMIS
338
+ Fatigue 4a instruments. Stress, anxiety, and depression combination
339
+ using the 10‐item NIH ToolBox Perceived Stress, 4‐item PROMIS Anxiety
340
+ 4a, and 4‐item PROMIS Depression 4a instruments. Symptoms using the
341
+ 4‐item FACT‐NTX‐4, the 4‐item PROMIS Cognitive Function 4a, and the
342
+ 4‐item PROMIS Sleep Disturbance 4a instruments. Support for daily
343
+ living using the 11‐item PROMIS Instrumental Support version 2.0
344
+ instrument. Focus assessments using validated instruments from
345
+ trusted sources and working to deploy novel surveys to address gaps
346
+ and support prevention, survivorship, palliative and end‐of‐life care
347
+ to strengthen cancer capabilities across the continuum from
348
+ prevention, early detection, treatment selection, mitigation of
349
+ effects, rehabilitation, and survivorship, including palliative and
350
+ end‐of‐life care. This may include assessments of barriers to care,
351
+ patient preferences regarding treatment and care, resilience, cancer
352
+ pain management, young adult survivorship, and serious adverse event
353
+ reporting. Open in a separate window AJCC, American Joint Commission
354
+ on Cancer; ALK, anaplastic lymphoma kinase; APOLLO, Applied
355
+ Proteogenomics OrganizationaL Learning and Outcomes; BRAF, B‐type Raf;
356
+ BRCA, breast cancer; CAP, College of American Pathologists; cGy,
357
+ centigray; CLIA, Clinical Laboratory Improvement Amendment; DoD,
358
+ Department of Defense; EGFR, epidermal growth factor receptor; ERBB,
359
+ erythroblastic leukemia viral oncogene; FACT‐G, functional assessment
360
+ of cancer therapy general; FDA, US Food and Drug Administration; HER2,
361
+ human epidermal growth factor receptor 2; ICD‐10, International
362
+ Classification of Disease‐10th edition; ICD‐O, International
363
+ Classification of Disease for Oncology; KRAS, Kirsten RAt Sarcoma
364
+ virus; NTRK, Neurotrophic tropomyosin receptor kinase; PGDx, Personal
365
+ Genome Diagnostics; PROMIS, Patient‐Reported Outcomes Measurement
366
+ Information System; RWD, real‐world data; TCIA, The Cancer Imaging
367
+ Archive; TNM, Tumor, Node, Metastasis staging system; VA, Veteran's
368
+ Affairs.
369
+
370
+ Translation of RWD into RWE is a key component of APOLLO with
371
+ integrated systems for enhancing capabilities across the cancer care
372
+ continuum, driving efficiencies, and enhancing quality, thereby
373
+ improving health outcomes and the readiness of warfighters and the
374
+ operational medical force. The full potential of APOLLO will be
375
+ realized when interoperable EHRs are readily and securely exchangeable
376
+ across the DoD and VA with enterprise solutions and clinical decision
377
+ tools for molecular pathology, clinical imaging, patient‐reported
378
+ outcomes, clinical trials, serious adverse events reporting,
379
+ prevention clinics, rehabilitative and other supportive services, pain
380
+ management, survivorship, palliative care, end‐of‐life care, research,
381
+ and education.
382
+
383
+ RETURN ON INVESTMENT: LEVERAGING RWD AND RWE FOR DOD, VA, AND
384
+ THE GLOBAL CANCER ECOSYSTEM Improvements in readiness, health care,
385
+ and outcomes for SMs, veterans, health beneficiaries, and civilians
386
+ will be achieved not only from deliverables generated by the APOLLO
387
+ network but also from release of RWD and RWE to the public for
388
+ secondary research. APOLLO patients may also benefit from release of
389
+ research data that qualify either for clinical certification or direct
390
+ release based on criteria, such as level and quality of the
391
+ evidence. Federal agencies may also benefit from the generated
392
+ agreements, established working groups, and taskforces with
393
+ representation from the stakeholders and invited nonfederal experts,
394
+ aligned resources and assets, integrated and expanded infrastructure
395
+ and workforces, and the capabilities developed for APOLLO and
396
+ operationalized across the DoD and VA for implementing precision
397
+ oncology solutions to acquire and translate RWD from APOLLO into RWE
398
+ for SMs, veterans, and the global cancer ecosystem.
399
+
400
+ Funding Funding for these efforts was provided from Uniformed
401
+ Services University of the Health Sciences (USUHS) awards from the
402
+ Defense Health Program to the Murtha Cancer Center Research Program
403
+ (HU0001‐16‐2‐0014, C.D. Shriver and J.S.H. Lee), the Gynecologic
404
+ Cancer Center of Excellence (HU0001‐16‐2‐0006, Y. Casablanca and
405
+ G. Larry Maxwell), and HU0001‐16‐2‐004 (L. Kvecher and H. Hu)
406
+ administered by the Henry M. Jackson Foundation for the Advancement of
407
+ Military Medicine. This project has also been funded in whole or in
408
+ part with federal funds from the National Cancer Institute, National
409
+ Institutes of Health, under Contract No. HHSN261200800001E
410
+ (J.B. Freymann).
411
+
412
+ Conflict of Interest The authors declared no competing
413
+ interests for this work.
414
+
415
+ Disclaimer The contents of this publication are the sole
416
+ responsibility of the authors and do not necessarily reflect the
417
+ views, opinions, or policies of the USUHS, the Henry M. Jackson
418
+ Foundation for the Advancement of Military Medicine, Inc., the
419
+ Department of Defense (DoD), the Departments of the Army, Navy, or Air
420
+ Force, Department of Health and Human Services, or Department of
421
+ Veterans Affairs. Mention of trade names, commercial products, or
422
+ organization does not imply endorsement by the U.S. Government.
423
+
424
+ Acknowledgments The authors would like to thank Joseph Shaw,
425
+ Sara Sakura, Autumn Beemer Phillips, Gregory Samuel, Olga Castellanos,
426
+ Jillian Infusino, and Mayada Aljehani for their critical review of the
427
+ figure and paper.
428
+
429
+ Contributor Information Jerry S. H. Lee, Email:
430
+ ude.csu@yrrej.rd.
431
+
432
+ Craig D. Shriver, Email: lim.liam@vic.revirhs.d.giarc.
433
+
434
+ References
435
+ 1. Goldberg, K.B. , Blumenthal, G.M. & Pazdur,
436
+ R. The first year of the Food and Drug Administration Oncology Center
437
+ of Excellence: landmark approvals in a dynamic regulatory
438
+ environment. Cancer J. 24, 131–135 (2018). [PubMed] [Google Scholar]
439
+
440
+ 2. US Framework for FDA's Real‐World Evidence Program. US Food and
441
+ Drug Administration (FDA)
442
+ <https://www.fda.gov/downloads/ScienceResearch/SpecialTopics/RealWorldEvidence/UCM627769.pdf>
443
+ (2018). Accessed February 12, 2019.
444
+
445
+ 3. Zullig, L.L. et al Cancer incidence among patients of the
446
+ U.S. Veterans Affairs Health Care System: 2010 update. Mil. Med. 182,
447
+ e1883–e1891 (2017). [PMC free article] [PubMed] [Google Scholar]
448
+
449
+ 4. Huang, G.D. , Ferguson, R.E. , Peduzzi, P.N. & O'Leary,
450
+ T.J. Scientific and organizational collaboration in comparative
451
+ effectiveness research: the VA cooperative studies program
452
+ model. Am. J. Med. 123, e24–e31 (2010). [PubMed] [Google Scholar]
453
+
454
+ 5. Shao, S. et al Diabetes diagnosis and overall survival among breast
455
+ cancer patients in the Military Health System. Cancer
456
+ Epidemiol. Biomarkers Prev. 27, 50–57 (2018). [PMC free article]
457
+ [PubMed] [Google Scholar]
458
+
459
+ 6. Manjelievskaia, J. , Brown, D. , McGlynn, K.A. , Anderson, W. ,
460
+ Shriver, C.D. & Zhu, K. Chemotherapy use and survival among young and
461
+ middle‐aged patients with colon cancer. JAMA Surg. 152, 452–459
462
+ (2017). [PMC free article] [PubMed] [Google Scholar]
463
+
464
+ 7. Hutter, C. & Zenklusen, J.C. The Cancer Genome Atlas: creating
465
+ lasting value beyond its data. Cell, 173, 283–285 (2018). [PubMed]
466
+ [Google Scholar]
467
+
468
+ 8. Liu, J. et al An integrated TCGA pan‐cancer clinical data resource
469
+ to drive high‐quality survival outcome analytics. Cell 173, 400–416
470
+ (2018). [PMC free article] [PubMed] [Google Scholar]
471
+
472
+ 9. Hu, H. et al DW4TR: a data warehouse for translational
473
+ research. J. Biomed. Inform. 44, 1004–1019 (2011). [PubMed] [Google
474
+ Scholar]
475
+
476
+ 10. Returning individual research results to participants: guidance
477
+ for a new research paradigm. National Academies of Sciences,
478
+ Engineering, and Medicine
479
+ <http://nationalacademies.org/hmd/Reports/2018/returning-individual-research-results-to-participants.aspx>
480
+ (2018). Accessed February 12, 2019.
481
+
482
+ 11. APOLLO‐1‐VA pilot lung cancer study data in The Cancer Imaging
483
+ Archive (TCIA)
484
+ <https://wiki.cancerimagingarchive.net/display/Public/APOLLO-1-VA>
485
+ (2018). Accessed February 12, 2019.
486
+
487
+ 12. APOLLO‐1‐VA pilot lung cancer study data in Genomic Data Commons
488
+ (GDC) <https://portal.gdc.cancer.gov/projects/VAREPOP-APOLLO>
489
+ (2018). Accessed February 12, 2019.
490
+
491
+ 13. College of American Pathologists: cancer protocol templates
492
+ <http://www.cap.org/cancerprotocols> (2019). Accessed February 12,
493
+ 2019.
samples/APOLLO-2_description.txt ADDED
@@ -0,0 +1,21 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ The Applied Proteogenomics Organizational Learning and Outcomes
2
+ (APOLLO) network is a collaboration between NCI, the Department of
3
+ Defense (DoD), and the Department of Veterans Affairs (VA) to
4
+ incorporate proteogenomics into patient care as a way of looking
5
+ beyond the genome, to the activity and expression of the proteins that
6
+ the genome encodes. The emerging field of proteogenomics aims to
7
+ better predict how patients will respond to therapy by screening their
8
+ tumors for both genetic abnormalities and protein information, an
9
+ approach that has been made possible in recent years due to advances
10
+ in proteomic technology.
11
+
12
+ The APOLLO network collects data that include a full set of medical
13
+ images, including CT and MRI scans, obtained before and during
14
+ treatment. Each set of images can be connected to the patient’s
15
+ genomic, proteomic, and clinical data. The real data will be made
16
+ available publicly through the Genomic Data Commons, Proteomics Data
17
+ Commons, and Cancer Imaging Archive. By using all available data,
18
+ researchers will be able to study the relationships among these data,
19
+ validate results, and develop predictive and prognostic models to
20
+ improve patient care. Aligning this data with the NCI Cancer Research
21
+ Data Commons is currently a time intensive manual effort.
samples/APOLLO-5-leaderboard.tsv ADDED
@@ -0,0 +1,2 @@
 
 
 
1
+ race ethnicity prior_malignancy alcohol_history smokestat_apollo2 any_family_cancer_history dxextent_prenact dzextent_beyondabd dzextentpriortosurg site_code stage_figo_2014 tumor_stage celltype tumor_grade sticpresent intraepilocation disease_distribution diseasehigh miliarypresent residual_disease anyresidualdisease last_known_disease_status progression_or_recurrence survival_status vital_status causeofdeath_apollo2 dtdatalocked project_id submitter_id encounter date_of_consent year_of_birth days_to_birth age_at_diagnosis gender specify_prior_malignancy height weight bmi alcohol_intensity cigarettes_per_day2 years_smoked2 family_cancer_history primary_diagnosis classification_of_tumor morphology brcastat pfstimeindays pfs_status survivaltimeindays year_of_death days_to_death days_to_treatment therapeutic_agents treatment_intent_type treatment_or_therapy dtform_apollo2 form_by_apollo2 dt_amend_apollo2 form_aby_apollo2 brca1ihc_apollo days_to_last_follow_up days_to_last_known_disease_status fuflag days_to_recurrence
2
+ 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1
samples/Apollo-5-headings.txt ADDED
@@ -0,0 +1 @@
 
 
1
+ race, ethnicity, prior_malignancy, alcohol_history, smokestat_apollo2, any_family_cancer_history, dxextent_prenact, dzextent_beyondabd, dzextentpriortosurg, site_code, stage_figo_2014, tumor_stage, celltype, tumor_grade, sticpresent, intraepilocation, disease_distribution, diseasehigh, miliarypresent, residual_disease, anyresidualdisease, last_known_disease_status, progression_or_recurrence, survival_status, vital_status, causeofdeath_apollo2, dtdatalocked, project_id, submitter_id, encounter, date_of_consent, year_of_birth, days_to_birth, age_at_diagnosis, gender, specify_prior_malignancy, height, weight, bmi, alcohol_intensity, cigarettes_per_day2, years_smoked2, family_cancer_history, primary_diagnosis, classification_of_tumor, morphology, brcastat, pfstimeindays, pfs_status, survivaltimeindays, year_of_death, days_to_death, days_to_treatment, therapeutic_agents, treatment_intent_type, treatment_or_therapy, dtform_apollo2, form_by_apollo2, dt_amend_apollo2, form_aby_apollo2, brca1ihc_apollo, days_to_last_follow_up, days_to_last_known_disease_status, fuflag, days_to_recurrence
samples/Outcome-Predictors-leaderboard.csv ADDED
@@ -0,0 +1,46 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ ,Round,TCGAID,MajorAxisLength,MinorAxisLength,PropnCET,T1FLAIR,nCETCrossMidline,rCBVmean,rCBVmax,rCBVfl,Machine,Subtype_by_phils,Subtype_by_Verhaak,age_at_initial_pathologic_diagno,date_of_initial_pathologic_diagn,days_to_birth,days_to_death,days_to_last_followup,ethnicity,gender,histological_type,karnofsky_performance_score,performance_status_scale_timing,person_neoplasm_cancer_status,prior_glioma,race,tumor_histologic_subtype,tumor_tissue_site,vital_status,days_to_tumor_recurrence,time_to_progression,progEvent,progSurvTime,OStime,rCBVmeanSD,rCBVmaxSD,rCBVflSD,rCBVmean2,rCBVmax2,rCBVfl2,age5,cohort,gcimp,egfrAmp,pdgfraAmp,ptenAmp,egfrMut,ptenMut,sequenced,Extent_of_Resection
2
+ 0,1,TCGA-06-0122,71.47023114,40.43448767,None (0%),Mixed T1<FLAIR Ratio,Crosses Midline No,6.86,9.483,1.467,3T,Mes,Mesenchymal,84,2006-00-00,-30967,181,8,NOT HISPANIC OR LATINO,FEMALE,,,,,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,181,181,7.0280743631,4.8061079943,2.9471538976,high,high,high,16.8,6,NEG,2.0,0.0,-1.0,,,1,STR
3
+ 1,1,TCGA-06-0127,86.97233129,55.44624428,6-33%,Mixed T1<FLAIR Ratio,Crosses Midline Yes,4.243,5.973,1.177,1.5T,PN,Neural,67,2002-00-00,-24502,121,108,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,60.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,90.0,1,90,121,4.3469561986,3.0271942476,2.3645536043,high,high,high,13.4,2,NEG,2.0,0.0,-1.0,"V292L,V292L,V292L",,1,STR
4
+ 2,2,TCGA-06-0128,68.54275415,55.25454465,34-67%,Expansive T1~FLAIR Ratio,Crosses Midline No,1.388,2.83,0.713,1.5T,PN,Proneural,66,2000-00-00,-24217,691,691,NOT HISPANIC OR LATINO,MALE,,80.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,189.0,1,189,691,1.4220068828,1.4342808841,1.432393135,low,low,low,13.2,0,POS,0.0,0.0,-1.0,,,1,GTR
5
+ 3,1,TCGA-06-0132,90.38251662,62.26697312,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.173,6.127,0.837,1.5T,PN,Neural,49,2007-00-00,-18125,761,570,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,482.0,1,482,761,2.2262398821,3.1052434547,1.6815049845,low,high,high,9.8,7,NEG,2.0,0.0,-1.0,,,1,STR
6
+ 4,1,TCGA-06-0133,102.8050346,62.89969015,34-67%,Mixed T1<FLAIR Ratio,Crosses Midline Yes,2.435,4.663,0.553,3T,PN,Neural,64,2007-00-00,-23402,435,428,NOT HISPANIC OR LATINO,MALE,,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,78.0,1,78,435,2.4946590487,2.363269174,1.1109584904,high,high,low,12.8,7,NEG,2.0,0.0,-1.0,,,1,STR
7
+ 5,1,TCGA-06-0137,101.8218393,56.31768036,None (0%),Mixed T1<FLAIR Ratio,Crosses Midline No,2.53,4.317,0.717,1.5T,Mes,Classical,63,2003-00-00,-23273,812,701,NOT HISPANIC OR LATINO,FEMALE,,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,487.0,1,487,812,2.5919866091,2.1879118645,1.4404290011,high,low,low,12.6,3,NEG,2.0,0.0,-1.0,P596L,,1,STR
8
+ 6,1,TCGA-06-0139,80.6709788,39.04821797,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.112,4.497,1.24,3T,Mes,Mesenchymal,40,2006-00-00,-14728,383,327,NOT HISPANIC OR LATINO,MALE,,60.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,152.0,1,152,383,2.1637453433,2.2791382105,2.4911184956,low,high,high,8.0,6,NEG,1.0,0.0,-1.0,,,1,STR
9
+ 7,1,TCGA-06-0143,114.8301745,65.62670418,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline Yes,2.393,5.657,0.353,3T,Mes,Mesenchymal,58,2006-00-00,-21386,357,357,NOT HISPANIC OR LATINO,MALE,,60.0,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,264.0,264.0,1,264,357,2.451630022,2.8670413291,0.7091651846,low,high,low,11.6,6,NEG,2.0,2.0,-1.0,,,1,STR
10
+ 8,1,TCGA-06-0147,74.49334479,51.14346067,None (0%),Expansive T1~FLAIR Ratio,Crosses Midline No,2.24,4.76,0.865,1.5T,Mes,Mesenchymal,51,1999-00-00,-18742,541,508,NOT HISPANIC OR LATINO,FEMALE,,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,92.0,1,92,541,2.2948814247,2.4124300383,1.7377560473,low,high,high,10.2,-1,NEG,1.0,0.0,-1.0,,,1,STR
11
+ 9,1,TCGA-06-0149,90.87360905,48.56049078,68-95%,Infiltrative T1<<FLAIR Ratio,Crosses Midline Yes,2.51,3.99,2.057,3T,Mes,Mesenchymal,74,2005-00-00,-27315,262,238,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,80.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,203.0,1,203,262,2.5714965964,2.0221840027,4.1324441495,high,low,high,14.8,5,NEG,1.0,0.0,-1.0,,,0,STR
12
+ 10,1,TCGA-06-0164,66.76339735,46.13628674,None (0%),Expansive T1~FLAIR Ratio,Crosses Midline No,1.768,4.363,0.607,1.5T,Mes,Mesenchymal,47,2000-00-00,-17510,1730,1729,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,100.0,Pre-Operative,WITH TUMOR,NO,ASIAN,Glioblastoma multiforme,BRAIN,DECEASED,,1428.0,1,1428,1730,1.8113171245,2.2112252641,1.2194426829,low,low,low,9.4,0,NEG,1.0,0.0,-1.0,,,0,GTR
13
+ 11,1,TCGA-06-0166,79.76086734,74.46603485,< 5%,Mixed T1<FLAIR Ratio,Crosses Midline No,3.038,4.47,0.763,1.5T,PN,Proneural,51,2001-00-00,-18902,178,161,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,66.0,1,66,178,3.1124329322,2.2654542586,1.5328414614,high,low,high,10.2,1,NEG,1.0,2.0,-1.0,,"T167S,T167S",1,STR
14
+ 12,1,TCGA-06-0168,44.13221598,24.29751635,6-33%,Infiltrative T1<<FLAIR Ratio,Crosses Midline No,1.37,3.303,0.62,1.5T,Mes,Neural,59,2002-00-00,-21776,598,579,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,100.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,461.0,1,461,598,1.4035658713,1.6740034488,1.2455592478,low,low,low,11.8,2,NEG,1.0,0.0,-1.0,,A328fs,1,STR
15
+ 13,1,TCGA-06-0173,107.1367651,52.88218154,6-33%,Infiltrative T1<<FLAIR Ratio,Crosses Midline Yes,2.027,3.73,0.96,3T,Prolif,Neural,72,2003-00-00,-26548,171,7,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,171,171,2.0766627892,1.890412614,1.9286078676,low,low,high,14.4,3,NEG,2.0,2.0,-1.0,,,1,STR
16
+ 14,1,TCGA-06-0175,58.76606943,37.35171235,None (0%),Infiltrative T1<<FLAIR Ratio,Crosses Midline No,2.12,3.63,0.577,1.5T,Mes,Mesenchymal,69,2003-00-00,-25558,123,83,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,100.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,39.0,1,39,123,2.1719413484,1.8397313107,1.1591736871,low,low,low,13.8,3,NEG,1.0,0.0,-1.0,,,0,GTR
17
+ 15,1,TCGA-06-0177,70.2938662,47.73769856,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,1.368,3.067,0.253,1.5T,Prolif,Proneural,64,2004-00-00,-23498,127,60,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,60.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,127,127,1.4015168701,1.554395573,0.5082685318,low,low,low,12.8,4,NEG,2.0,2.0,-1.0,,,0,STR
18
+ 16,1,TCGA-06-0179,64.06461812,44.22389414,< 5%,Mixed T1<FLAIR Ratio,Crosses Midline No,2.51,5.493,0.623,3T,PN,Neural,64,2004-00-00,-23449,616,578,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,60.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,250.0,1,250,616,2.5714965964,2.7839239916,1.2515861474,high,high,low,12.8,4,NEG,2.0,0.0,-1.0,,,0,GTR
19
+ 17,1,TCGA-06-0184,82.84546219,64.92510487,6-33%,Expansive T1~FLAIR Ratio,Crosses Midline No,1.672,3.067,0.71,1.5T,Mes,Mesenchymal,63,2005-00-00,-23317,2126,1228,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,1276.0,1,1276,2126,1.7129650634,1.554395573,1.4263662354,low,low,low,12.6,5,NEG,1.0,0.0,-1.0,,"G36E,G36E,G36E",1,GTR
20
+ 18,1,TCGA-06-0185,72.07177397,44.71617175,6-33%,Mixed T1<FLAIR Ratio,Crosses Midline No,1.99,4.267,0.413,1.5T,PN,Neural,54,2005-00-00,-19922,2366,2246,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,100.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,711.0,1,711,2366,2.0387562657,2.1625712129,0.8297031764,low,low,low,10.8,5,NEG,2.0,0.0,-2.0,"V651M,V651M,V651M",,1,GTR
21
+ 19,1,TCGA-06-0187,65.87112795,44.41677325,None (0%),Expansive T1~FLAIR Ratio,Crosses Midline No,1.835,3.233,0.517,1.5T,Mes,Classical,69,2006-00-00,-25317,828,801,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,60.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,531.0,531.0,1,531,828,1.8799586671,1.6385265365,1.0386356953,low,low,low,13.8,6,NEG,2.0,0.0,-1.0,,"G132D,G132D,G132D",1,STR
22
+ 20,2,TCGA-06-0189,85.25564146,56.4416166,< 5%,Mixed T1<FLAIR Ratio,Crosses Midline No,2.708,4.99,0.573,3T,Mes,Mesenchymal,55,2006-00-00,-20296,468,454,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,468,468,2.7743477223,2.5289970359,1.1511378209,high,high,low,11.0,6,NEG,2.0,0.0,-1.0,,,1,STR
23
+ 21,2,TCGA-06-0190,74.76880933,60.24171094,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,4.376,6.68,1.333,1.5T,Mes,Mesenchymal,62,2006-00-00,-22835,317,313,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,88.0,1,88,317,4.4832147832,3.3855110621,2.6779523828,high,high,high,12.4,6,NEG,1.0,1.0,-1.0,,"D92E,D92E,D92E",1,STR
24
+ 22,1,TCGA-06-0192,79.16025289,42.82469307,6-33%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.425,4.16,1.22,3T,Mes,Mesenchymal,58,2007-00-00,-21243,1185,556,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,100.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,648.0,1,648,1185,2.4844140423,2.1083422183,2.450939165,high,low,high,11.6,7,NEG,2.0,2.0,-1.0,,Y315*,1,GTR
25
+ 23,1,TCGA-06-0213,72.9466112,43.71555926,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.167,3.763,0.767,1.5T,Mes,Mesenchymal,55,1998-00-00,-20134,16,6,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,16,16,2.2200928782,1.9071374441,1.5408773275,low,low,high,11.0,-2,NEG,1.0,0.0,-2.0,,"R335*,R335*",1,STR
26
+ 24,1,TCGA-06-0237,70.35187771,28.66265438,68-95%,Infiltrative T1<<FLAIR Ratio,Crosses Midline No,3.365,5.537,0.613,1.5T,PN,Neural,75,2006-00-00,-27735,415,314,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,415,415,3.4474446402,2.8062237651,1.2314964821,high,high,low,15.0,6,NEG,2.0,1.0,-1.0,"L62R,L62R",,1,STR
27
+ 25,2,TCGA-06-0238,76.80296366,48.00510283,6-33%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.346,4.023,0.62,3T,PN,Proneural,46,2007-00-00,-17037,405,359,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,311.0,1,311,405,2.4034784921,2.0389088328,1.2455592478,low,low,low,9.2,7,NEG,1.0,0.0,-1.0,,,1,STR
28
+ 26,1,TCGA-06-0241,53.7870232,33.79282207,None (0%),Mixed T1<FLAIR Ratio,Crosses Midline No,2.912,4.43,0.66,3T,Prolif,Proneural,65,2007-00-00,-24101,1481,455,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,100.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,196.0,196.0,1,196,1481,2.9833458521,2.2451817373,1.3259179089,high,low,low,13.0,7,NEG,1.0,2.0,-1.0,,,1,STR
29
+ 27,1,TCGA-06-0644,51.55889274,40.19048554,None (0%),Expansive T1~FLAIR Ratio,Crosses Midline No,2.555,4.19,0.617,3T,Mes,Mesenchymal,71,2007-00-00,-26246,384,375,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Pre-Operative,WITH TUMOR,NO,BLACK OR AFRICAN AMERICAN,Glioblastoma multiforme,BRAIN,DECEASED,85.0,85.0,1,85,384,2.617599125,2.1235466093,1.2395323482,high,low,low,14.2,7,NEG,1.0,0.0,-1.0,,"T202fs,I203fs",1,STR
30
+ 28,1,TCGA-06-0646,47.0471611,36.60848593,34-67%,Expansive T1~FLAIR Ratio,Crosses Midline No,3.393,4.86,1.033,3T,Mes,Proneural,60,2007-00-00,-22272,175,136,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,90.0,1,90,175,3.476130658,2.4631113416,2.0752624241,high,high,high,12.0,7,NEG,2.0,1.0,-2.0,,,1,STR
31
+ 29,1,TCGA-06-0648,89.38226471,60.540485,6-33%,Expansive T1~FLAIR Ratio,Crosses Midline No,1.925,4.0,0.76,3T,PN,Proneural,77,2007-00-00,-28477,298,293,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Pre-Operative,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,201.0,1,201,298,1.9721637243,2.027252133,1.5268145618,low,low,high,15.4,7,NEG,1.0,0.0,-1.0,,,1,STR
32
+ 30,1,TCGA-08-0352,115.0557228,66.04929696,< 5%,Mixed T1<FLAIR Ratio,Crosses Midline No,2.752,5.727,0.56,1.5T,Mes,Mesenchymal,79,2003-00-00,-29106,39,39,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,39,39,2.8194257503,2.9025182414,1.1250212561,high,high,low,15.8,3,NEG,2.0,0.0,-1.0,,K263*,1,
33
+ 31,1,TCGA-08-0353,79.35522434,44.92923295,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.748,4.98,0.94,1.5T,Prolif,Proneural,58,2003-00-00,-21332,256,195,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Post-Adjuvant Therapy,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,164.0,1,164,256,2.8153277478,2.5239289056,1.888428537,high,high,high,11.6,3,NEG,,,,,,1,
34
+ 32,1,TCGA-08-0354,53.97760228,38.37069558,34-67%,Expansive T1~FLAIR Ratio,Crosses Midline Yes,3.038,5.393,0.827,1.5T,Mes,Classical,52,2003-00-00,-19198,546,458,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,60.0,Post-Adjuvant Therapy,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,253.0,1,253,546,3.1124329322,2.7332426883,1.6614153192,high,high,high,10.4,3,NEG,2.0,0.0,-1.0,,,1,
35
+ 33,1,TCGA-08-0355,77.67814706,56.93133533,6-33%,Expansive T1~FLAIR Ratio,Crosses Midline No,3.073,5.27,0.463,1.5T,Mes,Classical,30,2003-00-00,-11098,747,685,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,100.0,Other,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,519.0,519.0,1,519,747,3.1482904545,2.6709046852,0.9301515028,high,high,low,6.0,3,,2.0,0.0,-1.0,A289D,,1,
36
+ 34,1,TCGA-08-0356,67.52694124,38.67870135,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.058,4.89,0.273,1.5T,Mes,Classical,59,2004-00-00,-21900,946,447,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,80.0,Pre-Adjuvant Therapy,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,447.0,447.0,1,447,946,2.1084223089,2.4783157326,0.5484478623,low,high,low,11.8,4,NEG,2.0,0.0,-1.0,,,1,
37
+ 35,1,TCGA-08-0357,65.42796422,41.479033,6-33%,Mixed T1<FLAIR Ratio,Crosses Midline No,2.972,4.567,1.01,1.5T,PN,Classical,49,2004-00-00,-18143,1143,1084,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Other,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,155.0,155.0,1,155,1143,3.0448158902,2.3146151229,2.029056194,high,high,high,9.8,4,NEG,2.0,0.0,-1.0,,,1,
38
+ 36,1,TCGA-08-0358,62.85813516,42.12099332,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.968,4.8,0.473,1.5T,PN,Classical,50,2005-00-00,-18383,678,593,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Pre-Adjuvant Therapy,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,263.0,263.0,1,263,678,3.0407178877,2.4327025596,0.9502411681,high,high,low,10.0,5,NEG,2.0,0.0,-1.0,,,1,
39
+ 37,1,TCGA-08-0359,57.70535869,53.43134242,6-33%,Mixed T1<FLAIR Ratio,Crosses Midline Yes,1.518,3.413,0.933,1.5T,PN,Proneural,59,2005-00-00,-21748,102,29,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,102,102,1.5551919655,1.7297528825,1.8743657713,low,low,high,11.8,5,NEG,,,,,"R47G,R47G",1,
40
+ 38,1,TCGA-08-0360,33.67883942,25.59985695,6-33%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.252,3.993,0.587,1.5T,Mes,Mesenchymal,76,2005-00-00,-27894,468,352,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Pre-Adjuvant Therapy,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,112.0,112.0,1,112,468,2.3071754323,2.0237044418,1.1792633523,low,low,low,15.2,5,NEG,,,,,,1,
41
+ 39,1,TCGA-08-0385,77.56004839,55.03851551,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,2.546,16.267,1.125,1.5T,PN,Proneural,71,2003-00-00,-26233,82,31,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,60.0,Pre-Adjuvant Therapy,WITH TUMOR,NO,,Glioblastoma multiforme,BRAIN,DECEASED,,,1,82,82,2.6083786193,8.2443276119,2.2600873448,high,high,high,14.2,3,,,,,,,1,
42
+ 40,1,TCGA-08-0389,62.87975086,36.42895664,68-95%,Mixed T1<FLAIR Ratio,Crosses Midline No,1.862,3.317,1.123,1.5T,PN,Proneural,59,2005-00-00,-21638,467,97,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,80.0,Other,WITH TUMOR,NO,,Glioblastoma multiforme,BRAIN,DECEASED,,,1,467,467,1.9076201843,1.6810988313,2.2560694117,low,low,high,11.8,5,NEG,1.0,0.0,-1.0,,F271S,1,
43
+ 41,1,TCGA-08-0392,96.34986554,56.55214654,< 5%,Expansive T1~FLAIR Ratio,Crosses Midline No,1.728,3.923,0.457,1.5T,Prolif,Mesenchymal,60,2005-00-00,-22056,22,9,NOT HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,,,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,,1,22,22,1.770337099,1.9882275294,0.9180977036,low,low,low,12.0,5,NEG,1.0,0.0,-1.0,,,0,
44
+ 42,1,TCGA-08-0521,71.92146975,45.32140912,None (0%),Mixed T1<FLAIR Ratio,Crosses Midline No,1.74,4.72,1.017,1.5T,Mes,Mesenchymal,17,2002-00-00,-6376,146,146,HISPANIC OR LATINO,MALE,Untreated primary (de novo) GBM,60.0,Post-Adjuvant Therapy,WITH TUMOR,NO,BLACK OR AFRICAN AMERICAN,Glioblastoma multiforme,BRAIN,DECEASED,,125.0,1,125,146,1.7826311067,2.3921575169,2.0431189597,low,high,high,3.4,2,NEG,2.0,0.0,-1.0,,,0,
45
+ 43,1,TCGA-08-0524,114.4584859,58.49940496,6-33%,Mixed T1<FLAIR Ratio,Crosses Midline Yes,2.216,5.003,1.103,1.5T,PN,Proneural,17,2002-00-00,-6464,220,198,NOT HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,80.0,Other,WITH TUMOR,NO,WHITE,Glioblastoma multiforme,BRAIN,DECEASED,,61.0,1,61,220,2.2702934094,2.5355856054,2.2158900812,low,high,high,3.4,2,NEG,0.0,2.0,0.0,,,0,
46
+ 44,1,TCGA-08-0529,70.00860426,43.9033772,None (0%),Expansive T1~FLAIR Ratio,Crosses Midline No,4.313,5.373,0.8,1.5T,Mes,Classical,56,2004-00-00,-20556,559,526,HISPANIC OR LATINO,FEMALE,Untreated primary (de novo) GBM,80.0,Pre-Adjuvant Therapy,WITH TUMOR,NO,,Glioblastoma multiforme,BRAIN,DECEASED,328.0,328.0,1,328,559,4.4186712431,2.7231064277,1.607173223,high,high,high,11.2,4,NEG,2.0,0.0,-1.0,,,0,
samples/Outcome-Predictors-leaderboard.tsv ADDED
@@ -0,0 +1,46 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ Round TCGAID MajorAxisLength MinorAxisLength PropnCET T1FLAIR nCETCrossMidline rCBVmean rCBVmax rCBVfl Machine Subtype_by_phils Subtype_by_Verhaak age_at_initial_pathologic_diagno date_of_initial_pathologic_diagn days_to_birth days_to_death days_to_last_followup ethnicity gender histological_type karnofsky_performance_score performance_status_scale_timing person_neoplasm_cancer_status prior_glioma race tumor_histologic_subtype tumor_tissue_site vital_status days_to_tumor_recurrence time_to_progression progEvent progSurvTime OStime rCBVmeanSD rCBVmaxSD rCBVflSD rCBVmean2 rCBVmax2 rCBVfl2 age5 cohort gcimp egfrAmp pdgfraAmp ptenAmp egfrMut ptenMut sequenced Extent_of_Resection
2
+ 1 TCGA-06-0122 71.47023114 40.43448767 None (0%) Mixed T1<FLAIR Ratio Crosses Midline No 6.86 9.483 1.467 3T Mes Mesenchymal 84 2006-00-00 -30967 181 8 NOT HISPANIC OR LATINO FEMALE null NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 181 181 7.0280743631 4.8061079943 2.9471538976 high high high 16.8 6 NEG 2 0 -1 NaN NaN 1 STR
3
+ 1 TCGA-06-0127 86.97233129 55.44624428 6-33% Mixed T1<FLAIR Ratio Crosses Midline Yes 4.243 5.973 1.177 1.5T PN Neural 67 2002-00-00 -24502 121 108 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 60 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 90 1 90 121 4.3469561986 3.0271942476 2.3645536043 high high high 13.4 2 NEG 2 0 -1 V292L,V292L,V292L NaN 1 STR
4
+ 2 TCGA-06-0128 68.54275415 55.25454465 34-67% Expansive T1~FLAIR Ratio Crosses Midline No 1.388 2.83 0.713 1.5T PN Proneural 66 2000-00-00 -24217 691 691 NOT HISPANIC OR LATINO MALE null 80 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 189 1 189 691 1.4220068828 1.4342808841 1.432393135 low low low 13.2 0 POS 0 0 -1 NaN NaN 1 GTR
5
+ 1 TCGA-06-0132 90.38251662 62.26697312 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 2.173 6.127 0.837 1.5T PN Neural 49 2007-00-00 -18125 761 570 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 482 1 482 761 2.2262398821 3.1052434547 1.6815049845 low high high 9.8 7 NEG 2 0 -1 NaN NaN 1 STR
6
+ 1 TCGA-06-0133 102.8050346 62.89969015 34-67% Mixed T1<FLAIR Ratio Crosses Midline Yes 2.435 4.663 0.553 3T PN Neural 64 2007-00-00 -23402 435 428 NOT HISPANIC OR LATINO MALE null WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 78 1 78 435 2.4946590487 2.363269174 1.1109584904 high high low 12.8 7 NEG 2 0 -1 NaN NaN 1 STR
7
+ 1 TCGA-06-0137 101.8218393 56.31768036 None (0%) Mixed T1<FLAIR Ratio Crosses Midline No 2.53 4.317 0.717 1.5T Mes Classical 63 2003-00-00 -23273 812 701 NOT HISPANIC OR LATINO FEMALE null WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 487 1 487 812 2.5919866091 2.1879118645 1.4404290011 high low low 12.6 3 NEG 2 0 -1 P596L NaN 1 STR
8
+ 1 TCGA-06-0139 80.6709788 39.04821797 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 2.112 4.497 1.24 3T Mes Mesenchymal 40 2006-00-00 -14728 383 327 NOT HISPANIC OR LATINO MALE null 60 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 152 1 152 383 2.1637453433 2.2791382105 2.4911184956 low high high 8 6 NEG 1 0 -1 NaN NaN 1 STR
9
+ 1 TCGA-06-0143 114.8301745 65.62670418 < 5% Expansive T1~FLAIR Ratio Crosses Midline Yes 2.393 5.657 0.353 3T Mes Mesenchymal 58 2006-00-00 -21386 357 357 NOT HISPANIC OR LATINO MALE null 60 WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 264 264 1 264 357 2.451630022 2.8670413291 0.7091651846 low high low 11.6 6 NEG 2 2 -1 NaN NaN 1 STR
10
+ 1 TCGA-06-0147 74.49334479 51.14346067 None (0%) Expansive T1~FLAIR Ratio Crosses Midline No 2.24 4.76 0.865 1.5T Mes Mesenchymal 51 1999-00-00 -18742 541 508 NOT HISPANIC OR LATINO FEMALE null WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 92 1 92 541 2.2948814247 2.4124300383 1.7377560473 low high high 10.2 -1 NEG 1 0 -1 NaN NaN 1 STR
11
+ 1 TCGA-06-0149 90.87360905 48.56049078 68-95% Infiltrative T1<<FLAIR Ratio Crosses Midline Yes 2.51 3.99 2.057 3T Mes Mesenchymal 74 2005-00-00 -27315 262 238 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 80 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 203 1 203 262 2.5714965964 2.0221840027 4.1324441495 high low high 14.8 5 NEG 1 0 -1 NaN NaN 0 STR
12
+ 1 TCGA-06-0164 66.76339735 46.13628674 None (0%) Expansive T1~FLAIR Ratio Crosses Midline No 1.768 4.363 0.607 1.5T Mes Mesenchymal 47 2000-00-00 -17510 1730 1729 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 100 Pre-Operative WITH TUMOR NO ASIAN Glioblastoma multiforme BRAIN DECEASED 1428 1 1428 1730 1.8113171245 2.2112252641 1.2194426829 low low low 9.4 0 NEG 1 0 -1 NaN NaN 0 GTR
13
+ 1 TCGA-06-0166 79.76086734 74.46603485 < 5% Mixed T1<FLAIR Ratio Crosses Midline No 3.038 4.47 0.763 1.5T PN Proneural 51 2001-00-00 -18902 178 161 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 66 1 66 178 3.1124329322 2.2654542586 1.5328414614 high low high 10.2 1 NEG 1 2 -1 NaN T167S,T167S 1 STR
14
+ 1 TCGA-06-0168 44.13221598 24.29751635 6-33% Infiltrative T1<<FLAIR Ratio Crosses Midline No 1.37 3.303 0.62 1.5T Mes Neural 59 2002-00-00 -21776 598 579 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 100 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 461 1 461 598 1.4035658713 1.6740034488 1.2455592478 low low low 11.8 2 NEG 1 0 -1 NaN A328fs 1 STR
15
+ 1 TCGA-06-0173 107.1367651 52.88218154 6-33% Infiltrative T1<<FLAIR Ratio Crosses Midline Yes 2.027 3.73 0.96 3T Prolif Neural 72 2003-00-00 -26548 171 7 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 171 171 2.0766627892 1.890412614 1.9286078676 low low high 14.4 3 NEG 2 2 -1 NaN NaN 1 STR
16
+ 1 TCGA-06-0175 58.76606943 37.35171235 None (0%) Infiltrative T1<<FLAIR Ratio Crosses Midline No 2.12 3.63 0.577 1.5T Mes Mesenchymal 69 2003-00-00 -25558 123 83 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 100 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 39 1 39 123 2.1719413484 1.8397313107 1.1591736871 low low low 13.8 3 NEG 1 0 -1 NaN NaN 0 GTR
17
+ 1 TCGA-06-0177 70.2938662 47.73769856 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 1.368 3.067 0.253 1.5T Prolif Proneural 64 2004-00-00 -23498 127 60 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 60 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 127 127 1.4015168701 1.554395573 0.5082685318 low low low 12.8 4 NEG 2 2 -1 NaN NaN 0 STR
18
+ 1 TCGA-06-0179 64.06461812 44.22389414 < 5% Mixed T1<FLAIR Ratio Crosses Midline No 2.51 5.493 0.623 3T PN Neural 64 2004-00-00 -23449 616 578 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 60 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 250 1 250 616 2.5714965964 2.7839239916 1.2515861474 high high low 12.8 4 NEG 2 0 -1 NaN NaN 0 GTR
19
+ 1 TCGA-06-0184 82.84546219 64.92510487 6-33% Expansive T1~FLAIR Ratio Crosses Midline No 1.672 3.067 0.71 1.5T Mes Mesenchymal 63 2005-00-00 -23317 2126 1228 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1276 1 1276 2126 1.7129650634 1.554395573 1.4263662354 low low low 12.6 5 NEG 1 0 -1 NaN G36E,G36E,G36E 1 GTR
20
+ 1 TCGA-06-0185 72.07177397 44.71617175 6-33% Mixed T1<FLAIR Ratio Crosses Midline No 1.99 4.267 0.413 1.5T PN Neural 54 2005-00-00 -19922 2366 2246 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 100 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 711 1 711 2366 2.0387562657 2.1625712129 0.8297031764 low low low 10.8 5 NEG 2 0 -2 V651M,V651M,V651M NaN 1 GTR
21
+ 1 TCGA-06-0187 65.87112795 44.41677325 None (0%) Expansive T1~FLAIR Ratio Crosses Midline No 1.835 3.233 0.517 1.5T Mes Classical 69 2006-00-00 -25317 828 801 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 60 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 531 531 1 531 828 1.8799586671 1.6385265365 1.0386356953 low low low 13.8 6 NEG 2 0 -1 NaN G132D,G132D,G132D 1 STR
22
+ 2 TCGA-06-0189 85.25564146 56.4416166 < 5% Mixed T1<FLAIR Ratio Crosses Midline No 2.708 4.99 0.573 3T Mes Mesenchymal 55 2006-00-00 -20296 468 454 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 468 468 2.7743477223 2.5289970359 1.1511378209 high high low 11 6 NEG 2 0 -1 NaN NaN 1 STR
23
+ 2 TCGA-06-0190 74.76880933 60.24171094 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 4.376 6.68 1.333 1.5T Mes Mesenchymal 62 2006-00-00 -22835 317 313 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 88 1 88 317 4.4832147832 3.3855110621 2.6779523828 high high high 12.4 6 NEG 1 1 -1 NaN D92E,D92E,D92E 1 STR
24
+ 1 TCGA-06-0192 79.16025289 42.82469307 6-33% Expansive T1~FLAIR Ratio Crosses Midline No 2.425 4.16 1.22 3T Mes Mesenchymal 58 2007-00-00 -21243 1185 556 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 100 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 648 1 648 1185 2.4844140423 2.1083422183 2.450939165 high low high 11.6 7 NEG 2 2 -1 NaN Y315* 1 GTR
25
+ 1 TCGA-06-0213 72.9466112 43.71555926 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 2.167 3.763 0.767 1.5T Mes Mesenchymal 55 1998-00-00 -20134 16 6 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 16 16 2.2200928782 1.9071374441 1.5408773275 low low high 11 -2 NEG 1 0 -2 NaN R335*,R335* 1 STR
26
+ 1 TCGA-06-0237 70.35187771 28.66265438 68-95% Infiltrative T1<<FLAIR Ratio Crosses Midline No 3.365 5.537 0.613 1.5T PN Neural 75 2006-00-00 -27735 415 314 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 415 415 3.4474446402 2.8062237651 1.2314964821 high high low 15 6 NEG 2 1 -1 L62R,L62R NaN 1 STR
27
+ 2 TCGA-06-0238 76.80296366 48.00510283 6-33% Expansive T1~FLAIR Ratio Crosses Midline No 2.346 4.023 0.62 3T PN Proneural 46 2007-00-00 -17037 405 359 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 311 1 311 405 2.4034784921 2.0389088328 1.2455592478 low low low 9.2 7 NEG 1 0 -1 NaN NaN 1 STR
28
+ 1 TCGA-06-0241 53.7870232 33.79282207 None (0%) Mixed T1<FLAIR Ratio Crosses Midline No 2.912 4.43 0.66 3T Prolif Proneural 65 2007-00-00 -24101 1481 455 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 100 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 196 196 1 196 1481 2.9833458521 2.2451817373 1.3259179089 high low low 13 7 NEG 1 2 -1 NaN NaN 1 STR
29
+ 1 TCGA-06-0644 51.55889274 40.19048554 None (0%) Expansive T1~FLAIR Ratio Crosses Midline No 2.555 4.19 0.617 3T Mes Mesenchymal 71 2007-00-00 -26246 384 375 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Pre-Operative WITH TUMOR NO BLACK OR AFRICAN AMERICAN Glioblastoma multiforme BRAIN DECEASED 85 85 1 85 384 2.617599125 2.1235466093 1.2395323482 high low low 14.2 7 NEG 1 0 -1 NaN T202fs,I203fs 1 STR
30
+ 1 TCGA-06-0646 47.0471611 36.60848593 34-67% Expansive T1~FLAIR Ratio Crosses Midline No 3.393 4.86 1.033 3T Mes Proneural 60 2007-00-00 -22272 175 136 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 90 1 90 175 3.476130658 2.4631113416 2.0752624241 high high high 12 7 NEG 2 1 -2 NaN NaN 1 STR
31
+ 1 TCGA-06-0648 89.38226471 60.540485 6-33% Expansive T1~FLAIR Ratio Crosses Midline No 1.925 4 0.76 3T PN Proneural 77 2007-00-00 -28477 298 293 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Pre-Operative WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 201 1 201 298 1.9721637243 2.027252133 1.5268145618 low low high 15.4 7 NEG 1 0 -1 NaN NaN 1 STR
32
+ 1 TCGA-08-0352 115.0557228 66.04929696 < 5% Mixed T1<FLAIR Ratio Crosses Midline No 2.752 5.727 0.56 1.5T Mes Mesenchymal 79 2003-00-00 -29106 39 39 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 39 39 2.8194257503 2.9025182414 1.1250212561 high high low 15.8 3 NEG 2 0 -1 NaN K263* 1
33
+ 1 TCGA-08-0353 79.35522434 44.92923295 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 2.748 4.98 0.94 1.5T Prolif Proneural 58 2003-00-00 -21332 256 195 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Post-Adjuvant Therapy WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 164 1 164 256 2.8153277478 2.5239289056 1.888428537 high high high 11.6 3 NEG NaN NaN NaN NaN NaN 1
34
+ 1 TCGA-08-0354 53.97760228 38.37069558 34-67% Expansive T1~FLAIR Ratio Crosses Midline Yes 3.038 5.393 0.827 1.5T Mes Classical 52 2003-00-00 -19198 546 458 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 60 Post-Adjuvant Therapy WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 253 1 253 546 3.1124329322 2.7332426883 1.6614153192 high high high 10.4 3 NEG 2 0 -1 NaN NaN 1
35
+ 1 TCGA-08-0355 77.67814706 56.93133533 6-33% Expansive T1~FLAIR Ratio Crosses Midline No 3.073 5.27 0.463 1.5T Mes Classical 30 2003-00-00 -11098 747 685 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 100 Other WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 519 519 1 519 747 3.1482904545 2.6709046852 0.9301515028 high high low 6 3 2 0 -1 A289D NaN 1
36
+ 1 TCGA-08-0356 67.52694124 38.67870135 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 2.058 4.89 0.273 1.5T Mes Classical 59 2004-00-00 -21900 946 447 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 80 Pre-Adjuvant Therapy WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 447 447 1 447 946 2.1084223089 2.4783157326 0.5484478623 low high low 11.8 4 NEG 2 0 -1 NaN NaN 1
37
+ 1 TCGA-08-0357 65.42796422 41.479033 6-33% Mixed T1<FLAIR Ratio Crosses Midline No 2.972 4.567 1.01 1.5T PN Classical 49 2004-00-00 -18143 1143 1084 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Other WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 155 155 1 155 1143 3.0448158902 2.3146151229 2.029056194 high high high 9.8 4 NEG 2 0 -1 NaN NaN 1
38
+ 1 TCGA-08-0358 62.85813516 42.12099332 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 2.968 4.8 0.473 1.5T PN Classical 50 2005-00-00 -18383 678 593 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Pre-Adjuvant Therapy WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 263 263 1 263 678 3.0407178877 2.4327025596 0.9502411681 high high low 10 5 NEG 2 0 -1 NaN NaN 1
39
+ 1 TCGA-08-0359 57.70535869 53.43134242 6-33% Mixed T1<FLAIR Ratio Crosses Midline Yes 1.518 3.413 0.933 1.5T PN Proneural 59 2005-00-00 -21748 102 29 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 102 102 1.5551919655 1.7297528825 1.8743657713 low low high 11.8 5 NEG NaN NaN NaN NaN R47G,R47G 1
40
+ 1 TCGA-08-0360 33.67883942 25.59985695 6-33% Expansive T1~FLAIR Ratio Crosses Midline No 2.252 3.993 0.587 1.5T Mes Mesenchymal 76 2005-00-00 -27894 468 352 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Pre-Adjuvant Therapy WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 112 112 1 112 468 2.3071754323 2.0237044418 1.1792633523 low low low 15.2 5 NEG NaN NaN NaN NaN NaN 1
41
+ 1 TCGA-08-0385 77.56004839 55.03851551 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 2.546 16.267 1.125 1.5T PN Proneural 71 2003-00-00 -26233 82 31 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 60 Pre-Adjuvant Therapy WITH TUMOR NO Glioblastoma multiforme BRAIN DECEASED 1 82 82 2.6083786193 8.2443276119 2.2600873448 high high high 14.2 3 NaN NaN NaN NaN NaN 1
42
+ 1 TCGA-08-0389 62.87975086 36.42895664 68-95% Mixed T1<FLAIR Ratio Crosses Midline No 1.862 3.317 1.123 1.5T PN Proneural 59 2005-00-00 -21638 467 97 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 80 Other WITH TUMOR NO Glioblastoma multiforme BRAIN DECEASED 1 467 467 1.9076201843 1.6810988313 2.2560694117 low low high 11.8 5 NEG 1 0 -1 NaN F271S 1
43
+ 1 TCGA-08-0392 96.34986554 56.55214654 < 5% Expansive T1~FLAIR Ratio Crosses Midline No 1.728 3.923 0.457 1.5T Prolif Mesenchymal 60 2005-00-00 -22056 22 9 NOT HISPANIC OR LATINO MALE Untreated primary (de novo) GBM WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 1 22 22 1.770337099 1.9882275294 0.9180977036 low low low 12 5 NEG 1 0 -1 NaN NaN 0
44
+ 1 TCGA-08-0521 71.92146975 45.32140912 None (0%) Mixed T1<FLAIR Ratio Crosses Midline No 1.74 4.72 1.017 1.5T Mes Mesenchymal 17 2002-00-00 -6376 146 146 HISPANIC OR LATINO MALE Untreated primary (de novo) GBM 60 Post-Adjuvant Therapy WITH TUMOR NO BLACK OR AFRICAN AMERICAN Glioblastoma multiforme BRAIN DECEASED 125 1 125 146 1.7826311067 2.3921575169 2.0431189597 low high high 3.4 2 NEG 2 0 -1 NaN NaN 0
45
+ 1 TCGA-08-0524 114.4584859 58.49940496 6-33% Mixed T1<FLAIR Ratio Crosses Midline Yes 2.216 5.003 1.103 1.5T PN Proneural 17 2002-00-00 -6464 220 198 NOT HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 80 Other WITH TUMOR NO WHITE Glioblastoma multiforme BRAIN DECEASED 61 1 61 220 2.2702934094 2.5355856054 2.2158900812 low high high 3.4 2 NEG 0 2 0 NaN NaN 0
46
+ 1 TCGA-08-0529 70.00860426 43.9033772 None (0%) Expansive T1~FLAIR Ratio Crosses Midline No 4.313 5.373 0.8 1.5T Mes Classical 56 2004-00-00 -20556 559 526 HISPANIC OR LATINO FEMALE Untreated primary (de novo) GBM 80 Pre-Adjuvant Therapy WITH TUMOR NO Glioblastoma multiforme BRAIN DECEASED 328 328 1 328 559 4.4186712431 2.7231064277 1.607173223 high high high 11.2 4 NEG 2 0 -1 NaN NaN 0
samples/Outcome-Predictors-leaderboard_meta.csv ADDED
@@ -0,0 +1,51 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ dtype,count,unique,null,freq,min,max,PV_match,Enumerated,NonEnumerated
2
+ Round,int64,45,2,0,,1.0,2.0,1.0,True,True
3
+ TCGAID,object,45,45,0,1,,,,False,True
4
+ MajorAxisLength,float64,45,45,0,,33.67883942,115.0557228,,False,True
5
+ MinorAxisLength,float64,45,45,0,,24.29751635,74.46603485,,False,True
6
+ PropnCET,object,45,5,0,15,,,0.0,False,True
7
+ T1FLAIR,object,45,3,0,25,,,0.0,False,True
8
+ nCETCrossMidline,object,45,2,0,37,,,0.0,False,True
9
+ rCBVmean,float64,45,43,0,,1.368,6.86,,False,True
10
+ rCBVmax,float64,45,44,0,,2.83,16.267,,False,True
11
+ rCBVfl,float64,45,44,0,,0.253,2.057,,False,True
12
+ Machine,object,45,2,0,31,,,0.5,False,True
13
+ Subtype_by_phils,object,45,3,0,24,,,0.3333333333333333,False,True
14
+ Subtype_by_Verhaak,object,45,4,0,17,,,0.5,False,True
15
+ age_at_initial_pathologic_diagno,int64,45,30,0,,17.0,84.0,,False,True
16
+ date_of_initial_pathologic_diagn,object,45,10,0,8,,,0.0,False,True
17
+ days_to_birth,int64,45,45,0,,-30967.0,-6376.0,,False,True
18
+ days_to_death,int64,45,44,0,,16.0,2366.0,,False,True
19
+ days_to_last_followup,int64,45,45,0,,6.0,2246.0,,False,True
20
+ ethnicity,object,45,2,0,43,,,1.0,True,False
21
+ gender,object,45,2,0,30,,,1.0,True,False
22
+ histological_type,object,38,2,7,38,,,0.0,False,True
23
+ karnofsky_performance_score,float64,32,4,13,,60.0,100.0,,False,True
24
+ performance_status_scale_timing,object,30,5,15,18,,,1.0,True,False
25
+ person_neoplasm_cancer_status,object,44,2,1,44,,,1.0,True,False
26
+ prior_glioma,object,45,1,0,45,,,1.0,True,False
27
+ race,object,42,4,3,39,,,1.0,True,False
28
+ tumor_histologic_subtype,object,45,1,0,45,,,1.0,True,False
29
+ tumor_tissue_site,object,45,1,0,45,,,1.0,True,False
30
+ vital_status,object,45,1,0,45,,,1.0,True,False
31
+ days_to_tumor_recurrence,float64,10,11,35,,85.0,531.0,,False,True
32
+ time_to_progression,float64,34,34,11,,39.0,1428.0,,False,True
33
+ progEvent,int64,45,1,0,,1.0,1.0,1.0,True,True
34
+ progSurvTime,int64,45,43,0,,16.0,1428.0,,False,True
35
+ OStime,int64,45,44,0,,16.0,2366.0,,False,True
36
+ rCBVmeanSD,float64,45,43,0,,1.4015168701,7.0280743631,,False,True
37
+ rCBVmaxSD,float64,45,44,0,,1.4342808841,8.2443276119,,False,True
38
+ rCBVflSD,float64,45,44,0,,0.5082685318,4.1324441495,,False,True
39
+ rCBVmean2,object,45,2,0,23,,,1.0,True,False
40
+ rCBVmax2,object,45,2,0,23,,,1.0,True,False
41
+ rCBVfl2,object,45,2,0,23,,,1.0,True,False
42
+ age5,float64,45,30,0,,3.4,16.8,,False,True
43
+ cohort,int64,45,10,0,,-2.0,7.0,1.0,True,True
44
+ gcimp,object,43,3,2,42,,,0.5,False,True
45
+ egfrAmp,float64,41,4,4,,0.0,2.0,,False,True
46
+ pdgfraAmp,float64,41,4,4,,0.0,2.0,,False,True
47
+ ptenAmp,float64,41,4,4,,-2.0,0.0,,False,True
48
+ egfrMut,object,5,6,40,1,,,,False,True
49
+ ptenMut,object,11,12,34,1,,,,False,True
50
+ sequenced,int64,45,2,0,,0.0,1.0,1.0,True,True
51
+ Extent_of_Resection,object,30,3,15,23,,,0.0,False,True
samples/Outcome-Predictors.txt ADDED
@@ -0,0 +1,30 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ From: https://pubs.rsna.org/doi/10.1148/radiol.14131691
2
+
3
+ Outcome Prediction in Patients with Glioblastoma by Using Imaging, Clinical, and Genomic Biomarkers: Focus on the Nonenhancing Component of the Tumor
4
+ Rajan Jain1 , Laila M. Poisson, David Gutman, Lisa Scarpace, Scott N. Hwang, Chad A. Holder, Max Wintermark, Arvind Rao, Rivka R. Colen2, Justin Kirby, John Freymann, C. Carl Jaffe, Tom Mikkelsen, Adam Flanders
5
+ Author Affiliations
6
+ Published Online:Mar 17 2014https://doi.org/10.1148/radiol.14131691
7
+
8
+ Abstract
9
+ In the current study, we focused on the role of the nonenhancing region (NER) of glioblastomas and showed that there are imaging phenotypic features related specifically to the NER—most notably the NER crossing the midline and relative cerebral blood volume of NER, which provide important prognostic information; these are complementary to clinical and genomic features and can improve models of patient prognosis.
10
+
11
+ Purpose
12
+ To correlate patient survival with morphologic imaging features and hemodynamic parameters obtained from the nonenhancing region (NER) of glioblastoma (GBM), along with clinical and genomic markers.
13
+
14
+ Materials and Methods
15
+ An institutional review board waiver was obtained for this HIPAA-compliant retrospective study. Forty-five patients with GBM underwent baseline imaging with contrast material–enhanced magnetic resonance (MR) imaging and dynamic susceptibility contrast-enhanced T2*-weighted perfusion MR imaging. Molecular and clinical predictors of survival were obtained. Single and multivariable models of overall survival (OS) and progression-free survival (PFS) were explored with Kaplan-Meier estimates, Cox regression, and random survival forests.
16
+
17
+ Results
18
+ Worsening OS (log-rank test, P = .0103) and PFS (log-rank test, P = .0223) were associated with increasing relative cerebral blood volume of NER (rCBVNER), which was higher with deep white matter involvement (t test, P = .0482) and poor NER margin definition (t test, P = .0147). NER crossing the midline was the only morphologic feature of NER associated with poor survival (log-rank test, P = .0125). Preoperative Karnofsky performance score (KPS) and resection extent (n = 30) were clinically significant OS predictors (log-rank test, P = .0176 and P = .0038, respectively). No genomic alterations were associated with survival, except patients with high rCBVNER and wild-type epidermal growth factor receptor (EGFR) mutation had significantly poor survival (log-rank test, P = .0306; area under the receiver operating characteristic curve = 0.62). Combining resection extent with rCBVNER marginally improved prognostic ability (permutation, P = .084). Random forest models of presurgical predictors indicated rCBVNER as the top predictor; also important were KPS, age at diagnosis, and NER crossing the midline. A multivariable model containing rCBVNER, age at diagnosis, and KPS can be used to group patients with more than 1 year of difference in observed median survival (0.49–1.79 years).
19
+
20
+ Conclusion
21
+ Patients with high rCBVNER and NER crossing the midline and those with high rCBVNER and wild-type EGFR mutation showed poor survival. In multivariable survival models, however, rCBVNER provided unique prognostic information that went above and beyond the assessment of all NER imaging features, as well as clinical and genomic features.
22
+
23
+ © RSNA, 2014
24
+
25
+ Online supplemental material is available for this article.
26
+
27
+ Article History
28
+ Received August 16, 2013; revision requested September 18; revision received December 20; accepted January 10, 2014; final version accepted January 20.
29
+ Published online: Mar 17 2014
30
+ Published in print: Aug 2014
samples/Outcome-Predictors_description.txt ADDED
@@ -0,0 +1,16 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ The manuscript in which this dataset is used correlates patient
2
+ survival with morphologic imaging features and hemodynamic parameters
3
+ obtained from the non-enhancing region (NER) of glioblastoma (GBM),
4
+ along with clinical and genomic markers. Forty-five patients with GBM
5
+ underwent baseline imaging with contrast material-enhanced magnetic
6
+ resonance (MR) imaging and dynamic susceptibility contrast-enhanced
7
+ T2*-weighted perfusion MR imaging. Molecular and clinical predictors
8
+ of survival were obtained. Single and multivariable models of overall
9
+ survival (OS) and progression-free survival (PFS) were explored with
10
+ Kaplan-Meier estimates, Cox regression, and random survival
11
+ forests. This dataset correlates with images on the TCIA web portal
12
+ and includes supplemental data including clinical, genomic, and
13
+ radiologist derived information from
14
+ JainPoisson_2014_Radiology_Dataset.xlsx. The xlsx file contains 2
15
+ sheets: the dataset, and a second worksheet of descriptions of the
16
+ variables.
samples/README.md ADDED
@@ -0,0 +1,27 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ # Metadata Challenge Input Files
2
+
3
+ [Metadata Automation DREAM Challenge Data Sources](https://www.synapse.org/#!Synapse:syn18065891/wiki/600450)
4
+
5
+ ## Tables and Files
6
+
7
+ There are 4 input tables:
8
+ - APOLLO-2
9
+ - REMBRANDT
10
+ - Outcome-Predictors
11
+ - ROI-Masks
12
+
13
+ APOLLO-5 was an attempt of getting more data to train on.
14
+
15
+ Each has:
16
+ - A -leaderboard.tsv spreadsheet of the RCT data
17
+ - a PDF paper of the results of the RCT trail
18
+ - a txt version of the PDF paper
19
+ - a _description.txt short paragraph description of the data provided at the competition
20
+
21
+ While ALL files for the train data were available at the start of the
22
+ competition, the compute environment for the test data did not provide
23
+ access to the txt and PDF files.
24
+
25
+ Still, it seems obvious that those files should be available at the
26
+ time of annotation, and we should find a way to use them.
27
+
samples/REMBRANDT-leaderboard.tsv ADDED
@@ -0,0 +1,128 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ Sample Age at Dx (years) Survival (months) Race Karnofsky MRI Desc Followup Month Steroid Dose Status Anti-Convulsant Status Prior Therapy Radiation Fraction Dose Prior Therapy Radiation Fraction Number Prior Therapy Radiation Type Prior Therapy Chemo Course Count Prior Therapy Surgery Procedure Title Prior Therapy Surgery Outcome OnStudy Therapy Radiation Site OnStudy Therapy Radiation Neurosis Status OnStudy Therapy Radiation Fraction Number OnStudy Therapy Radiation Type OnStudy Therapy Chemo Agent Name OnStudy Therapy Chemo Course Count OnStudy Therapy Surgery Procedure Title OnStudy Therapy Surgery Indication OnStudy Therapy Surgery Histo Diagnosis OnStudy Therapy Surgery Outcome
2
+ 900_00_1961 15
3
+ 900_00_5332 15
4
+ 900_00_5308 17
5
+ 900_00_5316 17
6
+ 900_00_5317 17
7
+ 900_00_5338 15
8
+ 900_00_5339 15
9
+ 900_00_5341 15
10
+ 900_00_5342 15
11
+ 900_00_5345 16
12
+ 900_00_5346 10
13
+ 900_00_5379 16
14
+ 900_00_5380 16
15
+ 900_00_5381 4
16
+ 900_00_5382 16
17
+ 900_00_5384 9
18
+ 900_00_5385 13
19
+ 900_00_5393 13
20
+ 900_00_5396 13
21
+ 900_00_5404 12
22
+ 900_00_5412 12
23
+ 900_00_5413 2
24
+ 900_00_5414 2
25
+ 900_00_5445 2
26
+ 900_00_5458 7
27
+ 900_00_5459 10
28
+ 900_00_5462 2
29
+ 900_00_5468 10
30
+ 900_00_5476 9
31
+ 900_00_5477 5
32
+ 900_00_5299 6
33
+ 900_00_5303 5
34
+ HF0608 50-54 10.6 WHITE rim enhancement and nodular foci NASS NASS NONE EBRT 9-AC(2) subtotal x2 recurrence GBM with necrosis subtotal
35
+ HF0652.4 50-54 19.6
36
+ HF0763
37
+ HF0828 35-39
38
+ HF0835 35-39 45.3 WHITE 90 non contrast enhancing NASS Yes NASS EBRT (3D-CRT) PCV+CCNU; EMD; Temodar; PCV+CCNU(2; 1; 2; 1) subtotal x 2 recurrence anaplastic oligodenroglioma; glioblastoma multiforme subtotal
39
+ HF0855 55-59 13.7 WHITE 90 ring enhancement; thick, irregular and nodular wall NASS NONE EBRT BCNU(4) subtotal recurrence radiation necrosis
40
+ HF0868
41
+ HF0883
42
+ HF0899 55-59 66.7 WHITE noncontrast enhancing NASS NASS EBRT NONE(NONE) subtotal recurrence malignant glioma subtotal
43
+ HF0920 85-89 1 WHITE 90 noncontrast enhancing; minimal mass effect 12 mg/day Dilantin NONE NONE NONE(NONE) NONE NONE NONE NONE
44
+ HF0931 35-39 119 WHITE 90 noncontrast enhancing 8 mg/day Dilantin EBRT PCV(4) subtotal recurrence foreign body granuloma subtotal
45
+ HF0953 35-39 44.6 WHITE 100 complex multicompartment tumor NONE NONE NONE EBRT BCNU (2) subtotal recurrence Gemistocytic Astrocytoma subtotal
46
+ HF0960 30-34 88.7 WHITE subtle contrast enhancement; cystic; midline shift Yes dose unknown Dilantin EBRT (3D-CRT) Temozolomide; Bay 43; PCV/CCNU(13 1 1) subtotal x 2 recurrence
47
+ HF0962 30-34 116.5 WHITE 100 subtle contrast enhancement NONE Dilantin NONE NONE Temozolomide(6) NONE NONE NONE NONE
48
+ HF0966 50-54 137.7 WHITE noncontrast enhancing NONE Dilantin EBRT PCV (6) NONE NONE NONE NONE
49
+ HF0986 35-39 62.4 WHITE 90 thick ring enhancing nodular mass NASS NASS EBRT PCV(4) subtotal recurrence mixed oligoastrocytoma subtotal
50
+ HF0990 40-44 88.3 WHITE mixed signal intesnsity lesion; bi-frontal; inhomogeneous areas of intense enhancement NASS NASS EBRT 5-FU/Carboplatin/Gamma IFN/Thalidomide/plasmapheresis(4) subtotal recurrence GBM subtotal
51
+ HF0996 50-54 120.5 WHITE 90 3cm x 2cm slightly heterogeneously enahncment NASS NASS EBRT BCNU(6) NONE NONE NONE NONE
52
+ HF1000 30-34 40.3 WHITE 90 minimal enhancement Yes dose unkown Yes type unknown EBRT SU-101(6) subtotal recurrence anaplastic astrocytoma subtotal
53
+ HF1032 40-44 28.1 WHITE 100 non enhancing; small amount of edema NASS Dilantin EBRT PCV(5) subtotal recurrence abcess/infection arteritis subtotal
54
+ HF1057 60-64 24.5 WHITE 90 irregularly enhancing mass; multiple; nodular enhancing NASS NASS EBRT CI-980(4) subtotal recurrence GBM subtotal
55
+ HF1058 50-54 18 WHITE large mass; significant vasogenic edema; mass effect NASS NASS EBRT BCNU(1) NONE NONE NONE NONE
56
+ HF1059 60-64 11.5 WHITE diffuse abnormality; mild compression of R lateral ventrile with mild midline shift 4mg t.i.d. NASS EBRT BCNU(2) NONE NONE NONE NONE
57
+ HF1071 55-59 8.8 WHITE shaggy, irregular peripheral enahcnement; central necrosis and surrounding edema NASS EBRT TMZ+Thalidomide(3) NONE NONE
58
+ HF1077 40-44 73.4 BLACK enhancing lesion; very little mass effect NASS NASS EBRT PCV(4) subtotal recurrence GBM subtotal
59
+ HF1078 45-49 22.8 WHITE 80 ring enhancing; multicystic; vasogenic edema; significant mass effect and midline shift NASS NASS EBRT penicillamine/low copper diet(9) subtotal recurrence malignant astrocytoma subtotal
60
+ HF1097 50-54 9 WHITE 90 enhancing NASS Dilantin EBRT TMZ(2) NONE NONE NONE NONE
61
+ HF1113
62
+ HF1122 35-39 7.3 WHITE 90 multiple enhancing lesions; in leptomeninges NONE Dilantin EBRT (L Frontal) + WBRT ARA-C(5) NONE NONE NONE NONE
63
+ HF1136 35-39 45.3 WHITE 100 contrast enhancing NONE Dilantin EBRT (3D-CRT) PCV + Temozolomide(2 PCV; 5 TMZ) subtotal recurrence anaplastic oligodenroglioma with necrosis subtotal
64
+ HF1137 70-74 18.3 WHITE 80 irregular enhancign mass with vasogenic edema and mass effect NASS NASS EBRT NONE(NONE) NONE NONE NONE NONE
65
+ HF1139 40-44 15.8 WHITE 80 large 6cm x 5cm x 7.2 cm hetrogeneous enhancing mass; edema; mass effect 4mg/day Dilantin EBRT TMZ(2) subtotal recurrence malignant glioma subtotal
66
+ HF1150 70-74 21.2 WHITE 90 contrast enhancing; ring enhancing; edema NONE Dilantin EBRT Temozolomide(11) NONE NONE NONE NONE
67
+ HF1156 30-34 105.3 WHITE 90 large T2 abnormality; tiny focal areas of enhancement NONE NASS EBRT NONE(NONE) gross total recurrence oligodendroglioma Gross Total
68
+ HF1167 45-49 17.9 BLACK noncontrast enhancing; mild mass effect NONE Dilantin EBRT Gliadel Wafer(1) subtotal recurrence oligodenroglioma and radiation necrosis subtotal
69
+ HF1185 25-29 95.2 WHITE 90 contrast enhancing minimally NONE Dilantin + Tegratol XR EBRT Temozolomide(12) subtotal x 3 recurrence Atypical oligodenroglioma; anaplastic oligodenroglioma x 2 subtotal
70
+ HF1191 70-74 0.3 WHITE nodular enhancement NONE Dilantin NONE NONE(NONE) NONE NONE NONE NONE
71
+ HF1199
72
+ HF1219 50-54 27.1 WHITE 90 contrast enhancing; mass effect 16 mg/day Dilantin EBRT Temozolomide(12) subtotal x 2 recurrence necrosis; GBM subtotal
73
+ HF1226
74
+ HF1227 25-29 251.7 WHITE 100 large T2 abnormality with nodular patchy enhancement NASS Yes Unknown EBRT TMZ(11) subtotal recurrence atypical oligodenroglioma subtotal
75
+ HF1232 50-54 20.2 WHITE 100 calcified; enhancing NASS NASS EBRT TMZ(11) NONE NONE NONE NONE
76
+ HF1235 35-39 98.4 WHITE 90 solitary mass without significant edema or enhancemnt NONE NONE NONE NONE(NONE) NONE NONE NONE NONE
77
+ HF1242 35-39 149 WHITE 80 enhancing mass with significant edema 16mg/day Dilantin EBRT TMZ(8) subtotal recurrence GBM subtotal
78
+ HF1246 65-69 0.2 WHITE 80 enhancing and non enhancing; leptomeningeal 4 mg t.i.d. NASS NONE NONE(NONE) NONE NONE NONE NONE
79
+ HF1264 45-49 89.1 WHITE non contrast enhancing NONE Tegretol and Dilantin NONE NONE NONE(NONE) NONE NONE NONE NONE
80
+ HF1269 45-49 13 WHITE 80 intense ray-like and nodular enhancements 12 mg q 6 hours Dilantin EBRT BCNU(4) subtotal recurrence Anaplastic Astrocytoma subtotal
81
+ HF1280 40-44 15.8 WHITE mulitple large 4cm enhancing lesions; vasogenic edema Yes Unknown NONE EBRT TMZ(12) NONE NONE NONE NONE
82
+ HF1292 45-49 57.7 ASIAN NOS 100 2cm x 3cm well circumscribed lesion; vasogenic edema NASS NASS EBRT TMZ(15) subtotal recurrence GBM subtotal
83
+ HF1293
84
+ HF1297 60-64 17.2 WHITE 80 linear hyperintense lesion NASS NASS EBRT TMZ(7) NONE NONE NONE NONE
85
+ HF1300
86
+ HF1307
87
+ HF1316 70-74 6.9 BLACK small foci of enhancment; small, old hemorrhages NASS EBRT NONE(NONE) NONE NONE NONE
88
+ HF1325 35-39 84.5 WHITE 90 non contrast enhancing NONE Carbamazepine EBRT NONE(NONE) subtotal recurrence oligoastrocytoma subtotal
89
+ HF1331
90
+ HF1334 20-24 115.7 WHITE enhancing; mass effect NASS Tegretol-XR EBRT Temozolomide(2) sub total x 2 recurrence anaplastic gemistocytic glioma; gemistocytic glioma subtotal
91
+ HF1344 60-64 9
92
+ HF1345 15-19 83.5 BLACK enhancing NASS Tegretol NONE NONE NONE(NONE) NONE NONE NONE NONE
93
+ HF1357 20-24 30.8 WHITE cystic, abnormal nodular enhancement; large T2 signal Yes Dose Unknown EBRT TMZ(5) subtotal recurrence subtotal
94
+ HF1381 25-29 136.2 BLACK non enhancing NASS Depakote No No NO(NO) NO NO NO NO
95
+ HF1397 60-64 30 WHITE large enhancing mass with vasogenic edema 4 mg b.i.d. Trileptal and Neurontin EBRT TMZ(8) NONE NONE NONE NONE
96
+ HF1398 55-59 66.8 WHITE 80 thick irregular shaggy enhancement with vasogenic edema and mass effect 8mg q. daily Dilantin EBRT TMZ+cRA(4) NONE NONE NONE NONE
97
+ HF1407 55-59 5.2 WHITE 80 vasogenic edema; subfalcine herniation NASS NASS EBRT TMZ(0) NONE NONE NONE NONE
98
+ HF1409 50-54 12.7 WHITE 90 multicentric lesion with numerous enhancing lesions NONE Depakote EBRT TMZ(1) subtotal recurrence GBM subtotal
99
+ HF1420
100
+ HF1429
101
+ HF1433 45-49 78.2 WHITE mass effect; necrosis; heterogeneous enhancement NASS Phenytoin EBRT NONE(NONE) NONE NONE NONE NONE
102
+ HF1437
103
+ HF1442 35-39 47.2 WHITE 100 non enhancing; mass effect NASS NASS EBRT TMZ(2) subtotal recurrence Anaplastic Astrocytoma subtotal
104
+ HF1458 45-49 8.8 WHITE 80 hetergeneous, hyperintense lobular and multicentric lesion; ring enhancing NASS EBRT PZA(2) NONE NONE NONE
105
+ HF1463 40-44 72 WHITE non enhancing NASS NASS EBRT TMZ(12) subtotal recurrence GBM subtotal
106
+ HF1475 15-19 31.7 WHITE 100 unusual enhancment 6mg/day NONE EBRT TMZ(22) subtotal recurrence undifferentiated malignant glioma subtotal
107
+ HF1489 50-54 68.3 WHITE 100 non enhancing None Dilantin EBRT Temozolomide(2) NO NO No No
108
+ HF1490 65-69 4 WHITE 100 ring enhancing NASS NASS EBRT NONE(NONE) NONE NONE NONE NONE
109
+ HF1502 35-39 6.4 WHITE 100 cystic; non-enhancing; edema None Keppra NO No No(No) NO NO No No
110
+ HF1510
111
+ HF1511 25-29 56.6 OTHER 100 non enhancing NASS NASS EBRT TMZ(5) subtotal recurrence GBM subtotal
112
+ HF1517 55-59 8.3 WHITE 100 enhancing; edema 8mg/day Dilantin EBRT TMZ(3) subtotal recurrence radiation necrosis subtotal
113
+ HF1538 70-74 3.2 WHITE large enhancing mass NASS NASS EBRT TMZ(0) NONE NONE NONE NONE
114
+ HF1540 75-79 10.2 BLACK 90 4.5cm large enhancing mass with subependymal spread; mild mass effect NONE NONE NONE EBRT NONE(NONE) NONE NONE NONE NONE
115
+ HF1551 30-34 70.9 WHITE 80 cystic; enhancing None Topiramate NO No Temozolomide(12) NO NO No No
116
+ HF1553 10-14 71.2 WHITE 100 cystic; ring enhancement None Dilantin NONE No No(No) NO NO No No
117
+ HF1560 75-79 1.2 WHITE 80 extensive vasogenic edema; bilateral frontal enhancing mass NONE NONE NONE NONE NONE(NONE) NONE NONE NONE NONE
118
+ HF1568 30-34 42.6 WHITE 100 minimal areas of enhancement; large T2 abnormality NONE NONE NONE EBRT Temodar(2) subtotal recurrence GBM subtotal
119
+ HF1587 30-34 75.3 WHITE 90 compression of the left lateral horn of the lateral ventricle; high intensity on diffusion; scattered areas of enhancement NASS NASS NONE EBRT Temodar(6) NONE NONE NONE NONE
120
+ HF1588 40-44 75.2 WHITE 90 mildly hypointense on T1; no enhancement NASS NASS NONE NONE NONE(NONE) NONE NONE NONE NONE
121
+ HF1606 60-64 73.6 WHITE 90 Ill defined hypointense on T1 images; minimal mass effect; no enhancement NONE Tegretol NONE NONE NONE(NONE) NONE NONE NONE NONE
122
+ HF1613 35-39 66.8 WHITE 100 no enhancement NASS NASS NO NO NO(NO) NO NO NO NO
123
+ HF1628 45-49 14.6 WHITE 100 enhancing mass NONE NONE EBRT TMZ(4) subtotal recurrence GBM and radiation necrosis subtotal
124
+ HF1652
125
+ HF1671 65-69 13.3 WHITE pre op CT only NASS NASS EBRT TMZ(1) subtotal recurrence CRN + minimal residual GBM subtotal
126
+ HF1677 50-54 63.5 WHITE 100 no enhancement; mild mass effect none Dilantin NO NO NO(NO) NO No NO NO
127
+ HF1702 75-79 5.4 WHITE 80 enhancing mass with vasogenic edema NASS NASS Brachytherapy NASS(NASS) NASS NASS NASS NASS
128
+ HF1708 20-24 67.8 WHITE nonenhancing abnormal signal intensity NASS NASS NONE NONE NONE(NONE) NONE NONE NONE NONE
samples/REMBRANDT.txt ADDED
@@ -0,0 +1,196 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ Rembrandt: Helping Personalized Medicine Become a Reality through Integrative Translational Research
2
+ Subha Madhavan; Jean-Claude Zenklusen; Yuri Kotliarov; Himanso Sahni; Howard A. Fine; Kenneth Buetow
3
+ Crossmark: Check for Updates
4
+ Author & Article Information
5
+ Mol Cancer Res (2009) 7 (2): 157–167.
6
+ https://doi.org/10.1158/1541-7786.MCR-08-0435
7
+ Article history
8
+ Split-Screen
9
+ Views Icon
10
+ Views
11
+ Click here to open pdf in another window
12
+ PDFfor
13
+ Share Icon
14
+ Share
15
+ Tools Icon
16
+ Tools
17
+ Search Site
18
+ Article Versions Icon
19
+ Versions
20
+ Abstract
21
+ Finding better therapies for the treatment of brain tumors is hampered by the lack of consistently obtained molecular data in a large sample set and the ability to integrate biomedical data from disparate sources enabling translation of therapies from bench to bedside. Hence, a critical factor in the advancement of biomedical research and clinical translation is the ease with which data can be integrated, redistributed, and analyzed both within and across functional domains. Novel biomedical informatics infrastructure and tools are essential for developing individualized patient treatment based on the specific genomic signatures in each patient's tumor. Here, we present Repository of Molecular Brain Neoplasia Data (Rembrandt), a cancer clinical genomics database and a Web-based data mining and analysis platform aimed at facilitating discovery by connecting the dots between clinical information and genomic characterization data. To date, Rembrandt contains data generated through the Glioma Molecular Diagnostic Initiative from 874 glioma specimens comprising ∼566 gene expression arrays, 834 copy number arrays, and 13,472 clinical phenotype data points. Data can be queried and visualized for a selected gene across all data platforms or for multiple genes in a selected platform. Additionally, gene sets can be limited to clinically important annotations including secreted, kinase, membrane, and known gene-anomaly pairs to facilitate the discovery of novel biomarkers and therapeutic targets. We believe that Rembrandt represents a prototype of how high-throughput genomic and clinical data can be integrated in a way that will allow expeditious and efficient translation of laboratory discoveries to the clinic. (Mol Cancer Res 2009;7(2):157–67)
22
+
23
+ Introduction
24
+ Primary brain tumors are a leading cause of cancer mortality in adults and children in the United States (1). The molecular and genetic heterogeneity of gliomas undoubtedly contributes to the varied and often suboptimal response to treatment that is usually predicated on standard pathologic diagnoses. Improvement in the prognosis of patients with gliomas will likely come about through the use of new targeted therapies based on the biological knowledge of the tumors at a molecular level.
25
+
26
+ To identify glioma-specific targets, consistent molecular characterization of a large number of tumors is required. To date, all the studies published have limitations due to incomplete coverage of whole-genome expression due to the usage of small or outdated, legacy, microarray platforms (2, 3), limited number of samples studied and/or incomplete inclusion of various different glioma subtypes and grades (4, 5), or the narrow scope of targets being investigated. Thus, we have put together a national, publicly funded effort that we call the Glioma Molecular Diagnostic Initiative (GMDI), which, coupled with its bioinformatics counterpart, Repository of Molecular Brain Neoplasia Data (Rembrandt), is designed to breach the gap of biological information related to primary brain tumors to help patients receive a better, biologically oriented therapy tailored to their specific needs.
27
+
28
+ Rembrandt is a powerful and intuitive informatics system designed to integrate genetic and clinical information for improved research, disease diagnosis, and treatment (as shown in Fig. 1). The platform supports clinical genomic research and (as data are collected and analyzed) will create a knowledge base that allows physicians to predict clinical outcomes and therapeutic efficacy based on an individual's clinical and genetic profiles, thereby enabling personalized medicine.
29
+
30
+ FIGURE 1.
31
+ FIGURE 1. Data integration via the Rembrandt discovery platform.
32
+ VIEW LARGEDOWNLOAD SLIDE
33
+ Data integration via the Rembrandt discovery platform.
34
+
35
+ To support discovery, the Rembrandt platform also allows researchers to search, import, and aggregate additional data from internal and external databases (such as GenBank, University of California at Santa Cruz golden path data sets, and Biocarta pathways), analyze the combined data sets to identify meaningful patterns (including specific chromosomal abnormalities), and share their research with other physicians and researchers within their own institution or in other physical locations. Each user is assigned a specific role that governs how much of the study data are accessible. A series of intuitive tools enable users to easily analyze and interact with the integrated data to achieve greater insight into molecular signatures that characterize each tumor and correlate with clinical outcome.
36
+
37
+ Unlike many biomedical database systems, Rembrandt is a fully integrated platform that supports multiple facets of clinical and molecular research, discovery, and hypothesis generation. This shared environment crosses many disciplines including genetic research and clinical care. As such, the platform should serve to foster cooperation and integration between research and clinical disciplines and expedite the time and increase the depth to which molecular data become relevant to the clinical environment.
38
+
39
+ Materials and Methods
40
+ Glioma Molecular Diagnostic Initiative
41
+ Sample Acquisition and Diagnosis. To better understand the genetic pathogenesis of gliomas and begin to identify potential glioma-specific molecular therapeutic targets, consistent molecular characterization of a large number of tumors is required.
42
+
43
+ This process was undertaken under a national prospective clinical trial that would eventually be institutional review board approved both within the National Cancer Institute intramural program and through both Cancer Therapy Evaluation Program-sponsored adult brain tumor consortia (NABTT and NABTC protocol 01-07). With the activation of this study, we collected matched tumor, blood, and plasma from the 14 contributing institutions (NIH, Henry Ford Hospital, Thomas Jefferson University, University of California at San Francisco, H. Lee Moffitt Hospital, University of Wisconsin, University of Pittsburgh Medical Center, University of California at Los Angeles, The University of Texas M. D. Anderson Cancer Center, Dana-Farber Cancer Center, Duke University, Johns Hopkins University, Massachusetts General Hospital, and Memorial Sloan Kettering Cancer Center). All tissues collected are sent to the Neuro-Oncology Branch laboratory for processing. The samples were provided as snap-frozen sections of areas immediately adjacent to the region used for the histopathologic diagnosis. Initial histopathologic diagnosis is done at the tissue collecting institution following the WHO standards (6). The initial diagnosis is reviewed by in-house neuropathologists to assure a measure of consistency across samples. To date, 874 complete frozen sample sets have been accrued, of those 389 are glioblastoma multiformes, 122 are astrocytomas, 113 are oligodendrogliomas, and 33 are mixed, with the reminder still unclassified.
44
+
45
+ Clinical data on the patients are collected prospectively until the patient's death through the NABTC Operations Office at The University of Texas M. D. Anderson Cancer Center and the NABTT Operations office at the Johns Hopkins University. The clinical data collected are updated into the Rembrandt database on a quarterly basis.
46
+
47
+ To assure consistency in the collection, shipment, processing, assaying, storage, data retrieval, and dissemination, we have put together a series of standard operating procedures that have resulted in a streamlined, high-throughput operation capable of handling large numbers of samples in a consistent, operator-independent fashion. Consistency of data over time is continuously monitored by looking for any signs of batch effect in the analyses.
48
+
49
+ mRNA Extraction and Gene Expression Data Processing. Tissue (∼50-80 mg) from each tumor was used to extract total RNA using the Trizol reagent (Invitrogen) following the manufacturer's instructions. The quality of RNA obtained was verified with the Bioanalyzer System (ref. 7; Agilent Technologies) using the RNA Pico Chips. RNA (5 μg) extracted from the accrued samples has been processed using U133 2 Plus mRNA expression chips (Affymetrix), which contains >54,000 probe sets analyzing the expression level of >47,000 transcripts and variants, including 38,500 well-characterized human genes.
50
+
51
+ All arrays were confirmed to be within an acceptable minimal quality-control according to internal standard operating procedure variables following these criteria: (a) A scaling factor of <5 when the expression values are scaled to a target mean signal intensity of 500. (b) Signal intensity ratios of the 3′ to 5′ end of the internal control genes of β-actin and GAPDH < 3. (c) Affymetrix spike control (BioC, BioDN, and CreX) are always present, and percentage present calls is >35% for brain tissue.
52
+
53
+ The .cel and .txt files of all the arrays that passed the minimal quality-control were input into dChip for normalization. The model-based expression index algorithm implemented in dChip selects an invariant set with a small within-subset rank difference to serve as basis for adjusting the brightness of the arrays to a comparable level. The normalization was done at the PM and MM probe levels, and model-based expression levels were calculated using normalized probe level data. We choose the average difference model (PM > MM) to compute expression values; negative average differences were truncated to 1 or log-transformed values of zeros to flag negative signal intensities with no biological meaning.
54
+
55
+ For data preprocessing, probe-level data were consolidated into probe-set data using the Affymetrix MAS5 algorithm, with the target scaling value at 500. Probe-level data were also processed with custom Chip Definition Files (1) that rearranged Affymetrix probes into gene-based probe sets. Probes mapped to alternatively spliced exons were grouped into distinct probe sets. Most 3′ probes were selected for processing. Nonspecific probes were masked before processing.
56
+
57
+ Single tumor samples were compared with the nontumor pool and the sample average to the nontumor pool. Samples were averaged based on tumor subtypes in six categories: glioblastoma multiforme, oligodendroglioma, astrocytoma, mixed, unclassified, and unknown tumors. Group comparisons were done in R with two sample t tests. Signal values were first transformed to logarithm (base 2). The averages of the log2 signals of tumor and nontumor groups were computed. The magnitude of the differences between the geometric means of expression levels for each reporter from the two groups was computed. The significance of the differences between tumors (or each tumor subtype) and nontumor samples for each reporter was also evaluated.
58
+
59
+ For each individual tumor sample, signals for each tumor and the ratio between each tumor and the average of normal (geometric means, computed the same way as described above) were computed. All processes were done separately for various data groups (public data and institution-based data).
60
+
61
+ DNA Extraction and Genomic Alteration Analysis. Tissue (∼10 μg; as recommended by the manufacturer) from each tumor was used to extract high molecular weight, genomic DNA using QIAamp DNA Micro DNA extraction kit (Qiagen) following the manufacturer's instructions. The quality of DNA was checked by electrophoresis run in a 2% agarose gel.
62
+
63
+ Genomic DNA (250 ng) from samples received has been hybridized to 100K single nucleotide polymorphism chips (ref. 8; Affymetrix), which covers 116,204 single nucleotide polymorphism loci in the human genome with a mean intermarker distance of 23.6 kb. These arrays give two simultaneous data types: allelic calls and signal intensity, allowing for the determination of both copy number alterations and regions of allelic imbalances (loss of heterozygosity). Calls were determined by the GTYPE software version 3.0 with 25% level of confidence. Only samples with call rates of >90% were accepted for any analysis.
64
+
65
+ Clinical Data Processing. The University of Texas M. D. Anderson Cancer Center serves as the operating center for clinical data collection for the GMDI trial. Clinical data reports from the case report forms were accessed through the Data Management Initiative Web portal at The University of Texas M. D. Anderson Cancer Center, parsed, and uploaded to the Rembrandt data warehouse after various preprocessing and data validations steps. The clinical data collected are updated into the Rembrandt database on a quarterly basis.
66
+
67
+ Results
68
+ A Rembrandt Storyboard
69
+ To exemplify the powerful integration that Rembrandt provides to analyze a large data set of both molecular and clinical data, we would like to show how one could come about to explore the validity of a scientific hypothesis using the system.
70
+
71
+ Suppose that one would have come across two publications on Glioma Tumor Stem Cells that mentioned the irregular expression of BMPR1B in such cells (9, 10).
72
+
73
+ A typical Rembrandt usage scenario might be to ask if BMPR1B is a potential therapeutic target as it has been recently been postulated to be involved in cell differentiation. To answer this question, a researcher can take a stepwise workflow approach in Rembrandt as shown in Figs. 2 and 3.
74
+
75
+ FIGURE 2.
76
+ FIGURE 2. A. Gene expression box plot for BMPR1B. Samples are categorized by histologic type. Different Affymetrix probe sets are shown as different color bars. B. BMPR1B probe set in Affymetrix probe-set viewer. Information for selected probe set can be displayed, allowing the user to decide on the quality of information retrieved. C. BMPR1B probe set of interest showing outliers in glioblastoma multiforme samples. The ability to display expression graphs in different formats allows the use to gain a wealth of information without having to redo the queries.
77
+ VIEW LARGEDOWNLOAD SLIDE
78
+ A. Gene expression box plot for BMPR1B. Samples are categorized by histologic type. Different Affymetrix probe sets are shown as different color bars. B. BMPR1B probe set in Affymetrix probe-set viewer. Information for selected probe set can be displayed, allowing the user to decide on the quality of information retrieved. C. BMPR1B probe set of interest showing outliers in glioblastoma multiforme samples. The ability to display expression graphs in different formats allows the use to gain a wealth of information without having to redo the queries.
79
+
80
+ FIGURE 3.
81
+ FIGURE 3. A. Kaplan-Meier survival plot showing survival comparing BMRP1 up-regulating samples and the rest of the gliomas in the database. This plot allows the identification of putative clinically relevant genes to explore as new targets for therapy. Users can query gene expression and graph changes in survival rate at each time point on the study. Kaplan-Meier estimates are calculated based on the last follow-up time and the censor status (0, alive; 1, dead) from the samples of interest. Kaplan-Meier estimates are then plotted against the survival time. Users can dynamically modify the fold change (up-regulation and down-regulation) thresholds and redraw the plot. A log-rank P value is provided as an indication of significance of the difference in survival between any two groups of samples segregated based on gene expression of the gene of interest. B. Performing PCA and correlating with clinical data. An example of PCA report from the Rembrandt application. These two-dimensional (top) and three-dimensional (bottom) graphs plot the various principal components from the gene expression PCA. Various analysis options are provided to the user to select from an array of gene/reporter filtering and sample selection settings. Users can select samples in the two-dimensional plot to retrieve related clinical information on the selected patients.
82
+ VIEW LARGEDOWNLOAD SLIDE
83
+ A. Kaplan-Meier survival plot showing survival comparing BMRP1 up-regulating samples and the rest of the gliomas in the database. This plot allows the identification of putative clinically relevant genes to explore as new targets for therapy. Users can query gene expression and graph changes in survival rate at each time point on the study. Kaplan-Meier estimates are calculated based on the last follow-up time and the censor status (0, alive; 1, dead) from the samples of interest. Kaplan-Meier estimates are then plotted against the survival time. Users can dynamically modify the fold change (up-regulation and down-regulation) thresholds and redraw the plot. A log-rank P value is provided as an indication of significance of the difference in survival between any two groups of samples segregated based on gene expression of the gene of interest. B. Performing PCA and correlating with clinical data. An example of PCA report from the Rembrandt application. These two-dimensional (top) and three-dimensional (bottom) graphs plot the various principal components from the gene expression PCA. Various analysis options are provided to the user to select from an array of gene/reporter filtering and sample selection settings. Users can select samples in the two-dimensional plot to retrieve related clinical information on the selected patients.
84
+
85
+ Explore the expression levels of BMPR1B in different subtypes of glioma. Analysis of the box plots in Rembrandt (Fig. 2A) indicates that probe-set 210523_at is differentially expressed in glioblastoma multiformes when compared with nontumors (borderline significance: P < 0.04).
86
+
87
+ Where does this probe map onto the transcripts of BMPR1B? Review of probe mapping in Affymetrix probe viewer integrated into Rembrandt (Fig. 2B) shows that this probe maps to coding region.
88
+
89
+ Are there two subpopulations of BMPR1B regulating samples? Review of the “box and whisker” plot in Fig. 2C indicates that glioblastoma multiformes have low-end outliers for BMPR1B expression.
90
+
91
+ Now, can we identify samples that show high (up >2) and low (down <1.5) expression of BMPR1B? Advanced queries can be set up in the Rembrandt application to create sample sets with separate up-regulation and down-regulation criteria for BMPR1 expression.
92
+
93
+ Does BMPR1 up-regulation affect survival? Can this sample group be compared with the rest of the gliomas? Figure 3A shows the difference in probability of survival between BMRP1 up-regulating group and the rest of the gliomas. Results indicate that BMPR1B up-regulation is bad as a prognostic factor and could be a good target for therapy.
94
+
95
+ How different are these sample groups beyond BMPR1B expression? By analyzing the whole gene expression patterns in both groups using the high-order analysis tool of principal component analysis (PCA; Fig. 3B), it is possible to see that BMPR1B overexpressors and underexpressors are indeed quite different at a global expression level, suggesting that this gene may hold a key to glioma diversity.
96
+
97
+ The storyboard here presented indicates that Rembrandt can effectively be used to test in silico a scientific hypothesis and allow for additional experimentation to occur. In fact, this has been the case with the scenario here presented and we have shown that BMPR1B is able in fact to modulate the tumorigenic potential of glioma cells (11). Additionally, a Rembrandt search of newly identified (NF1) and well-known (IGFBP2) targets of deregulation in gliomas shows that the result produced by our data set are concordant with the current knowledge of clinical features (Supplementary Fig. S1).
98
+
99
+ Key Features in Rembrandt
100
+ Integrating Genome Characterization Data with Clinical Outcomes
101
+ Users can query gene expression or copy number data and graph changes in survival rate at each time point in the study. Kaplan-Meier estimates are calculated based on the last follow-up time and the censor status (0 = alive, 1 = dead) from the samples of interest. Kaplan-Meier estimates are then plotted against survival time (Fig. 3A). The points that correspond to the events with a censor status of 0 are indicated on the graph. Users can dynamically modify the fold change (up-regulation and down-regulation) thresholds and redraw the plot. A log-rank P value is provided as an indication of significance of the difference in survival between any two groups of samples segregated based on gene expression of the gene of interest. The log-rank P value is calculated using the Mantel-Haenszel procedure (12). P values are recalculated every time a new threshold is selected. Users can toggle to a unified gene expression view with lesser reporters to get a gene-based view of the expression data. To obtain the unified gene expression values, the probe-level data are processed with custom Chip Definition Files that rearrange Affymetrix probes into gene-based probe sets. Probes mapped to alternatively spliced exons are grouped into distinct probe sets. Most 3′ probes are selected for processing. Nonspecific probes are masked before processing. Similar to Kaplan-Meier plots for differential fold change analysis, Kaplan-Meier plots can be drawn for copy number data where genes are mapped to single nucleotide polymorphism probe sets by aligning the probe's physical position to aligned mRNA sequences plus 50 kb upstream and downstream for maximum coverage. Also, Kaplan-Meier plots can be drawn by selecting two patient groups of interest. These groups can be user-defined or predefined lists of patients.
102
+
103
+ Performing Higher-Order Statistical Analysis on Genomic and Clinical Data Set
104
+ Rembrandt supports computer-intensive, high-memory utilizing tasks such as higher-order gene expression analyses (such as class comparison, clustering, and PCA), where the data sets could be as large as 4 GB with an analytic cluster to allow for several simultaneous analytic jobs.
105
+
106
+ Figure 3B shows an example of a PCA report from the Rembrandt application. This two-dimensional graph plots the various principal components from the gene expression PCA. Various analysis options are provided from which users can select gene/reporter filtering and sample selection settings. Users can click on the three tabs at the top of the graph to display PC1 versus PC2, PC1 versus PC3, or PC2 versus PC3. Each point on the graph represents a sample. The samples are colored by disease type. Users can click on the link on the top left-hand corner of the graph to color by gender. Patients with different survival ranges are indicated by different shapes on the graph. Users can select samples of interest by clicking on the graph and drawing a rectangle around samples to save them for future use.
107
+
108
+ GenePattern Link
109
+ Broad's GenePattern (13) combines a powerful scientific workflow platform with >90 computational and visualization tools for the analysis of genomic data. To expand a researcher's ability to analyze the glioma data sets, Rembrandt has been seamlessly integrated with GenePattern. Shown in Fig. 4A is an expression heat map of 50 additional genes that have expression patterns related to stem cell factor (14) in glioblastoma multiforme.
110
+
111
+ FIGURE 4.
112
+ FIGURE 4. A. Heat-map view in GenePattern. Subsets of data from Rembrandt can be transferred to GenePattern using standard interfaces to invoke several run-time data analysis capabilities. A heat map for 50 neighbors of stem cell factor is shown for astrocytoma and mixed glioma samples in Rembrandt. B. Scatter plot for copy number data across physical genomic locations. Scatter plots display measured copy number against physical genome location in an application called webGenome, which has been integrated with Rembrandt via standard data interfaces. These plots are context sensitive to the copy number reports generated from the copy number queries in the caIntegrator application. Users can view data at arbitrary resolutions from the entire genome on down.
113
+ VIEW LARGEDOWNLOAD SLIDE
114
+ A. Heat-map view in GenePattern. Subsets of data from Rembrandt can be transferred to GenePattern using standard interfaces to invoke several run-time data analysis capabilities. A heat map for 50 neighbors of stem cell factor is shown for astrocytoma and mixed glioma samples in Rembrandt. B. Scatter plot for copy number data across physical genomic locations. Scatter plots display measured copy number against physical genome location in an application called webGenome, which has been integrated with Rembrandt via standard data interfaces. These plots are context sensitive to the copy number reports generated from the copy number queries in the caIntegrator application. Users can view data at arbitrary resolutions from the entire genome on down.
115
+
116
+ Plotting Copy Number Data from Patient DNA Samples against Genomic Location
117
+ Scatter plots (shown in Fig. 4B) display measured copy number against the physical genome location in an application called webGenome, which has been integrated with Rembrandt. These plots are context sensitive to the copy number reports generated from the copy number queries in the Rembrandt application. Users can view data at arbitrary resolutions from the entire genome on down. When users move the mouse over specific probes, the system provides mouse-over probe names. Clicking on the name of an experiment or bioassay in the plot legend will highlight the corresponding data.
118
+
119
+ Advanced Query and Report Interfaces
120
+ Biomedical researchers struggle to meaningfully integrate their findings across multiple data types. Cancer is a complex disease requiring genomic, proteomic, pathology, imaging, and clinical data for a true understanding of the scope of the problem. Advanced query interfaces (as shown in Fig. 5) in Rembrandt enable this meaningful integration across data types. It allows users to mine the Rembrandt database using various genomic and clinical criteria. These queries can be combined to arrive at reports (shown in Fig. 6) that integrate data from various data domains, such as gene expression, copy number analysis, and clinical trials. Several filtering and data download options are presented in Rembrandt reports.
121
+
122
+ FIGURE 5.
123
+ FIGURE 5. User-friendly data query interface. Query pages enable users to restrict their searches in the database to specific genomic and/or clinical criteria.
124
+ VIEW LARGEDOWNLOAD SLIDE
125
+ User-friendly data query interface. Query pages enable users to restrict their searches in the database to specific genomic and/or clinical criteria.
126
+
127
+ FIGURE 6.
128
+ FIGURE 6. Gene expression fold report. All reports in Rembrandt are fully customizable at the report window, making unnecessary to re-run queries to refine the results.
129
+ VIEW LARGEDOWNLOAD SLIDE
130
+ Gene expression fold report. All reports in Rembrandt are fully customizable at the report window, making unnecessary to re-run queries to refine the results.
131
+
132
+ Rembrandt System Architecture
133
+ Rembrandt was developed using a n-tier architecture. The system was developed using Java 2 Enterprise Edition, a hybrid star data warehouse schema and various open source technologies. The back end consists of an Oracle 10g database for storing precomputed microarray differential expression, computed copy number, clinical data, and user security information. For performance reasons, normalized gene expression data used by the real-time analysis module are stored as R-binary files. The middle tier, which handles application logic and core functionality, was developed using Java and cancer Biomedical Informatics Grid software development and compatibility guidelines (15). Rembrandt application consists of standard interfaces that enable integration with third-party tools such as caArray, webGenome, and GenePattern. Rembrandt has an Analytical Server that provides on-the-fly computational analysis capability. The Analytical Server communicates asynchronously with Rembrandt's middle tier via the Java Messaging Service. Java Messaging Service allows Rembrandt to abstract the statistical packages being used for heavy computational tasks.
134
+
135
+ Rembrandt Cancer Biomedical Informatics Grid Service
136
+ Basic and clinical research has increasingly become dependent on advanced information technologies for management, exchange, and analysis of diverse biomedical data. Although a wealth of information is collected by the cancer research community, any one given researcher is faced with challenges in discovering, extracting, and analyzing the information relevant to his/her research. To address this need, the National Cancer Institute has initiated a national-scale effort, called the cancer Biomedical Informatics Grid, to develop a federation of interoperable research information systems. At the heart of the cancer Biomedical Informatics Grid approach to federated interoperability effort is a Grid middleware infrastructure, called caGrid (16). caGrid Data Services provide the means to share data via the caGrid federated infrastructure. One of the major goals of the current release of Rembrandt was to create a clinical genomic object model and expose the domain model through a caGrid data service. The purpose of the object model is to help capture the relationships between the clinical study and its associated experimental observations. The Rembrandt caGrid service can be used to obtain programmatic access to public data in Rembrandt in a federated fashion and can be found at http://caintegrator.nci.nih.gov/wsrf-rbt/services/cagrid/RembrandtGridService.
137
+
138
+ Conclusion
139
+ Large-scale data sets from genomics, proteomics, population genetics, and imaging are driving research at a previously unprecedented pace. Bioinformatics data management providers must serve these data sets in a usable way that helps find the needle in a haystack effectively and accurately. The goal of the “omic” sciences is not to generate numbers but rather “insight.” The Web interfaces are burdened with displaying terabytes of data in ways that physician scientists can comprehend and use the results to develop hypothesis for their next study or trial. Ultimately, we feel that information must be standardized, integrated, and made available at the point of care to help patients and physicians make optimal decisions.
140
+
141
+ Tools such as Rembrandt have primarily focused on the usability aspect of high-throughput heterogeneous data and yet enabling power users and bioinformaticians to tap into runtime analysis tools such as gene pattern or use the programmatic interfaces that are provided via the caGrid service. From a technical standpoint, the Rembrandt platform provides developer tools for a highly scalable system to include new data types (as shown in Fig. 1) and connect with existing ones to present integrated data views to users. This flexible discovery informatics platform has aided in implementing data portals to host several other cancer clinical data sets including those from the I-SPY stage III breast cancer study and The Cancer Genome Atlas (TCGA; ref. 17) project data included in the Cancer Molecular Analysis Portal. In this respect, it is worth to point out that the new Cancer Molecular Data portal has reutilized many of the features available in Rembrandt to suit a more general set of tumor sample analysis. At the sample level, GMDI and TCGA are complementary in many levels. GMDI is a prospective study wherein 14 institutions recruited patients with any type of glioma giving a wide spectrum of demographic sampling due to the geographic dispersion of the sites. The TCGA sample collection pipeline included two centers that had retrospective sample collections of glioblastoma multiforme. Thus, TCGA focused its analysis on high-grade glioblastoma multiformes, whereas the samples in GMDI represent all glioma grades and subtypes described in the WHO classification, allowing for studies on the differences of gliomas as they progress. The clinical data obtained by the GMDI project are comprehensive, because the study was conceived as a prospective, natural history clinical trial, thus allowing for the collection of a wide range of clinical data points. On the other hand, the TCGA project, in virtue of its more focused nature, has produced more molecular data types (methylation, sequencing, and miRNA expression) than GMDI. However, the GMDI samples are being used to acquire those data types, and they will be incorporated to Rembrandt as sufficient numbers of samples are processed.
142
+
143
+ The ultimate beneficiaries of Rembrandt are the brain tumor patients themselves. Rembrandt is designed to bridge the gap between biological and clinical information to help patients receive a better, biologically oriented therapy tailored to their specific needs. As such, we plan to incorporate new and useful capabilities in future releases that are not available at present time, such as the ability for researchers to incorporate their own data to the system to compare with the large data set already in the database. It is hoped that the GMDI and Rembrandt will provide a much needed resource for scientists and physicians combating brain cancer, and ultimately other forms of cancer, for providing the data and bioinformatics tool set that may allow the development of a biologically and clinically significant pathologic classification of brain tumors and help elucidate novel molecular targets for therapy.
144
+
145
+ Availability
146
+ Rembrandt is freely available to all users at https://caintergator.nci.nih.gov/rembrandt. The source code for Rembrandt is also available under a nonviral cancer Biomedical Informatics Grid license at https://gforge.nci.nih.gov/frs/download.php/1489/rembrandt_1_0.zip. The Rembrandt caGrid service is accessible at http://caintegrator.nci.nih.gov/wsrf-rbt/services/cagrid/RembrandtGridService.
147
+
148
+ Web Resources
149
+ Rembrandt clinical genomics object model: http://Rembrandt.nci.nih.gov/content/Rembrandtlfs/RembrandtEA1.0docs/index.htm.
150
+
151
+ Rembrandt clinical genomics data model: http://Rembrandt.nci.nih.gov/developers/images/db_model2.jpg.
152
+
153
+ Rembrandt application: http://rembrandt-db.nci.nih.gov.
154
+
155
+ Rembrandt information site: http://rembrandt.nci.nih.gov.
156
+
157
+ webGenome: http://webgenome.nci.nih.gov/webgenome/home.do.
158
+
159
+ GenePattern: http://www.broad.mit.edu/cancer/software/genepattern/.
160
+
161
+ caArray: https://array.nci.nih.gov/caarray/home.action.
162
+
163
+ I-SPY trial: http://ncicb.nci.nih.gov/tools/translation_research/ispy.
164
+
165
+ TCGA: http://cancergenome.nih.gov/.
166
+
167
+ Cancer molecular analysis portal (access to TCGA data sets): http://cma.nci.nih.gov.
168
+
169
+ Disclosure of Potential Conflicts of Interest
170
+ No potential conflicts of interest were disclosed.
171
+
172
+ Grant support: Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research and National Institute of Neurological Disorders and Stroke.
173
+ The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
174
+ Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/).
175
+ S. Madhavan and J-C. Zenklusen contributed equally to this work.
176
+ Acknowledgments
177
+ We thank Anand Basu, Shine Jacobs, Alex Jiang, Huaitian Liu, Ram Bhattaru, Michael Harris, Kevin Rosso, Ryan Landy, Hangjiong Chen, and Ying Long for contributions to Rembrandt software development and data loading; George Komatsoulis for reviewing data sharing policies and contributing to the Rembrandt domain information model; Carl Schaefer and Tracy Lively for reviewing usecases and interim releases of the software; Juli Klemm for helping with the integration of Rembrandt with caArray data repository; Jill Hadfield for technical documentation; David Hall, Dean Jackman, and Vessalina Bakalov for efforts on webGenome; and The University of Texas M. D. Anderson Cancer Center Data Management Initiative team for making the clinical reports available from the NABTC GMDI study for populating the Rembrandt database.
178
+
179
+ References
180
+ 1Cancer Statistics Branch, NIH. Cancer survival rates. In: Harras A, editor. Cancer: rates & risks. Washington (DC): U.S. Department of Health & Human Services, NIH; 1996. p. 28–34.
181
+ 2Nutt CL, Mani DR, Betensky RA, et al. Gene expression-based classification of malignant gliomas correlates better with survival than histological classification. Cancer Res 2003;63:1602–7.
182
+ 3Mischel PS, Shai R, Shi T, et al. Identification of molecular subtypes of glioblastoma by gene expression profiling. Oncogene 2003;22:2361–73.
183
+ 4Phillips HS, Kharbanda S, Chen R, et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 2006;9:157–73.
184
+ 5Nigro JM, Misra A, Zhang L, et al. Integrated array-comparative genomic hybridization and expression array profiles identify clinically relevant molecular subtypes of glioblastoma. Cancer Res 2005;65:1678–86.
185
+ 6Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol (Berl) 2007;114:97–109.
186
+ 7Miller CL, Diglisic S, Leister F, Webster M, Yolken RH. Evaluating RNA status for RT-PCR in extracts of postmortem human brain tissue. Biotechniques 2004;36:628–33.
187
+ 8Matsuzaki H, Dong S, Loi H, et al. Genotyping over 100,000 SNPs on a pair of oligonucleotide arrays. Nat Methods 2004;1:109–11.
188
+ 9Lee J, Kotliarova S, Kotliarov Y, et al. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 2006;9:391–403.
189
+ 10Galli R, Binda E, Orfanelli U, et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 2004;64:7011–21.
190
+ 11Lee J, Son MJ, Woolard K, et al. Epigenetic-mediated dysfunction of the bone morphogenetic protein pathway inhibits differentiation of glioblastoma-initiating cells. Cancer Cell 2008;13:69–80.
191
+ 12Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958;53:457–81.
192
+ 13Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Genet 2006;38:500–1.
193
+ 14Sun L, Hui AM, Su Q, et al. Neuronal and glioma-derived stem cell factor induces angiogenesis within the brain. Cancer Cell 2006;9:287–300.
194
+ 15Phillips J, Chilukuri R, Fragoso G, Warzel D, Covitz PA. The caCORE Software Development Kit: streamlining construction of interoperable biomedical information services. BMC Med Inform Decis Mak 2006;6:2.
195
+ 16Oster S, Langella S, Hastings S, et al. caGrid 1.0: an enterprise Grid infrastructure for biomedical research. J Am Med Inform Assoc 2008;15:138–49.
196
+ 17McLendon R, Friedman A, Bigner D, et al. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008;455:1061–8.
samples/REMBRANDT_description.txt ADDED
@@ -0,0 +1,23 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ Finding better therapies for the treatment of brain tumors, is
2
+ hampered for several reasons: 1) lack of consistently obtained
3
+ molecular data in a large sample sets; and 2) the ability to integrate
4
+ biomedical data from disparate sources, which would enable translation
5
+ of therapies from bench to bedside. Hence, a critical factor in the
6
+ advancement of biomedical research and clinical translation is the
7
+ ease with which data can be obtained, integrated, and analyzed both
8
+ within and across functional domains. Novel biomedical informatics
9
+ infrastructure and tools are essential for developing individualized
10
+ patient treatment based on the specific genomic signatures in each
11
+ patient's tumor. The Repository of Molecular Brain Neoplasia Data
12
+ (REMBRANDT) aims to facilitate discovery by connecting the dots
13
+ between clinical information and genomic characterization data.
14
+
15
+ REMBRANDT contains data generated through the Glioma Molecular
16
+ Diagnostic Initiative from 874 glioma specimens, comprised of
17
+ approx. 566 gene expression arrays, 834 copy number arrays, and 13,472
18
+ clinical phenotype data points. These data are currently housed in
19
+ Georgetown University's G-DOC System and are described in a related
20
+ manuscript. The TCIA image collection was created as a companion data
21
+ set to augment the larger REMBRANDT project. It contains the
22
+ pre-surgical magnetic resonance (MR) multi-sequence images from 130
23
+ REMBRANDT patients.
samples/ROI-Masks-leaderboard.tsv ADDED
@@ -0,0 +1,189 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ Tumor 1p_19q_co_del_status neoplasm_histologic_grade ProgFreeSurvEvent01 ProgFreeSurvTime_days IDH/1p19q Subtype death01 TERT-Promoter ATRX CIC EGFR TP53 histological_type gender age_at_initial_pathologic cqcf_method_of_sample_procuremen karnofsky_performance_sco primary_radiation_therapy primary_pharma_therapy RNASeqCluster miRNACluster CNCluster OncosignCluster
2
+ TCGA-CS-4938 0 G2 0 143 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 31 Gross Total Resection 90 NA NA R1 mi1 C1 O2
3
+ TCGA-CS-4941 0 G3 1 9 0 1 NA wt wt wt wt Astrocytoma MALE 67 Gross Total Resection 90 NO NA R2 mi2 C2 O3
4
+ TCGA-CS-4942 0 G3 1 1184 2 1 NA Mutant wt wt Mutant Astrocytoma FEMALE 44 Gross Total Resection 90 NO NO R1 mi2 C1 O2
5
+ TCGA-CS-4943 0 G3 0 552 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 37 Gross Total Resection 50 NA NO R1 mi2 C1 O2
6
+ TCGA-CS-4944 0 G2 NA NA 2 0 NA wt wt wt wt Astrocytoma MALE 50 Gross Total Resection 90 NA NA NA mi2 C1 O1
7
+ TCGA-CS-5390 1 G2 0 1966 1 0 NA wt wt wt wt Oligodendroglioma FEMALE 47 Gross Total Resection 100 YES NO R3 mi2 C3 O1
8
+ TCGA-CS-5393 0 G3 0 1222 2 0 NA wt wt wt Mutant Astrocytoma MALE 39 Gross Total Resection 100 NA NA R4 mi2 C1 O3
9
+ TCGA-CS-5394 1 G3 NA NA 1 0 NA wt Mutant wt wt Astrocytoma MALE 40 Gross Total Resection NA NA NA R3 mi2 C3 O1
10
+ TCGA-CS-5395 0 G2 1 287 0 1 Mutant wt wt wt wt Oligodendroglioma MALE 43 Gross Total Resection 90 YES NO R2 mi2 C2 O3
11
+ TCGA-CS-5396 1 G3 0 303 1 0 NA wt wt wt Mutant Oligodendroglioma FEMALE 53 Gross Total Resection 90 YES YES R3 mi2 C3 O2
12
+ TCGA-CS-5397 0 G3 1 194 0 1 NA wt wt wt wt Astrocytoma FEMALE 54 Gross Total Resection 80 YES YES NA mi1 C2 O3
13
+ TCGA-CS-6186 0 G3 1 188 0 1 NA wt wt wt wt Oligoastrocytoma MALE 58 Gross Total Resection 90 YES NO R2 mi1 C2 O3
14
+ TCGA-CS-6188 0 G3 1 647 0 0 NA wt wt wt wt Astrocytoma MALE 48 Gross Total Resection 90 YES NA R2 mi3 C2 O3
15
+ TCGA-CS-6290 0 G3 0 546 2 0 NA wt wt wt Mutant Astrocytoma MALE 31 Gross Total Resection 90 NA NA R1 mi2 C1 O2
16
+ TCGA-CS-6665 0 G3 0 378 2 0 NA wt wt wt Mutant Astrocytoma FEMALE 51 Gross Total Resection 90 YES NA R2 mi1 C1 O2
17
+ TCGA-CS-6666 0 G3 0 258 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 22 Gross Total Resection 90 NA NA NA mi2 C1 O2
18
+ TCGA-CS-6667 0 G2 0 230 2 0 NA wt wt wt Mutant Astrocytoma FEMALE 39 Gross Total Resection 90 YES NA R1 mi1 C1 O2
19
+ TCGA-CS-6668 1 G2 0 244 1 0 Mutant wt Mutant wt wt Oligodendroglioma FEMALE 57 Gross Total Resection 90 NA NA R3 mi2 C3 O1
20
+ TCGA-CS-6669 0 G2 0 225 0 0 wt wt wt wt wt Oligodendroglioma FEMALE 26 Gross Total Resection 90 NA YES R4 mi1 C1 O1
21
+ TCGA-DU-5849 1 G2 0 443 1 0 NA wt wt wt wt Oligodendroglioma MALE 48 Biopsy Only NA NA YES R4 mi2 C3 O1
22
+ TCGA-DU-5851 0 G3 0 531 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 40 Biopsy Only 90 YES YES R4 mi1 C1 O2
23
+ TCGA-DU-5852 0 G3 1 24 0 1 NA wt wt Mutant wt Oligoastrocytoma FEMALE 61 Subtotal Resection 80 NO NA R2 mi4 C2 O3
24
+ TCGA-DU-5853 0 G2 0 407 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 29 Gross Total Resection 100 YES YES R1 mi2 C1 O2
25
+ TCGA-DU-5854 0 G3 1 202 0 0 NA wt wt wt wt Astrocytoma FEMALE 57 Biopsy Only 90 NA NA R2 mi1 C2 O3
26
+ TCGA-DU-5855 0 G3 0 207 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 49 Subtotal Resection 100 YES YES R1 mi4 C1 O2
27
+ TCGA-DU-5871 0 G2 0 576 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 37 Biopsy Only 100 NA NA R1 mi2 C1 O2
28
+ TCGA-DU-5872 0 G2 1 265 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 43 Biopsy Only NA YES YES NA mi4 C1 O2
29
+ TCGA-DU-5874 1 G2 0 461 1 0 Mutant wt wt wt wt Oligodendroglioma FEMALE 62 Subtotal Resection 100 NA YES R3 mi2 C3 O1
30
+ TCGA-DU-6395 0 G2 1 1197 2 1 NA Mutant wt wt Mutant Oligoastrocytoma MALE 31 Subtotal Resection NA NA NO R1 mi2 C1 O2
31
+ TCGA-DU-6397 1 G3 1 837 1 1 NA wt wt wt wt Oligodendroglioma MALE 45 Subtotal Resection NA YES NO R3 mi2 C3 O1
32
+ TCGA-DU-6399 0 G2 1 1629 0 1 NA Mutant wt wt Mutant Oligodendroglioma MALE 54 Subtotal Resection NA YES NA R1 mi2 C1 O2
33
+ TCGA-DU-6400 1 G2 1 37 1 1 NA wt Mutant wt wt Oligodendroglioma FEMALE 66 Subtotal Resection NA NA NA R3 mi2 C3 O1
34
+ TCGA-DU-6401 0 G2 1 1886 2 1 wt Mutant wt wt Mutant Oligodendroglioma FEMALE 31 Subtotal Resection NA NO NO R1 mi2 C1 O2
35
+ TCGA-DU-6402 0 G3 1 103 0 1 NA wt wt wt wt Astrocytoma MALE 52 Subtotal Resection NA YES NO R2 mi3 C2 O3
36
+ TCGA-DU-6404 0 G3 1 1154 0 1 NA wt wt wt wt Oligodendroglioma FEMALE 24 Gross Total Resection 100 NA NA R2 mi1 C1 O2
37
+ TCGA-DU-6405 0 G3 1 486 0 1 NA wt wt wt wt Astrocytoma FEMALE 51 Subtotal Resection 90 YES YES R2 mi1 C2 O3
38
+ TCGA-DU-6407 0 G2 1 2397 2 1 wt Mutant wt wt Mutant Oligodendroglioma FEMALE 35 Subtotal Resection 90 NA NA R1 mi1 C1 O2
39
+ TCGA-DU-6408 0 G3 1 2097 2 1 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 23 Subtotal Resection 90 NA NO NA mi2 C1 O2
40
+ TCGA-DU-6410 1 G3 0 242 1 0 NA wt wt wt wt Oligodendroglioma MALE 56 Subtotal Resection 50 NA YES R3 mi4 C3 O1
41
+ TCGA-DU-6542 0 G3 NA NA 2 0 NA wt wt wt Mutant Oligoastrocytoma MALE 25 Subtotal Resection NA NA NA NA mi2 C1 O2
42
+ TCGA-DU-7008 0 G2 1 330 2 0 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 41 Subtotal Resection NA NA NA R4 mi2 C1 O2
43
+ TCGA-DU-7010 0 G3 1 193 2 1 NA wt wt wt Mutant Astrocytoma FEMALE 58 Subtotal Resection NA NA NA R2 mi2 C2 O2
44
+ TCGA-DU-7013 0 G3 1 187 0 1 NA wt wt wt Mutant Astrocytoma MALE 59 Subtotal Resection 80 NA NA R2 mi3 C2 O2
45
+ TCGA-DU-7014 0 G2 1 3437 NA 1 NA NA NA NA NA Oligodendroglioma MALE 59 Subtotal Resection 100 NA NA R1 mi1 C1 NA
46
+ TCGA-DU-7015 0 G2 1 591 2 0 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 41 Subtotal Resection 90 NA NA R1 mi2 C1 O2
47
+ TCGA-DU-7018 1 G3 1 338 1 1 NA wt Mutant wt wt Oligodendroglioma FEMALE 57 Subtotal Resection 90 NA NA R3 mi2 C3 O1
48
+ TCGA-DU-7019 0 G3 0 800 2 0 NA wt wt wt Mutant Oligoastrocytoma MALE 39 Subtotal Resection 100 YES YES NA mi2 C1 O2
49
+ TCGA-DU-7294 1 G2 0 2869 1 0 NA wt Mutant wt wt Oligodendroglioma FEMALE 53 Subtotal Resection 100 NA NA R3 mi2 C3 O1
50
+ TCGA-DU-7298 0 G3 1 200 2 1 NA Mutant wt wt Mutant Astrocytoma FEMALE 38 Subtotal Resection 80 NA NA R4 mi2 C1 O2
51
+ TCGA-DU-7299 0 G3 1 675 2 1 NA wt wt wt Mutant Astrocytoma MALE 33 Subtotal Resection 90 YES NA NA mi4 C1 O2
52
+ TCGA-DU-7300 1 G3 1 317 1 1 NA wt wt wt Mutant Oligodendroglioma FEMALE 53 Subtotal Resection 90 NA NA R4 mi1 C3 O1
53
+ TCGA-DU-7301 0 G2 1 366 2 1 wt Mutant wt wt Mutant Oligodendroglioma MALE 53 Subtotal Resection 100 NA NA R1 mi2 C1 O2
54
+ TCGA-DU-7302 1 G3 1 1374 1 0 NA wt Mutant wt wt Oligodendroglioma FEMALE 48 Subtotal Resection 90 NA NA R4 mi2 C3 O1
55
+ TCGA-DU-7304 0 G3 1 309 2 1 NA Mutant wt wt Mutant Oligoastrocytoma MALE 43 Gross Total Resection 80 NA NA R4 mi1 C1 O2
56
+ TCGA-DU-7306 0 G2 1 1250 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 67 Subtotal Resection 100 YES YES NA mi2 C1 O2
57
+ TCGA-DU-7309 0 G3 0 84 2 0 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 41 Gross Total Resection 90 NA NA R4 mi1 C1 O2
58
+ TCGA-HT-7684 0 G3 0 184 2 0 NA wt Mutant wt wt Oligoastrocytoma FEMALE 29 Gross Total Resection NA NO NO R4 mi2 C3 O1
59
+ TCGA-HT-7681 1 G2 0 1359 1 0 NA wt wt wt wt Oligoastrocytoma MALE 58 Gross Total Resection NA NA NA NA mi1 C3 O1
60
+ TCGA-HT-7686 0 G3 0 1300 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 29 Gross Total Resection NA YES YES R2 mi3 C1 O2
61
+ TCGA-HT-7687 1 G3 NA NA 1 0 NA wt wt wt wt Oligodendroglioma MALE 74 Gross Total Resection NA NA NA R3 mi2 C3 O1
62
+ TCGA-HT-7688 0 G3 0 964 2 0 NA wt wt wt Mutant Oligodendroglioma MALE 59 Gross Total Resection NA YES YES R4 mi1 C1 O2
63
+ TCGA-HT-7689 0 G2 0 455 2 0 wt Mutant wt wt Mutant Oligodendroglioma FEMALE 58 Gross Total Resection 60 YES YES R1 mi2 C1 O2
64
+ TCGA-HT-7690 0 G3 0 3 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 29 Gross Total Resection NA NA NA R2 mi2 C1 O2
65
+ TCGA-HT-7691 0 G2 0 3 0 0 NA wt wt wt wt Astrocytoma FEMALE 31 Gross Total Resection NA NA NA R2 mi1 C1 O1
66
+ TCGA-HT-7692 1 G2 0 90 1 0 NA wt wt wt wt Oligoastrocytoma MALE 43 Gross Total Resection 100 YES NA R3 mi2 C3 O1
67
+ TCGA-HT-7693 0 G2 0 533 2 0 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 51 Gross Total Resection 90 NA NA R1 mi2 C3 O2
68
+ TCGA-HT-7694 1 G3 0 210 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 60 Gross Total Resection 90 YES NA R4 mi1 C3 O1
69
+ TCGA-HT-7695 1 G2 0 442 1 0 Mutant wt Mutant wt wt Oligodendroglioma FEMALE 29 Gross Total Resection NA YES YES R4 mi1 C3 O1
70
+ TCGA-HT-7854 0 G2 1 1147 0 0 NA wt wt wt wt Astrocytoma MALE 62 Gross Total Resection 100 NO NA R2 mi1 C1 O3
71
+ TCGA-HT-7855 0 G3 0 585 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 39 Gross Total Resection NA YES YES R1 mi2 C1 O2
72
+ TCGA-HT-7856 1 G3 0 1189 1 0 NA wt wt wt wt Oligodendroglioma MALE 35 Gross Total Resection NA YES YES R4 mi1 C3 O1
73
+ TCGA-HT-7857 0 G3 0 7 0 0 NA wt wt wt wt Astrocytoma FEMALE 24 Gross Total Resection NA NA NA R2 mi3 C1 O2
74
+ TCGA-HT-7858 0 G2 0 1540 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 28 Gross Total Resection NA YES NA R1 mi1 C1 O2
75
+ TCGA-HT-7860 0 G3 NA NA 0 0 NA wt wt wt wt Astrocytoma FEMALE 60 Gross Total Resection NA NA NA R2 mi1 C2 O3
76
+ TCGA-HT-7873 0 G2 0 718 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 29 Gross Total Resection NA YES NA R1 mi2 C1 O2
77
+ TCGA-HT-7874 1 G3 0 1130 1 0 NA wt wt wt wt Oligodendroglioma FEMALE 41 Gross Total Resection NA YES YES R4 mi1 C3 O1
78
+ TCGA-HT-7875 1 G2 0 10 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 56 Gross Total Resection 80 NA NA R4 mi2 C3 O1
79
+ TCGA-HT-7877 1 G2 0 4 1 0 NA wt Mutant wt wt Oligodendroglioma FEMALE 20 Gross Total Resection NA NA NA R4 mi2 C3 O1
80
+ TCGA-HT-7879 0 G3 0 112 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 31 Gross Total Resection NA YES YES R1 mi2 C1 O2
81
+ TCGA-HT-7880 0 G2 0 162 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 30 Gross Total Resection NA NA NA R4 mi1 C1 O2
82
+ TCGA-HT-7881 1 G2 0 89 1 0 NA wt wt wt wt Oligodendroglioma MALE 38 Gross Total Resection NA YES NA R4 mi1 C3 O1
83
+ TCGA-HT-7882 0 G3 1 113 0 1 NA wt wt wt wt Oligodendroglioma MALE 66 Gross Total Resection NA NA NA R2 mi3 C1 O1
84
+ TCGA-HT-7884 0 G2 0 343 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 44 Gross Total Resection 80 YES YES NA mi2 C1 O2
85
+ TCGA-HT-7902 0 G2 0 956 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 30 Gross Total Resection NA NA NA NA mi1 C1 O2
86
+ TCGA-HT-8010 1 G2 0 50 1 0 NA wt wt wt wt Oligodendroglioma FEMALE 64 Gross Total Resection NA NA NA R4 mi1 C3 O1
87
+ TCGA-HT-8011 0 G3 1 278 0 0 NA wt wt wt wt Astrocytoma MALE 55 Gross Total Resection NA YES YES R2 mi4 C2 O3
88
+ TCGA-HT-8012 1 G2 1 133 1 0 NA wt Mutant wt wt Oligodendroglioma FEMALE 30 Gross Total Resection NA NA NA R3 mi2 C3 O1
89
+ TCGA-HT-8013 0 G2 1 1306 2 1 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 37 Gross Total Resection NA NO NO R1 mi1 C1 O2
90
+ TCGA-HT-8015 0 G2 NA NA 0 0 NA wt wt wt wt Astrocytoma MALE 21 Gross Total Resection NA NA NA R4 mi1 C1 O3
91
+ TCGA-HT-8018 0 G2 NA NA 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 40 Gross Total Resection NA NA NA R4 mi1 C1 O2
92
+ TCGA-HT-8019 0 G3 NA NA 0 0 NA wt wt wt wt Oligodendroglioma FEMALE 34 Gross Total Resection NA NA NA R4 mi1 C1 O1
93
+ TCGA-HT-8104 0 G3 NA NA 0 0 NA wt wt Mutant wt Astrocytoma FEMALE 51 Gross Total Resection NA YES NA R2 mi1 C2 O3
94
+ TCGA-HT-8105 1 G3 0 190 1 0 NA wt Mutant wt Mutant Oligodendroglioma MALE 54 Gross Total Resection NA YES NA R3 mi2 C3 O1
95
+ TCGA-HT-8106 0 G3 0 3 2 0 NA wt wt wt Mutant Astrocytoma MALE 53 Gross Total Resection NA NA NA R2 mi3 C1 O2
96
+ TCGA-HT-8107 0 G2 0 14 0 0 NA wt wt Mutant wt Oligodendroglioma MALE 62 Gross Total Resection NA NA NA R4 mi1 C1 O3
97
+ TCGA-HT-8108 0 G2 0 76 2 0 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 26 Gross Total Resection NA NA NA R1 mi2 C1 O2
98
+ TCGA-HT-8109 1 G3 NA NA 1 0 NA wt wt wt wt Oligodendroglioma MALE 64 Gross Total Resection NA NA NA R4 mi1 C3 O1
99
+ TCGA-HT-8110 0 G3 NA NA 0 0 NA wt wt Mutant wt Astrocytoma MALE 57 Gross Total Resection NA YES NA R2 mi1 C2 O3
100
+ TCGA-HT-8111 0 G3 NA NA 2 0 NA wt wt wt Mutant Oligoastrocytoma MALE 32 Gross Total Resection NA NA NA R1 mi1 C1 O2
101
+ TCGA-HT-8113 0 G2 0 900 2 0 NA wt wt wt wt Oligodendroglioma FEMALE 49 Gross Total Resection NA YES YES R4 mi1 C1 O1
102
+ TCGA-HT-8114 0 G3 0 118 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 36 Gross Total Resection NA YES YES R1 mi2 C1 O2
103
+ TCGA-HT-8558 NA G2 0 52 0 0 NA wt wt wt wt Oligodendroglioma FEMALE 29 Gross Total Resection NA NO NO R4 mi1 NA NA
104
+ TCGA-HT-8563 0 G3 0 488 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 30 Gross Total Resection NA YES NO R2 mi3 C1 O2
105
+ TCGA-HT-8564 0 G3 1 120 0 0 NA wt wt wt Mutant Astrocytoma MALE 47 Gross Total Resection 80 YES YES R4 mi4 C1 O2
106
+ TCGA-HT-A4DS 0 G3 NA NA 0 0 NA wt wt wt Mutant Astrocytoma FEMALE 55 Gross Total Resection NA NA NA R2 mi2 C1 O2
107
+ TCGA-HT-A4DV 1 G3 0 319 1 0 NA wt wt wt wt Oligodendroglioma FEMALE 51 Gross Total Resection 90 YES YES R4 mi2 C3 O1
108
+ TCGA-HT-A5R5 0 G2 NA NA 2 0 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 33 Gross Total Resection NA NA NA R4 mi2 C1 O2
109
+ TCGA-HT-A5R7 0 NA NA NA 2 NA NA Mutant wt wt wt NA NA NA Gross Total Resection NA NA NA R4 mi2 C1 O2
110
+ TCGA-HT-A5RA 0 G3 0 447 0 0 NA wt wt wt wt Astrocytoma FEMALE 65 Gross Total Resection 70 YES YES R2 mi2 C2 O3
111
+ TCGA-HT-A5RB 0 G2 NA NA 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 24 Gross Total Resection NA NA NA R4 mi2 C1 O2
112
+ TCGA-HT-A5RC 0 G3 1 162 0 1 NA wt wt Mutant wt Astrocytoma FEMALE 70 Gross Total Resection 40 YES YES R2 mi2 C2 O3
113
+ TCGA-HT-A61A 0 G2 NA NA NA 0 NA wt wt wt wt Oligodendroglioma FEMALE 20 Subtotal Resection 80 YES YES NA mi2 C1 O2
114
+ TCGA-HT-A61B 1 G2 0 396 2 NA NA wt wt wt Mutant NA NA NA Gross Total Resection NA NA NA NA mi2 C3 O1
115
+ TCGA-HT-A61C 0 G2 NA NA 0 NA NA wt wt Mutant wt NA NA NA Gross Total Resection NA NA NA NA mi2 C1 O2
116
+ TCGA-HT-A614 0 G2 1 395 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 47 Gross Total Resection NA NA NA NA mi2 C2 O3
117
+ TCGA-HT-A615 0 G3 0 162 1 0 NA wt wt wt wt Oligodendroglioma FEMALE 38 Gross Total Resection 90 YES NO NA mi2 C1 O2
118
+ TCGA-HT-A616 1 NA NA NA 2 0 NA Mutant wt wt wt Astrocytoma FEMALE 36 Gross Total Resection NA NO NO NA mi2 C3 O1
119
+ TCGA-HT-A617 NA G2 NA NA 0 0 NA wt wt wt wt Oligodendroglioma MALE 47 Gross Total Resection 60 NA NA NA mi2 NA NA
120
+ TCGA-HT-A618 0 NA NA NA 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 37 Gross Total Resection 80 YES YES NA mi2 C1 O2
121
+ TCGA-HT-A619 0 NA NA NA 1 NA NA wt Mutant wt wt NA NA NA Gross Total Resection NA NA NA NA mi4 C2 O3
122
+ TCGA-DU-8158 0 G3 NA NA 0 1 NA wt wt Mutant wt Astrocytoma FEMALE 57 Subtotal Resection 80 YES YES R2 mi4 C2 O3
123
+ TCGA-DU-8162 0 G3 1 402 0 1 NA wt wt Mutant wt Oligoastrocytoma FEMALE 61 Subtotal Resection 80 YES YES R4 mi1 C1 O3
124
+ TCGA-DU-8163 0 G3 1 509 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 29 Subtotal Resection 90 YES YES R1 mi1 C1 O2
125
+ TCGA-DU-8164 1 G2 0 651 1 0 NA wt wt wt wt Oligodendroglioma MALE 51 Subtotal Resection NA NA YES R4 mi2 C3 O3
126
+ TCGA-DU-8165 0 G3 0 582 0 0 NA wt wt wt Mutant Oligodendroglioma FEMALE 60 Subtotal Resection 90 YES YES R2 mi3 C2 O2
127
+ TCGA-DU-8166 0 G2 1 193 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 29 Subtotal Resection NA YES YES R4 mi2 C1 O2
128
+ TCGA-DU-8167 0 G2 0 471 2 0 NA wt wt wt Mutant Oligoastrocytoma FEMALE 69 Subtotal Resection 100 YES NA R1 mi1 C1 O2
129
+ TCGA-DU-8168 1 G3 0 431 1 0 NA wt Mutant wt wt Oligodendroglioma FEMALE 55 Gross Total Resection 70 YES YES R3 mi2 C3 O1
130
+ TCGA-DU-A5TP 0 G3 1 203 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 33 Subtotal Resection 70 YES YES R2 mi2 C3 O2
131
+ TCGA-DU-A5TR 0 G2 0 383 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 51 Biopsy Only 90 YES YES R2 mi2 C1 O2
132
+ TCGA-DU-A5TS 0 G2 0 460 2 0 NA Mutant wt wt wt Oligodendroglioma MALE 42 Subtotal Resection 100 YES YES NA mi2 C1 O2
133
+ TCGA-DU-A5TT 0 G3 0 151 0 0 NA wt wt Mutant wt Oligodendroglioma MALE 70 Subtotal Resection NA YES YES R2 mi2 C2 O2
134
+ TCGA-DU-A5TU 0 G2 NA NA 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 62 Subtotal Resection 50 NO NO R2 mi4 C1 O2
135
+ TCGA-DU-A5TW 0 G3 0 172 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 33 Subtotal Resection 100 NO NO R1 mi2 C1 O2
136
+ TCGA-DU-A5TY 0 G3 0 371 0 0 NA wt wt Mutant wt Astrocytoma FEMALE 46 Subtotal Resection NA YES YES R2 mi4 C2 O3
137
+ TCGA-EZ-7264 1 G2 0 436 1 0 Mutant wt Mutant wt wt Oligodendroglioma FEMALE 47 Gross Total Resection NA NA NA R3 mi2 C3 O1
138
+ TCGA-FG-5962 1 G3 0 1453 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 54 Subtotal Resection NA YES YES R4 mi1 C3 NA
139
+ TCGA-FG-5963 0 G3 1 497 0 1 NA Mutant wt wt Mutant Astrocytoma MALE 23 Subtotal Resection 90 YES NO R2 mi1 C1 NA
140
+ TCGA-FG-5964 1 G2 0 1045 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 62 Subtotal Resection NA NO NA NA mi2 C3 NA
141
+ TCGA-FG-5965 0 G2 1 1035 2 1 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 39 Subtotal Resection 90 YES NO NA mi1 C3 NA
142
+ TCGA-FG-6688 0 G3 1 405 0 0 NA wt wt wt wt Astrocytoma FEMALE 59 Subtotal Resection 80 YES NO R2 mi1 C2 O3
143
+ TCGA-FG-6689 0 G2 1 218 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 30 Subtotal Resection 70 NO NO R4 mi2 C1 O2
144
+ TCGA-FG-6690 0 G2 0 760 2 0 NA Mutant wt wt Mutant Oligodendroglioma MALE 70 Subtotal Resection 90 YES NA R1 mi2 C3 O2
145
+ TCGA-FG-6691 0 G2 0 729 2 0 NA Mutant wt wt Mutant Astrocytoma FEMALE 23 Gross Total Resection 100 NA NA R1 mi2 C1 O2
146
+ TCGA-FG-6692 0 G3 1 561 0 1 NA wt wt Mutant wt Oligodendroglioma MALE 63 Subtotal Resection NA YES NO R2 mi4 C2 O3
147
+ TCGA-FG-7634 1 G2 0 467 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 28 Subtotal Resection NA NA NA NA mi2 C2 O1
148
+ TCGA-FG-7637 NA G2 0 1219 NA 0 NA wt wt wt wt Oligoastrocytoma MALE 49 Subtotal Resection NA NA NA R3 mi2 NA NA
149
+ TCGA-FG-7641 1 G2 0 627 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 31 Gross Total Resection NA NA NA R4 mi2 C3 O1
150
+ TCGA-FG-7643 0 G2 1 254 0 0 NA wt wt wt wt Oligoastrocytoma FEMALE 49 Subtotal Resection NA NA NA R4 mi1 C2 O1
151
+ TCGA-FG-8186 1 G3 0 487 1 0 NA wt wt wt wt Oligoastrocytoma FEMALE 42 Subtotal Resection 70 YES NA NA mi2 C3 O1
152
+ TCGA-FG-8189 0 G2 0 361 2 0 NA wt wt wt wt Oligodendroglioma FEMALE 33 Subtotal Resection 70 YES YES R4 mi2 C1 O1
153
+ TCGA-FG-A4MT 0 G2 1 499 2 0 NA Mutant wt wt Mutant Oligodendroglioma FEMALE 27 Gross Total Resection 100 NO NO R1 mi2 C1 O2
154
+ TCGA-FG-A4MU 0 G3 NA NA 0 0 NA wt wt Mutant wt Oligoastrocytoma MALE 58 Subtotal Resection NA NA NA R2 mi3 C2 O3
155
+ TCGA-FG-A60K 1 G2 0 202 1 0 NA wt Mutant wt wt Oligoastrocytoma FEMALE 34 Gross Total Resection NA NO NO NA mi2 C3 O1
156
+ TCGA-HT-7467 1 G2 NA NA 1 0 NA wt wt wt wt Oligodendroglioma MALE 54 Gross Total Resection NA NA NA R4 mi1 C3 O1
157
+ TCGA-HT-7468 1 G3 0 203 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 30 Gross Total Resection 80 NA YES R3 mi2 C3 O1
158
+ TCGA-HT-7469 0 G3 1 92 0 1 NA Mutant wt wt Mutant Oligodendroglioma MALE 30 Gross Total Resection NA YES YES R2 mi2 C1 O2
159
+ TCGA-HT-7470 0 G3 0 268 2 0 NA Mutant wt wt Mutant Oligodendroglioma MALE 37 Gross Total Resection 90 YES YES R4 mi1 C1 O2
160
+ TCGA-HT-7471 1 G3 NA NA 1 0 NA wt Mutant wt wt Oligodendroglioma FEMALE 37 Gross Total Resection NA NA NA R3 mi2 C3 O1
161
+ TCGA-HT-7472 0 G2 0 1 2 0 NA Mutant wt wt Mutant Oligodendroglioma MALE 38 Gross Total Resection NA NA NA R1 mi2 C3 O2
162
+ TCGA-HT-7473 0 G2 0 503 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 28 Gross Total Resection NA NO NA R1 mi2 C1 O2
163
+ TCGA-HT-7474 0 G2 0 114 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 52 Gross Total Resection NA NA NA R4 mi1 C1 O2
164
+ TCGA-HT-7475 0 G3 0 530 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 67 Gross Total Resection 70 YES NO R4 mi2 C1 O2
165
+ TCGA-HT-7476 0 G2 0 199 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 26 Gross Total Resection NA YES YES R4 mi1 C1 O2
166
+ TCGA-HT-7477 0 G3 0 738 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 62 Gross Total Resection 80 YES YES R2 mi2 C2 O2
167
+ TCGA-HT-7478 0 G2 0 194 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 36 Gross Total Resection NA YES NA R2 mi4 C1 O2
168
+ TCGA-HT-7479 0 G3 0 216 2 0 NA wt wt wt Mutant Astrocytoma MALE 44 Gross Total Resection NA NA YES R1 mi1 C1 O2
169
+ TCGA-HT-7480 1 G2 1 1943 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 33 Gross Total Resection NA YES YES R4 mi2 C3 O1
170
+ TCGA-HT-7481 1 G2 1 880 1 0 NA wt wt wt wt Oligodendroglioma MALE 39 Gross Total Resection NA NO NA R4 mi2 C3 O1
171
+ TCGA-HT-7482 0 G2 1 374 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 18 Gross Total Resection NA NO NO R1 mi1 C1 O2
172
+ TCGA-HT-7483 0 G2 NA NA 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 14 Gross Total Resection NA NA NA R4 mi2 C1 O2
173
+ TCGA-HT-7485 0 G2 0 122 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 42 Gross Total Resection 100 NA NA R1 mi1 C1 O2
174
+ TCGA-HT-7601 0 G3 0 153 2 0 NA wt wt wt Mutant Astrocytoma FEMALE 30 Gross Total Resection NA YES YES R2 mi3 C3 O2
175
+ TCGA-HT-7602 0 G2 NA NA 2 0 wt wt wt wt Mutant Oligodendroglioma MALE 21 Gross Total Resection NA NA NA R1 mi1 C1 O2
176
+ TCGA-HT-7603 0 G2 0 705 2 0 NA Mutant wt wt Mutant Oligodendroglioma MALE 29 Gross Total Resection NA YES NA R4 mi1 C1 O2
177
+ TCGA-HT-7604 0 G2 NA NA 2 0 NA Mutant wt wt Mutant Astrocytoma MALE 50 Gross Total Resection NA NA NA R1 mi2 C1 O2
178
+ TCGA-HT-7605 1 G2 0 139 1 0 NA wt wt wt wt Oligodendroglioma MALE 38 Gross Total Resection NA NA NA R4 mi1 C3 O1
179
+ TCGA-HT-7606 0 G2 0 526 2 0 NA wt wt wt Mutant Astrocytoma FEMALE 30 Gross Total Resection NA YES YES R1 mi2 C1 O2
180
+ TCGA-HT-7607 1 G2 1 96 1 1 NA wt wt wt wt Astrocytoma FEMALE 61 Gross Total Resection 70 NA NA R4 mi1 C3 O1
181
+ TCGA-HT-7608 1 G2 0 671 1 0 NA wt wt wt wt Oligoastrocytoma MALE 61 Gross Total Resection NA NA NA NA mi2 C3 O1
182
+ TCGA-HT-7609 0 G3 0 1399 2 0 NA wt wt wt Mutant Oligoastrocytoma MALE 34 Gross Total Resection 90 YES YES R1 mi2 C1 O2
183
+ TCGA-HT-7610 0 G2 1 1205 2 0 NA Mutant wt wt Mutant Oligoastrocytoma FEMALE 25 Gross Total Resection NA NA NA R4 mi2 C3 O2
184
+ TCGA-HT-7611 0 G2 0 1752 2 0 NA Mutant wt wt Mutant Oligoastrocytoma MALE 36 Gross Total Resection 90 NO NO R1 mi2 C1 O2
185
+ TCGA-HT-7616 1 G3 1 7 1 1 NA wt Mutant wt wt Oligodendroglioma MALE 75 Gross Total Resection NA NA NA R3 mi2 C3 O1
186
+ TCGA-HT-7620 1 G3 1 280 1 0 NA wt wt wt wt Oligodendroglioma MALE 40 Gross Total Resection NA YES NO NA mi1 C3 O1
187
+ TCGA-HT-7676 0 G2 NA NA 2 0 NA Mutant wt wt Mutant Oligodendroglioma MALE 26 Gross Total Resection NA NA NA R1 mi2 C1 O2
188
+ TCGA-HT-7677 1 G3 0 494 1 0 NA wt Mutant wt wt Oligodendroglioma MALE 53 Gross Total Resection NA YES YES R3 mi2 C3 O1
189
+ TCGA-HT-7680 0 G2 0 23 0 0 NA wt wt wt wt Astrocytoma FEMALE 32 Gross Total Resection NA NA NA R2 mi1 C1 O2
samples/ROI-Masks.txt ADDED
@@ -0,0 +1,211 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ From : https://academic.oup.com/neuro-oncology/article/19/6/862/2948265
2
+
3
+ MRI features predict survival and molecular markers in diffuse lower-grade gliomas
4
+ Hao Zhou, Martin Vallières, Harrison X. Bai, Chang Su, Haiyun Tang, Derek Oldridge, Zishu Zhang, Bo Xiao, Weihua Liao, Yongguang Tao ... Show moreAuthor Notes
5
+ Neuro-Oncology, Volume 19, Issue 6, 1 June 2017, Pages 862–870, https://doi.org/10.1093/neuonc/now256
6
+ Published: 24 January 2017
7
+ A correction has been published: Neuro-Oncology, Volume 19, Issue 12, December 2017, Page 1701, https://doi.org/10.1093/neuonc/nox110
8
+ pdfPDF
9
+ Split View
10
+ Cite
11
+ Permissions Icon Permissions
12
+ Share Icon Share
13
+ Abstract
14
+ Background.
15
+ Previous studies have shown that MR imaging features can be used to predict survival and molecular profile of glioblastoma. However, no study of a similar type has been performed on lower-grade gliomas (LGGs).
16
+
17
+ Methods.
18
+ Presurgical MRIs of 165 patients with diffuse low- and intermediate-grade gliomas (histological grades II and III) were scored according to the Visually Accessible Rembrandt Images (VASARI) annotations. Radiomic models using automated texture analysis and VASARI features were built to predict isocitrate dehydrogenase 1 (IDH1) mutation, 1p/19q codeletion status, histological grade, and tumor progression.
19
+
20
+ Results.
21
+ Interrater analysis showed significant agreement in all imaging features scored (k = 0.703–1.000). On multivariate Cox regression analysis, no enhancement and a smooth non-enhancing margin were associated with longer progression-free survival (PFS), while a smooth non-enhancing margin was associated with longer overall survival (OS) after taking into account age, grade, tumor location, histology, extent of resection, and IDH1 1p/19q subtype. Using logistic regression and bootstrap testing evaluations, texture models were found to possess higher prediction potential for IDH1 mutation, 1p/19q codeletion status, histological grade, and progression of LGGs than VASARI features, with areas under the receiver-operating characteristic curves of 0.86 ± 0.01, 0.96 ± 0.01, 0.86 ± 0.01, and 0.80 ± 0.01, respectively.
22
+
23
+ Conclusion.
24
+ No enhancement and a smooth non-enhancing margin on MRI were predictive of longer PFS, while a smooth non-enhancing margin was a significant predictor of longer OS in LGGs. Textural analyses of MR imaging data predicted IDH1 mutation, 1p/19q codeletion, histological grade, and tumor progression with high accuracy.
25
+
26
+ 1p/19q codeletion, IDH1 mutation, lower-grade gliomas, MR imaging, texture analysis
27
+ Topic: magnetic resonance imagingmutationglioblastomagliomadiagnostic imagingneoplasmscox proportional hazards modelstumor progressionidh1 geneprogression-free survivalradiomics
28
+ Issue Section: CLINICAL INVESTIGATIONS
29
+ Importance of the study
30
+ Previous studies have shown that MRI features scored according to VASARI annotations can be used to predict survival and molecular profile of glioblastoma. However, no study of similar type has been performed on LGGs. On multivariate analysis, we showed that no enhancement on MRI was associated with longer PFS, while a smooth non-enhancing margin was associated with longer PFS and OS compared with an irregular non-enhancing margin. Our results demonstrated that imaging features scored using a standardized vocabulary had prognostic value in addition to traditionally recognized clinical and molecular markers in LGGs. In addition, multivariable texture models extracted from baseline MRI scans have the potential to accurately classify LGGs in terms of IDH1 mutation, 1p/19q codeletion, histological grade, and tumor progression. With proven accuracy, textural analysis will complement invasive tissue sampling, guiding patient management at an earlier stage of disease and in follow-up. Textural analysis may even serve as the standard in cases where invasive procedure is not available or appropriate.
31
+
32
+ Diffuse lower-grade gliomas (LGGs) are infiltrative neoplasms that generally include diffuse low- and intermediate-grade gliomas (World Health Organization [WHO] grade II or III).1 The outcomes of these tumors are variable—some recur after treatment within months and even progress to glioblastoma (WHO grade IV gliomas), while others remain indolent for years.2 Traditionally, the natural progression of LGGs is thought to be dependent on their histological class (astrocytomas vs oligoastrocytomas vs oligodendrogliomas). However, a recent study from The Cancer Genome Atlas (TCGA) Research Network classified LGGs into 3 categories based on isocitrate dehydrogenase (IDH) mutation and 1p/19q codeletion status: gliomas with an IDH mutation and 1p/19q codeletion, gliomas with an IDH mutation but no 1p/19q codeletion, and gliomas with wild-type IDH.1 This new classification scheme has been shown to capture the biologic characteristics of LGGs with greater fidelity than does histological class.1
33
+
34
+ MRI has served as an important noninvasive method for diagnosing gliomas and monitoring their treatment response. Previous radiogenomic analyses of glioblastoma have shown that the proportion of contrast enhancement (CE) and longest axis length of tumor on MR were significantly associated with poor survival.3 In a recent study of 120 patients with primary grades III (n = 35) and IV (n = 85) gliomas, Zhang et al. built a model using nonredundant preoperative MRIs and clinical data with a random forest algorithm that achieved accuracies of 86% in the training cohort and 89% in the validation cohort in predicting IDH genotype.4 However, no similar study has been performed to determine the association of MR imaging features with survival and molecular markers in LGGs.
35
+
36
+ To make the assessment of MR imaging features in gliomas more accurate and reproducible, a comprehensive feature set known as the Visually Accessible Rembrandt Images (VASARI) was developed in 2008. The VASARI annotations include 30 distinct imaging features with corresponding criteria clustered by categories related to lesion location, morphology of the lesion substance, morphology of the lesion margin, alterations in the vicinity of the lesion, and remote alterations (https://wiki.nci.nih.gov/display/CIP/VASARI).5 Previous studies have shown that measurements of these features by VASARI were highly reproducible, clinically meaningful, and biologically relevant in glioblastoma.3,6
37
+
38
+ Radiomics extracts and mines a large number of medical imaging features.7 These approaches based on machine learning have been increasingly used to quantify tumor phenotypic characteristics and to predict clinical outcomes.7 In 2014, Aerts et al.8 demonstrated that imaging features quantifying tumor image intensity, shape, and texture extracted from computed tomography data of 1019 patients with lung or head-and-neck cancer not only had prognostic power in the independent datasets of lung and head-and-neck cancer patients, but also had associations with gene-expression patterns. In 2015, Vallières et al.9 built a radiomics model from joint 2-fluoro-2-deoxy-D-glucose PET and MRI texture features using bootstrapping evaluations that predicted lung metastases in soft tissue sarcomas of the extremities with high sensitivity and specificity.
39
+
40
+ The purpose of our study was to perform a comprehensive analysis of MR imaging features of LGGs by using the VASARI feature set, as they relate to patient survival and molecular markers. In addition, we explored the possibility of using textural features extracted from MR imaging to make binary predictions of wild-type IDH versus IDH1 mutation; IDH1 mutation with 1p/19q codeletion versus IDH1 mutation without 1p/19q codeletion; grade II versus grade III LGGs; and progression versus nonprogression of LGGs.
41
+
42
+ Materials and Methods
43
+ Study Data
44
+ We identified 165 patients with diffuse low- and intermediate-grade gliomas (histological grades II and III) from TCGA who have overlapping presurgical MRI data from The Cancer Imaging Archive (TCIA),10,11 an imaging sharing resource that houses images corresponding to TCGA patients. A flowchart of the number of patients included for each analysis, along with the number of and reason why patients were excluded from each analysis, is shown in Fig. 1. As the patients had been previously de-identified by TCGA/TCIA, and their relevant information was available for public download, no institutional review board or Health Insurance Portability and Accountability Act approval was required for our study.
45
+
46
+ Fig. 1
47
+ Flowchart of patients included and excluded for each analysis.
48
+ Open in new tabDownload slide
49
+ Flowchart of patients included and excluded for each analysis.
50
+
51
+ Imaging Review: VASARI Scores and Computation of Textures
52
+ For each patient, 2 neuroradiologists (H.T. and W.L.) with 7 and 20 years of experience, respectively, independently reviewed axial T1-weighted MR images before (T1W) and after gadolinium-based CE material administration (T1CE) as well as axial T2-weighted (T2W) and axial T2-weighted fluid attenuated inversion recovery (FLAIR) (T2F) images of the 165 LGG patients. The readers were blinded to the clinical data. Clear Canvas workstation, which allows visualization as well as annotation and markup of Digital Imaging and Communications in Medicine (DICOM) images, was used for imaging review. Each tumor was independently scored by the readers using the 30 imaging features defined according to the VASARI scoring system as previously described. The reader is referred to the Supplementary material for a complete description of the VASARI feature set. The interreader agreement for the imaging features was assessed using the kappa consistency test. Kappa values >0.81, in the range of 0.61–0.80, and <0.60 were considered to reflect excellent, good, and poor agreement, respectively. Final disagreement was resolved in a panel format including 2 additional coauthors (H.X.B. and L.Y.). For texture analysis, 3D regions of interest (ROIs) for each MR imaging set of each patient were manually drawn slice-by-slice in the axial plane for each of the available sequences (T1W, T2W, T1CE, T2F) by an expert radiologist. A total of 42 texture features were then extracted using 3D analysis from the T1W, T2W, T1CE, and T2F scans: 3 histogram-based textures, 8 texture features from the Gray-Level Co-occurrence Matrix (GLCM), 13 texture features from the Gray-Level Run-Length Matrix (GLRLM), 13 texture features from the Gray-Level Size Zone Matrix (GLSZM), and 5 texture features from the Neighborhood Gray-Tone Difference Matrix (NGTDM). The reader is referred to the Supplementary material for a complete description of all texture features, acronyms, and references.
53
+
54
+ Association of VASARI Imaging Features with Survival
55
+ Progression-free survival (PFS) was defined as the time that passes from the day on which a patient is enrolled and the date on which tumor progresses. Overall survival (OS) was defined as the time between initial diagnosis and death or last follow-up. We examined the association between each VASARI imaging feature with PFS and OS using the Kaplan–Meier survival curves and log-rank analyses. Features that were significant on the univariate analysis (P < .05) were entered into multivariate survival analysis based on the Cox proportional hazards ratio model, after incorporating clinical and pathological variables, including age, gender, extent of resection, histological type, histological grade, pretreatment Karnofsky performance scale (KPS), and IDH1 1p/19q codeletion status.
56
+
57
+ Prediction of IDH1 Mutation, 1p/19q Codeletion Status, Histological Grade, and LGG Progression with Textural Imaging Features
58
+ For this part of the work, the patient imaging data and outcome availability for each modeled outcome were as follows: (i) IDH1 mutation: 63 mutations, 21 wild type; (ii) 1p/19q codeletion status: 17 codeletion, 50 non-codeletion; (iii) histological grade: 35 grade II gliomas, 49 grade III gliomas; and (iv) LGG progression: 28 progressions, 47 nonprogressions. Binary clinical outcomes were modeled by predicting the class with the lowest number of instances.
59
+
60
+ Forty-two texture features were computed as described by Vallières et al.9 using the 40 possible combinations of the following extraction parameters: (i) 3D isotropic scales of 0.5 mm, 1 mm, 2 mm, 3 mm, and 4 mm; (ii) “uniform” and “equal-probability” quantization; (iii) number of gray-levels of 8, 16, 32, and 64. The 4 initial feature sets that were tested comprised one T1-based and one T2-based MR imaging sequence (each containing 2 × 42 × 40 = 3360 scan-texture-parameters): (i) T1W-T2W, (ii) T1W-T2F, (iii) T1CE-T2W, and (iv) T1CE-T2F.
61
+
62
+ Multivariate models were constructed for each initial feature set and modeled outcome using imbalanced-adjusted logistic regression, an adaptation of the method based on Schiller et al9 following the general methodology developed by Vallières et al.12 All initial feature sets first underwent feature set reduction using 100 bootstrap training samples to yield reduced feature sets of 25 different scan-texture features. Then, feature selection was performed by maximizing the area under the receiver-operating characteristic curve (AUC)632+ metric in 100 bootstrap training and testing samples to obtain texture models combining 1 to 10 variables (model order).13 For each outcome, the feature set and model order providing the combination of texture variables with the best parsimonious properties (ie, the simplest model with the best predictive properties) were chosen. The model was built using the feature set that provided the highest prediction curve while using the lowest model order before the AUC632+ metric started reaching a plateau or decreasing. Finally, the prediction performance of the 4 chosen texture models was estimated using average AUCs, sensitivities, and specificities obtained in 100 bootstrap testing samples.
63
+
64
+ Prediction of IDH1 Mutation, 1p/19q Codeletion Status, Histological Grade, and LGG Progression with VASARI Features
65
+ In order to directly compare the prognostic potential of VASARI features and texture features, we built VASARI multivariable models to predict IDH1 mutation, 1p/19q codeletion status, histological grade, and LGG progression. From the full set of VASARI features, the feature selection, model choice, and prediction estimation methods were the same as described in the section above for texture analysis.
66
+
67
+ Comparison of the Predictive Potential of Models Using Clinical Variables, VASARI Features, and Texture Features with Random Forest Analysis
68
+ Logistic or Cox regression analysis is best suited for modeling continuous variables only (eg, texture and VASARI features). These types of analyses provide a fast way to mine the best features for that category of inputs. On the other hand, random forests are more complex classifiers and are designed to implement any type of inputs, either categorical, as is often the case for clinical information, or continuous for radiomics data. In order to directly compare their predictive potential, clinical variables (categorical data), VASARI features, and texture features (continuous data) were thus included in a random forest analysis to predict IDH1 mutation, 1p/19q codeletion status, histological grade, and LGG progression. The clinical variables included age, KPS, histological type, grade (removed for histological grade outcome), laterality, location, gender, radiotherapy (yes vs no; removed for all outcomes except LGG progression), chemotherapy (yes vs no; removed for all outcomes except LGG progression), and IDH1 1p/19q subtype (removed for IDH1, 1p/19q outcomes). The VASARI and texture features included in the random forest analysis were the variables forming the best multivariable models as determined using the methods described in the 2 previous sections above. Random forest classifiers were trained (inherently using bootstrapping) on the whole cohort using 500 trees. Prediction performance was estimated using out-of-bag observations. Prediction balance between sensitivity and specificity was achieved by finding the optimal cost (ie, emphasis) on the classification of positive instances, and by maximizing 0.5*AUC + 0.5*(1 − |Sensitivity − Specificity|) on out-of-bag estimates. Random forest analysis was performed using clinical variables only; VASARI features only; texture features only; a combination of VASARI and texture features; and a combination of clinical variables, VASARI, and texture features.
69
+
70
+ Results
71
+ Interreader Agreement
72
+ Interrater analysis showed significant agreement in all VASARI imaging features scored. Interreader agreements for all the imaging features were good to excellent (kappa value = 0.703–1.000) (Supplementary Table 1).
73
+
74
+ Correlation between VASARI Imaging Features and Progression-Free Survival
75
+ Kaplan–Meier analysis showed that 7 VASARI features were significantly associated with PFS (P < .1): enhancement quality, proportion of enhancing tumor, proportion of non-enhancing tumor, definition of the non-enhancing margin, diffusion, satellites, and lesion size. On multivariate Cox regression analysis, enhancement quality, definition of the non-enhancing margin, the presurgical KPS score, and IDH1 1p/19q subtype were significantly associated with PFS after taking into account gender, tumor location, and histology (Table 1). Specifically, tumors with no enhancement had longer PFS than tumors with either mild or marked enhancement (P = .048 by log-rank test; Supplementary Fig. 1A). Tumors with a smooth margin were associated with improved PFS compared with those with an irregular margin (P = .02 by log-rank test; Supplementary Fig. 1B).
76
+
77
+
78
+ Table 1Correlation between VASARI imaging features and PFS on multivariate Cox analysis
79
+ Variable Hazard Ratio P-value
80
+ Enhancement quality 1.485 (1.051, 2.099) .025
81
+ Definition of the non- enhancing margin 2.056 (1.168, 3.518) .012
82
+ KPS 0.969 (0.942, 0.996) .023
83
+ IDH1 1p/19q subtype 3.035 (1.937, 4.756) <.001
84
+ Note: Data are hazard ratio estimates, with 95% CIs in parentheses, for variables included in the Cox regression model involving imaging features plus clinical variables, for the analysis of the association between the imaging features and PFS after adjusted for standard clinical variables.
85
+
86
+ Open in new tab
87
+ Correlation between VASARI Imaging Features and Overall Survival
88
+ Kaplan–Meier analysis showed that 10 VASARI features were significantly associated with OS: eloquent brain involved, proportion of enhancing tumor, proportion of non-enhancing tumor, cysts, multifocal or multicentric, definition of the non-enhancing margin, proportion of edema, diffusion, enhancing tumor crosses midline, and satellites. On multivariate Cox regression analysis, definition of the non-enhancing margin, age, tumor grade, and IDH1 1p/19q subtype were significantly associated with OS after taking into account gender, tumor location, and histology (Table 2). Specifically, tumors with a smooth margin had longer OS than those with an irregular margin (P = .002 by log-rank test; Supplementary Fig. 1C).
89
+
90
+
91
+ Table 2Correlation between VASARI imaging features and OS on multivariate Cox analysis
92
+ Variable Hazard Ratio P-value
93
+ Definition of the non-enhancing margin 1.088 (1.047, 1.131) .017
94
+ Age 1.088 (1.047, 1.131) <.001
95
+ WHO grade 5.298 (2.027, 13.849) .001
96
+ IDH1 1p/19q subtype 2.655 (1.489, 4.732) .001
97
+ Note: Data are hazard ratio estimates, with 95% CIs in parentheses, for variables included in the Cox regression model involving imaging features plus clinical variables, for the analysis of the association between the imaging features and OS after adjusted for standard clinical variables.
98
+
99
+ Open in new tab
100
+ Prediction of IDH1 Mutation, 1p/19q Codeletion Status, Histological Grade, and LGG Progression with Textural Imaging Features
101
+ Following feature set reduction and feature selection using imbalanced-adjusted logistic regression and bootstrap resampling, texture models with orders 1 to 10 that maximized the AUC632+ metric were computed for each of the 4 feature sets to model the 4 outcomes (Fig. 2A–D). By inspecting the curves in Fig. 1, we determined that the combinations of 3 textures from the T1CE-T2W set, 3 textures from the T1CE-T2W set, 4 textures from the T1CE-T2W set, and 4 textures from the T1W-T2W set provided the best predictive properties for IDH1 mutation, 1p/19q codeletion status, histological grade, and LGG progression outcomes, respectively. For the IDH1 mutation outcome, the optimal set of features included global-skewness (T2W), GLRLM run-length variance (T2W), and GLRLM short run low Gray-level emphasis (T2W), which reached an AUC of 0.86 ± 0.01, a sensitivity of 0.75 ± 0.03, and a specificity of 0.78 ± 0.02. For 1p/19q codeletion status, the optimal set of features included GLRLM low Gray-level run emphasis (LGRE) (T1CE), GLSZM short zone low Gray-level emphasis (SZHGE) (T2W), and GLRLM long run high Gray-level emphasis (T2W), which reached an AUC of 0.96 ± 0.01, a sensitivity of 0.90 ± 0.02, and a specificity of 0.89 ± 0.02. For the histological grade outcome, the optimal set of features included GLCM-homogeneity (T1CE), GLSZM short-zone emphasis (SZE) (T2W), GLSZM-SZE (T1CE), and global-kurtosis (T1CE), which reached an AUC of 0.86 ± 0.01, a sensitivity of 0.74 ± 0.02, and a specificity of 0.79 ± 0.02. For the LGG progression outcome, the optimal set of features included GLRLM long run low Gray-level emphasis (T1W), GLRLM-LGRE (T2W), GLSZM-SZHGE (T1W),and GLSZM zone size variance (T2W), which reached an AUC of 0.80 ± 0.01, a sensitivity of 0.76 ± 0.03, and a specificity of 0.72 ± 0.03. The direction of correlation between each significant texture feature included in the final model and outcome is shown in Supplementary Table 2.
102
+
103
+ Fig. 2
104
+ Inspection of predictive properties of multivariable texture models constructed from 4 feature sets: (i) T1W-T2W, (ii) T1W-T2F, (iii) T2CE-T2W, and (iv) T1CE-T2F. Estimation of prediction performance is shown for combinations of 1 to 10 texture features (model orders) in terms of the AUC632+ metric for: (A) IDH1 mutation, (B) 1p/19q codeletion status, (C) histological grade, and (D) LGG progression.
105
+ Open in new tabDownload slide
106
+ Inspection of predictive properties of multivariable texture models constructed from 4 feature sets: (i) T1W-T2W, (ii) T1W-T2F, (iii) T2CE-T2W, and (iv) T1CE-T2F. Estimation of prediction performance is shown for combinations of 1 to 10 texture features (model orders) in terms of the AUC632+ metric for: (A) IDH1 mutation, (B) 1p/19q codeletion status, (C) histological grade, and (D) LGG progression.
107
+
108
+ To demonstrate the classification capability of the optimal models shown in Supplementary Table 2, final logistic regression coefficients were computed for all models using 100 bootstrap training samples. The posterior probability of observing a given outcome as a function of the logistic regression response of the models was calculated along with the associated 95% CIs of the model responses in the bootstrap samples (Fig. 3).
109
+
110
+ Fig. 3
111
+ Probability of observing a given outcome as a function of the response of optimal multivariable texture models developed in this work, for all patients of the cohort: (A) IDH1 mutation (nonIDH1), (B) 1p/19q codeletion status (IDHcodel), (C) histological grade (lowGrade), and (D) LGG progression (progression). It can be seen that the optimal texture models can significantly separate the patients of the 2 classes for each outcome, especially in the case of the 1p/19q codeletion status.
112
+ Open in new tabDownload slide
113
+ Probability of observing a given outcome as a function of the response of optimal multivariable texture models developed in this work, for all patients of the cohort: (A) IDH1 mutation (nonIDH1), (B) 1p/19q codeletion status (IDHcodel), (C) histological grade (lowGrade), and (D) LGG progression (progression). It can be seen that the optimal texture models can significantly separate the patients of the 2 classes for each outcome, especially in the case of the 1p/19q codeletion status.
114
+
115
+ Prediction of IDH1 Mutation, 1p/19q Codeletion Status, Histological Grade, and LGG Progression with VASARI Features
116
+ For the IDH1 mutation outcome, the optimal set of features included proportion necrosis and lesion size, which reached an AUC of 0.73 ± 0.02, a sensitivity of 0.69 + 0.03, and a specificity of 0.69 ± 0.02. For the 1p/19q codeletion outcome, the optimal set of features included multifocal or multicentric, edema proportion, tumor location, and enhancing proportion, which reached an AUC of 0.78 ± 0.01, a sensitivity of 0.72 ± 0.03, and a specificity of 0.67 ± 0.03. For the histological grade outcome, the optimal set of features included enhancing proportion, definition of the non-enhancing margin, and diffusion, which reached an AUC of 0.78 ± 0.01, a sensitivity of 0.72 ± 0.03, and a specificity of 0.67 ± 0.03. For the LGG progression outcome, the optimal set of features included tumor location, enhancement quality, necrosis proportion, T1/FLAIR ratio, and thickness of enhancing margin, which reached an AUC of 0.58 ± 0.02, a sensitivity of 0.54 ± 0.04, and a specificity of 0.58 ± 0.03. The results are shown in Fig. 4. The direction of correlation between each significant VASARI feature included in the final model and outcome is shown in Supplementary Table 3.
117
+
118
+ Fig. 4
119
+ Inspection of predictive properties of VASARI models constructed for 4 different outcomes: (i) IDH1 mutation, (ii) 1p/19q codeletion status, (iii) histological grade, and (iv) LGG progression. Estimation of prediction performance is shown for combinations of 1 to 10 texture features (model orders) in terms of the AUC632+ metric.
120
+ Open in new tabDownload slide
121
+ Inspection of predictive properties of VASARI models constructed for 4 different outcomes: (i) IDH1 mutation, (ii) 1p/19q codeletion status, (iii) histological grade, and (iv) LGG progression. Estimation of prediction performance is shown for combinations of 1 to 10 texture features (model orders) in terms of the AUC632+ metric.
122
+
123
+ Comparison of the Predictive Potential of Models Using Clinical Variables, VASARI Features, and Texture Features with Random Forest Analysis
124
+ Random forest analysis was used to directly compare the predictive potential of clinical, VASARI, and texture features (Supplementary Table 4). For IDH1 mutation, 1p/19q codeletion, and LGG progression, we found that the best category of predictor was texture features, with AUCs of 0.79, 0.88, and 0.70, respectively. For histological grade, the best category of predictor was VASARI features, with an AUC of 0.73. However, the combination of clinical, VASARI, and texture variables showed that clinical variables can successfully complement imaging features for the prediction of IDH1 mutation and histological grade, with respective AUCs of 0.86 and 0.78. On the other hand, 1p/19q codeletion was best modeled with a combination of texture and VASARI features only (AUC = 0.89), and LGG progression was best modeled with texture features only (AUC = 0.70). The detailed results are shown in Supplementary Tables 5, 6, 7, 8, and 9.
125
+
126
+ Discussion
127
+ We found a significant association between enhancement quality (none vs mild vs marked) and PFS. Specifically, no enhancement was associated with longer PFS than either mild or marked enhancement. In addition, a smooth definition of the non-enhancing margin was associated with longer PFS and OS than an irregular non-enhancing margin. Of note, these imaging features were still strongly associated with patients’ survival after incorporating age, KPS, extent of resection, tumor grade, and IDH 1p/19q subtype.
128
+
129
+ In glioblastoma, previous studies have shown that the proportion of CE on MRI was associated with survival.3,14 However, in LGG, the prognostic significance of CE on survival remains less well understood, and differing results have been reported depending on LGG and mutation status.15–17 In our study, the proportion of CE was not a significant predictor of either PFS or OS, but the lack of CE was a positive predictor of PFS. There was no significant difference in survival between patients who had tumors with mild enhancement and those who had tumors with marked enhancement. Grade III tumors were more likely to enhance than grade II tumors (84% vs 44%, respectively), but a significant proportion of the grade II tumors demonstrated CE as well.
130
+
131
+ We found that a smooth non-enhancing margin was associated with longer PFS and OS compared with an irregular non-enhancing margin. This is in contrast to a prior study in glioblastoma which found that a smooth edge of CE predicted poor prognosis, while a sharp edge was a positive factor.6 However, a previous study of 43 grade III gliomas found no significant association of non-enhancing margin with survival.18 To our knowledge, our study was the first to demonstrate that the definition of the non-enhancing margin on MRI can predict survival in a combined cohort of grade II and grade III gliomas.
132
+
133
+ In this study, we constructed a multivariable texture model extracted from baseline MRI scans that achieved an AUC of 0.86, a sensitivity of 0.75, and a specificity of 0.78 in predicting IDH1 mutation in LGGs. This logistic regression model performed better than a model using VASARI features via the same methods and comparable to the models built with random forest analysis incorporating clinical variables, VASARI, and texture features. In comparison to previous studies that relied on complete clinical data19 or advanced imaging techniques,20 our results demonstrated the ability of machine-learning algorithms to achieve accurate prediction of IDH mutation in LGGs with very few preoperative MRI texture features alone. These results, if validated with other datasets, may affect clinical management in the future, since these imaging studies were obtained routinely before surgery. They can also be useful in a research setting where a large amount of samples have to be genotyped for IDH mutation.
134
+
135
+ The codeletion of chromosomal arms 1p and 19q is a characteristic and early genetic event in oligodendroglial tumors.15 Tumors with 1p/19q codeletion are associated with a better prognosis and enhanced response to therapy.16 Moreover, grades II and III gliomas with 1p/19q codeletion are also mutated in IDH1/2.21 In a smaller study of 55 patients with oligodendrogliomas, Brown et al.17 used S-transform-based texture analysis of preoperative MR images to predict 1p/19q codeletion with an AUC of 0.94, a sensitivity of 0.93, and a specificity of 0.96, versus a sensitivity of 0.70 and a specificity of 0.63 for genotype prediction by blinded experts. We corroborated these results in a larger cohort of patients that included both astrocytomas and oligodendrogliomas, and our optimal texture model achieved an average bootstrap testing AUC of 0.96, a sensitivity of 0.90, and a specificity of 0.89 in predicting 1p/19q codeletion status in LGGs. This logistic regression multivariable model was superior to that built using either VASARI features or random forest analysis. Our results afforded a method for predicting 1p/19q codeletion that is both noninvasive and uses data from routinely acquired MR images.
136
+
137
+ The current standard for glioma grading is based on histopathological assessment, which has 2 major limitations. First, it is an invasive procedure. Second, it has an inherent sampling error, especially with stereotactic biopsy. Our multivariable texture model achieved an AUC of 0.86, a sensitivity of 0.74, and a specificity of 0.79 in distinguishing grade II from grade III gliomas, a performance which compared favorably with previous studies.20–22 However, these prior studies relied on carefully selected ROIs drawn by neuroradiologists and multiple advanced MR imaging modalities, including diffusion-weighted imaging, diffusion-tensor imaging, MR spectroscopy, and perfusion-weighted imaging. These resources may not be available in non-academic institutions. Our algorithm demonstrated the potential of achieving high predictive accuracy using only conventional MRI sequences.
138
+
139
+ One texture feature in our final model that predicted histological grade was GLCM-homogeneity on contrast-enhanced T1-weighted images. We found that high tumor “homogeneity” was associated with higher grade. This result may be counterintuitive, since previous studies in breast cancer23 and soft tissue sarcomas24 have demonstrated that tumor heterogeneity is associated with higher tumor grade and more aggressive pathological features. We have 2 possible explanations for our results. First, MR CE in grade II tumors is more heterogeneous than CE in grade III tumors. This characteristic of CE in grade II gliomas was not captured by the corresponding VASARI feature key, but was demonstrated by Pallud et al.,25 where among the 143 cases of grade II tumor with CE, CE was characterized as “patchy and faint” in 93 and “nodular-like” in 50. Second, for those tumors without CE, our previous observation in soft tissue sarcoma was that high texture homogeneity was mostly due to large homogeneous necrotic centers.9 This suggests that the tumor has a fast growing rate, thus compatible with higher grade.
140
+
141
+ The ultimate goal of using machine-learning algorithm to study glioma is to predict patient outcome. Our multivariable texture model constructed using logistic regression achieved an AUC of 0.80, a sensitivity of 0.76, and a specificity of 0.72 in predicting LGG progression. These results were superior to those obtained using either VASARI features (AUC of 0.58) or random forest analysis (AUC of 0.69). Few studies have investigated the use of radiomics, specifically texture analysis based on baseline MR imaging, to predict outcomes in patients with gliomas, and existing studies focused exclusively on glioblastoma.21,26 Our study was the first in the literature to demonstrate the potential of using textural analysis from baseline MR imaging to predict glioma progression.
142
+
143
+ We acknowledge several limitations to our study. First, some patients could not be included due to incompleteness of MR sequence data from TCIA. In addition, the heterogeneity of different imaging parameters used by different investigators contributing to TCGA/TCIA data could not be controlled. Second, clinical variables, such as PFS and extent of resection, are not available for all patients from TCGA. Third, this study was partly based on the radiologist-scored imaging features. Although a good to excellent interreader agreement based on kappa consistency test has been achieved, the scores can be subject to interreader variability and random errors during manual contour tracing. Lastly, collection and assessment of new LGGs is still ongoing by our group, and we need more cases to validate our initial conclusions made in this paper.
144
+
145
+ In conclusion, tumor enhancement and an irregular non-enhancing margin on MR imaging were associated with shorter PFS, while a smooth non-enhancing margin was a positive predictor of OS. Multivariable texture models extracted from baseline MRI scans were able to classify LGGs in terms of IDH1 mutation, 1p/19q codeletion, histological grade, and tumor progression with high sensitivity and specificity.
146
+
147
+ Online Resources
148
+ The ROI masks used for texture analysis are made available in DICOM format on the TCIA website: http://www.cancerimagingarchive.net/. All MATLAB software code used to compute the texture features and multivariable model results is freely shared under the GNU General Public License at: https://github.com/mvallieres/radiomics.
149
+
150
+ Supplementary Material
151
+ Supplementary material is available at Neuro-Oncology online.
152
+
153
+ Funding
154
+ This work was supported by the Natural Science Foundation of China (81301988 to L.Y., 2014-1 to 2016-12), and China Ministry of Education Doctoral Program Spot Foundation (20130162120061 to L.Y.), Shenghua Yuying Project of Central South University to L.Y., and Natural Science Foundation of Hunan Province of China (14JJ2042, 2014-1 to 2016-12) to J.Z.
155
+
156
+ Conflict of interest statement. None declared.
157
+
158
+ References
159
+ 1. Brat DJ Verhaak RG Aldape KD et al. . Cancer Genome Atlas Research Network. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372(26):2481–2498.
160
+ Google ScholarCrossrefPubMedWorldCat
161
+ 2. van den Bent MJ Wefel JS Schiff D et al. . Response assessment in neuro-oncology (a report of the RANO group): assessment of outcome in trials of diffuse low-grade gliomas. Lancet Oncol. 2011;12(6):583–593.
162
+ Google ScholarCrossrefPubMedWorldCat
163
+ 3. Gutman DA Cooper LA Hwang SN et al. . MR imaging predictors of molecular profile and survival: multi-institutional study of the TCGA glioblastoma data set. Radiology. 2013;267(2):560–569.
164
+ Google ScholarCrossrefPubMedWorldCat
165
+ 4. Zhang B Chang K Ramkissoon S et al. . Multimodal MRI features predict isocitrate dehydrogenase genotype in high-grade gliomas. Neuro Oncol. 2016;0(0):1–9.
166
+ Google ScholarWorldCat
167
+ 5. Wiki for the VASARI feature set The National Cancer Institute Web site. Available at https://wikicancerimagingarchivenet/display/Public/VASARI+Research+Project. Updated July 25, 2016.
168
+ Google ScholarGoogle PreviewWorldCatCOPAC
169
+ 6. Gevaert O Mitchell LA Achrol AS et al. . Glioblastoma multiforme: exploratory radiogenomic analysis by using quantitative image features. Radiology. 2014;273(1):168–174.
170
+ Google ScholarCrossrefPubMedWorldCat
171
+ 7. Bai HX Lee AM Yang L et al. . Imaging genomics in cancer research: limitations and promises. Br J Radiol. 2016;89(1061):20151030.
172
+ Google ScholarCrossrefPubMedWorldCat
173
+ 8. Aerts HJ Velazquez ER Leijenaar RT et al. . Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat Commun. 2014;5:4006.
174
+ Google ScholarPubMedWorldCat
175
+ 9. Vallières M Freeman CR Skamene SR et al. . A radiomics model from joint FDG-PET and MRI texture features for the prediction of lung metastases in soft-tissue sarcomas of the extremities. Phys Med Biol. 2015;60(14):5471–5496.
176
+ Google ScholarCrossrefPubMedWorldCat
177
+ 10. Pedano N Flanders AE Scarpace L et al. Radiology data from The Cancer Genome Atlas Low Grade Glioma [TCGA-LGG] collection. Cancer Imaging Arch. 2016. doi:10.7937/K9/TCIA.2016.L4LTD3TK.
178
+ WorldCat
179
+ 11. Clark K Vendt B Smith K et al. The cancer imaging archive (TCIA): maintaining and operating a public information repository. J Digit Imaging. 2013;26(6):1045–1057.
180
+ WorldCat
181
+ 12. Schiller TW Chen Y Naqa IE et al. . Modeling radiation-induced lung injury risk with an ensemble of support vector machines. Neurocomputing. 2010;73(10–12):1861–1867.
182
+ Google ScholarCrossrefWorldCat
183
+ 13. Sahiner B Chan HP Hadjiiski L . Classifier performance prediction for computer-aided diagnosis using a limited dataset. Med Phys. 2008;35(4):1559–1570.
184
+ Google ScholarCrossrefPubMedWorldCat
185
+ 14. Nicolasjilwan M Hu Y Yan C et al. . TCGA Glioma Phenotype Research Group. Addition of MR imaging features and genetic biomarkers strengthens glioblastoma survival prediction in TCGA patients. J Neuroradiol. 2015;42(4):212–221.
186
+ Google ScholarCrossrefPubMedWorldCat
187
+ 15. Pope WB Sayre J Perlina A et al. . MR imaging correlates of survival in patients with high-grade gliomas. AJNR Am J Neuroradiol. 2005;26(10):2466–2474.
188
+ Google ScholarPubMedWorldCat
189
+ 16. Jenkins RB Blair H Ballman KV et al. . A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res. 2006;66(20):9852–9861.
190
+ Google ScholarCrossrefPubMedWorldCat
191
+ 17. Woehrer A Sander P Haberler C et al. . Research Committee of the European Confederation of Neuropathological Societies. FISH-based detection of 1p 19q codeletion in oligodendroglial tumors: procedures and protocols for neuropathological practice—a publication under the auspices of the Research Committee of the European Confederation of Neuropathological Societies (Euro-CNS). Clin Neuropathol. 2011;30(2):47–55.
192
+ Google ScholarCrossrefPubMedWorldCat
193
+ 18. Brown R Zlatescu M Sijben A et al. . The use of magnetic resonance imaging to noninvasively detect genetic signatures in oligodendroglioma. Clin Cancer Res. 2008;14(8):2357–2362.
194
+ Google ScholarCrossrefPubMedWorldCat
195
+ 19. Law M, Yang S, Wang H, et al. Glioma grading: sensitivity, specificity, and predictive values of perfusion MR imaging and proton MR spectroscopic imaging compared with conventional MR imaging. AJNR Am J Neuroradiol. 2003;24(10):1989-1998.
196
+ 20. Zacharaki EI, Wang S, Chawla S, et al. Classification of brain tumor type and grade using MRI texture and shape in a machine learning scheme. Magn Reson Med. 2009;62(6):1609-1618.
197
+ 21. Caulo M, Panara V, Tortora D, et al. Data-driven grading of brain gliomas: a multiparametric MR imaging study. Radiology. 2014;272(2):494-503.
198
+ 22. Soussan M Orlhac F Boubaya M et al. . Relationship between tumor heterogeneity measured on FDG-PET/CT and pathological prognostic factors in invasive breast cancer. PLoS One. 2014;9(4):e94017.
199
+ Google ScholarCrossrefPubMedWorldCat
200
+ 23. Zhao F Ahlawat S Farahani SJ et al. . Can MR imaging be used to predict tumor grade in soft-tissue sarcoma? Radiology. 2014;272(1):192–201.
201
+ Google ScholarCrossrefPubMedWorldCat
202
+ 24. Pallud J Capelle L Taillandier L et al. . Prognostic significance of imaging contrast enhancement for WHO grade II gliomas. Neuro Oncol. 2009;11(2):176–182.
203
+ Google ScholarCrossrefPubMedWorldCat
204
+ 25. Lee J Jain R Khalil K et al. . Texture feature ratios from relative CBV maps of perfusion MRI are associated with patient survival in glioblastoma. AJNR Am J Neuroradiol. 2016;37(1):37–43.
205
+ Google ScholarCrossrefPubMedWorldCat
206
+ 26. Yang D Rao G Martinez J et al. . Evaluation of tumor-derived MRI-texture features for discrimination of molecular subtypes and prediction of 12-month survival status in glioblastoma. Med Phys. 2015;42(11):6725–6735.
207
+ Google ScholarCrossrefPubMedWorldCat
208
+ Author notes
209
+ Corresponding Authors: Li Yang, MD, PhD, Department of Neurology, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, P.R. China (yangli762@gmail.com). Bo Xiao, MD,PhD, Department of Neurology, First Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, P.R. China (xiaobo1962xy@163.com).
210
+ *Hao Zhou and Martin Vallières contributed equally to this work.
211
+ © The Author(s) 2017. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.
samples/ROI-Masks_description.txt ADDED
@@ -0,0 +1,15 @@
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
 
1
+ This collection contains 406 Region Of Interest (ROI) masks in MATLAB
2
+ format defining the low grade glioma (LGG) tumor region on T1-weighted
3
+ (T1W), T2-weighted (T2W), T1-weighted post-contrast (T1CE) and
4
+ T2-flair (T2F) MR images of 108 different patients from the TCGA-LGG
5
+ collection. 81 of the patients have ROI masks drawn for the four MRI
6
+ sequences (T1W, T2W, T1CE and T2F), and 27 patients have ROI masks
7
+ drawn for three or less of the four MRI sequences. The ROI masks were
8
+ used to extract texture features in order to develop radiomic-based
9
+ multivariable models for the prediction of isocitrate dehydrogenase 1
10
+ (IDH1) mutation, 1p/19q codeletion status, histological grade and
11
+ tumor progression. Clinical data (188 patients in total from the
12
+ TCGA-LGG collection, some incomplete depending on the clinical
13
+ attribute), VASARI scores (188 patients in total from the TCGA-LGG
14
+ collection, 178 complete) with feature keys, and source code used in
15
+ this study are also available with this collection.